Lipoprotein (A) (2023
Lipoprotein (A) (2023
Karam Kostner
Gerhard M. Kostner
Peter P. Toth Editors
Lipoprotein(a)
Contemporary Cardiology
Series Editor
Peter P. Toth, Ciccarone Center for the Prevention of Cardiovascular Disease
Johns Hopkins University School of Medicine
Baltimore, MD, USA
For more than a decade, cardiologists have relied on the Contemporary Cardiology
series to provide them with forefront medical references on all aspects of cardiology.
Each title is carefully crafted by world-renown cardiologists who comprehensively
cover the most important topics in this rapidly advancing field. With more than 75
titles in print covering everything from diabetes and cardiovascular disease to the
management of acute coronary syndromes, the Contemporary Cardiology series has
become the leading reference source for the practice of cardiac care.
Karam Kostner • Gerhard M. Kostner
Peter P. Toth
Editors
Lipoprotein(a)
Editors
Karam Kostner Gerhard M. Kostner
Mater Hospital Medical University of Graz
University of Queensland Graz, Austria
Brisbane, QLD, Australia
Peter P. Toth
Ciccarone Center for the Prevention of
Cardiovascular Disease
Johns Hopkins University School of
Medicine
Baltimore, MD, USA
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature
Switzerland AG 2023
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether
the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and
transmission or information storage and retrieval, electronic adaptation, computer software, or by similar
or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in this book
are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the
editors give a warranty, expressed or implied, with respect to the material contained herein or for any
errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
This Humana imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Preface
v
vi Preface
ix
x Contents
23
Lipoprotein Apheresis for Reduction of Lipoprotein(a)���������������������� 377
Ulrich Julius and Sergey Tselmin
24 Elevated Lp(a): Why Should I Test For It, If I Cannot Treat It?
A Patient’s Perspective���������������������������������������������������������������������������� 409
Sandra Revill Tremulis
25 Unresolved Questions������������������������������������������������������������������������������ 425
Gerhard M. Kostner and Karam Kostner
Index������������������������������������������������������������������������������������������������������������������ 437
Contributors
xiii
xiv Contributors
Gerd Utermann
A Historical Review
Lipoprotein(a) was first described in 1963 by the Norwegian Physician Kåre Berg
(1963) (Fig. 1.1). As frequently in science, the history of Lp(a) started with a smart
idea, but ended with an unexpected result. In 1961/1962, Allison and Blumberg
(Allison and Blumberg 1961; Blumberg et al. 1962) described a polymorphism of
beta-lipoproteins, which they designated the Ag-system. They had observed that
some sera from polytransfused patients with thalassemia contained antibodies,
which distinguished between Ag-positive and Ag-negative sera from normal indi-
viduals in a test called Ouchterlonys double diffusion (Fig. 1.2). The availability of
anti-Ag sera depended on luck, required testing of many patients and quality was
G. Utermann (*)
Institute for Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
Institute for Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
e-mail: [email protected]
a b
Fig. 1.1 Panel a: Kåre Berg and the author at the first “International Lp workshop” 1967 in
Marburg/Lahn (Germany). Panel b: Title page of Kåre Berg’s first publication on Lp(a) with dedi-
cation to the organizer of the workshop Gerhard G. Wendt
difficult to control. Because antisera from polytransfused patients were not readily
available, limited in quantity, and could not be reproduced in the laboratory Kåre
Berg, at the time at the Institute of Forensic Medicine, Rikshospitalet, University of
Oslo/Norway had an idea to overcome these limitations. If an antigen elicited an
immune response in humans, it should do so also in rabbits. He started a series of
experiments, in which he immunized rabbits with individual human sera or beta-
lipoproteins. The rabbit sera were then “absorbed” with different individual human
sera to remove antibodies against foreign antigens present in all human sera.
Subsequently, the absorbed rabbit sera were tested against a panel of human sera for
antibodies recognizing individual human sera. Indeed, the plan worked. Some rab-
bit antisera reacted positive with some human sera and negative with others in
Ouchterlonys double diffusion (Fig. 1.2). Moreover, Berg could show that the
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 3
antigen in fact was a beta-lipoprotein. However, it was not identical with the
Ag-antigen (Berg 1964). Therefore, Berg introduced the name “Lp-System,” which
was later changed in Lp(a) to distinguish Lp(a) from other “Lp” Antigens. One was
“Lp(x)” which was detected by antisera produced in horse but turned out to be an
artifact. Berg distinguished between Lp(a+) and Lp(a−) individuals and showed by
family studies that the Lp(a) trait seemed to follow an autosomal dominant mode of
inheritance (Berg and Mohr 1963). Soon following the breakthrough discovery of
Berg, several laboratories tried to reproduce his finding, but with mixed results. In
principle, all confirmed Berg’s findings, but several researchers noticed that with
their antibodies the immune reaction was not an all or none. Instead, they observed
strong reactions (Bergs positives), no reactions (Bergs negatives), but also weak and
very weak reactions. It followed a discussion on whether the weak reactions were
true Lp(a) reactions or whether the antisera which recognized weak reactions were
unspecific containing antibodies to other components.
To clarify the situation and exchange latest research results, the human geneticist
Gerhard G. Wendt initiated the first “International Lp workshop” in Marburg/Lahn,
Germany (Wendt 1967). In preparation of the conference researchers from six dif-
ferent laboratories, including Kåre Berg’s sent in 17 antisera, which were tested
against a standard panel of 71 individual sera and analyzed for identity. The result
was that all antisera recognized the same antigen Lp(a), but confirmed the existence
of weak and very weak reactions which occurred to different degrees depending on
the antiserum. The issue was only resolved when researchers developed methods to
semi-quantify and finally quantify Lp(a), which demonstrated that Lp(a) in fact is a
quantitative trait (Harvie and Schultz 1970; Ehnholm et al. 1971). Methods to quan-
tify Lp(a) demonstrated large differences in median Lp(a) levels between and within
major human ethnic groups. The distributions of Lp(a) levels were highly skewed in
European and East-Asian populations but less so in sub-Saharan Africans (Fig. 1.3).
Mean and median Lp(a) levels were two to fourfold higher in Africans than
Europeans. The distributions in Asian populations were heterogenous with higher
Lp(a) levels in South-East Asia (Sandholzer et al. 1991; Parra et al. 1987a; Helmhold
et al. 1991).
4 G. Utermann
a
30
25 Khoi San
20 (N = 173)
15
10
5
0
30
Black Africans
25
(N = 193)
20
15
Relative Frequency (%)
10
5
0
30
Caucasians
25
(N = 224)
20
15
10
5
0
30
25 Chinese
(N = 198)
20
15
10
5
0
0 25 50 75 100 125 150 175
Lp(a) Concentration (mg/dl)
Fig. 1.3 Histograms showing: Panel (a) the distribution of plasma Lp(a) levels in four popula-
tions. Panel (b) the frequency distribution of the KIV-2 VNTR alleles in the same populations. The
total number of KIV repeats including the “unique” kringles is given. Panel (c) the inverse correla-
tion of KIV-2 repeats with Lp(a) concentration in the four populations. The Black Africans in this
study were from South Africa and represent different ethnicities. The Chinese samples were from
Hongkong and the “Caucasians” from Austria. (Figure reproduced from Kraft et al. 1996b with
permission)
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 5
b 12 Khoi San
(N = 346)
8
0
12
Black
Africans
8
(N = 386)
Relative Frequency (%)
0
12
Caucasians
(N = 448)
8
0
12
Chinese
(N = 396)
8
0
11 15 20 25 30 35 40 45 50
Number of Kringle-IV Repeats
c
150 Khoi San
(N = 173)
100
50
0
Black Africans
Lipoprotein(a) Concentration [mg/dl]
150
(N = 193)
100
50
150 Caucasians
(N = 224)
100
50
0
150 Chinese
(N = 198)
100
50
0
8 18 28 38 48 58
Sum of Kringle-IV Repeats
Beginning in 1968, first attempts were made to isolate Lp(a) from plasma and it was
shown that the antigenic property of Lp(a) is associated with a lipoprotein distinct
from LDL (Wiegandt et al. 1968; Utermann and Wiegandt 1969; Schultz et al. 1968).
A major breakthrough in Lp(a) research was the purification and characterization
of Lp(a) in 1970 by Christian Ehnholm (Fig. 1.4) in Kai Simons group in Helsinki,
Finland. They purified Lp(a) by a combination of preparative ultracentrifugation
and gel filtration on Sepharose 2B/4B columns and determined the physicochemical
properties of the particle (Ehnholm et al. 1971; Simons et al. 1970). Characteristics
of Lp(a) were a hydrated density of 1.09 g/mL, a molecular weight estimated by gel
filtration of 4.8 MDa and by electron microscopy of 5.6 MDa. Lp(a) had pre-beta
mobility in agarose gel electrophoresis and appeared as a spherical particle upon
electron microscopy. Notably it differed from LDL in amino acid composition and
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 7
a b
Fig. 1.4 Panel (a) Christian Ehnholm during a visit in Marburg/Lahn 1971 with the author. Panel
(b) laboratory equipment with Sepharose 4B column (red arrows) for final purification of Lp(a)
according to Ehnholm et al. (Ehnholm et al. 1971; Simons et al. 1970)
Lp(a) is usually depicted in cartoons as a global particle with an LDL in its center
and apo(a) wrapped around. Studies by electron microscopy of negatively stained
Lp(a) (Sines et al. 1994) and small-angel X-ray scattering (Prassl et al. 1995) sup-
ported this model but other studies suggested that apo(a) may protrude as a “tail”
from the particle depending on the environment (Weisel et al. 2001) and that Lp(a)
can switch between globular and “flexible tail” structures (Becker et al. 2004).
Metabolism
Studies of the metabolism of Lp(a) started in 1979 when Gerhard Kostner’s group
in Graz/Austria initiated a series of in vivo turnover studies in humans (Krempler
et al. 1979, 1980, 1983). Healthy individuals with different concentrations of Lp(a)
were injected with radioiodinated Lp(a) and in subsequent experiments with radio-
iodinated LDL. They demonstrated that (1) Lp(a) is not a metabolic product of other
apoB-containing lipoproteins, (2) that Lp(a) concentrations in plasma are deter-
mined by the rate of synthesis rather than by its catabolism, and (3) that Lp(a) is
catabolized at a slower rate than LDL.
Binding studies of radioactively labeled Lp(a) to human fibroblasts in compari-
son with LDL confirmed results of Havekes et al. (1981) and demonstrated that
Lp(a) binds with high affinity to the same cell surface receptor as LDL. However,
binding capacity for Lp(a) was lower than for LDL (Krempler et al. 1983). Lp(a) did
not bind to fibroblasts from patients with homozygous FH. These findings are in line
with later binding studies in fibroblasts and experiments in transgenic mice by
Goldstein and Brown (Hofmann et al. 1990) which indicated that Lp(a) is removed
from plasma by the LDL receptor pathway and with the observation that patients
with FH due to LDL receptor mutations or apoB100 mutations have elevated Lp(a)
in plasma (Seed et al. 1990; Utermann et al. 1989; Lingenhel et al. 1998; Van der
Hoek et al. 1997; Kraft et al. 2000). Together, these findings give a consistent pic-
ture. However, later studies on these topics were highly controversial and neither
confirmed the binding of Lp(a) to fibroblasts, nor the in vivo turnover studies in
humans and transgenic mice or the family studies (Soutar et al. 1991; Knight et al.
1991; Cain et al. 2005; Rader et al. 1995). Other receptors and pathways were impli-
cated to be involved in the removal of Lp(a) from plasma (reviewed in McCormick
and Schneider 2019). The liver (Cain et al. 2005) and the kidney (Kronenberg et al.
1997) both have been suggested as major sites of Lp(a) clearance from plasma. A
role for the kidney in Lp(a) clearance from the circulation was championed by the
group of Florian Kronenberg and Hans Dieplinger in Innsbruck and is supported by
several lines of evidence. Turnover studies demonstrated a reduced clearance of
Lp(a) in patients with kidney disease (Frischmann et al. 2007). Large arteriovenous
differences between Lp(a) concentrations were observed in the renovascular system
(Kronenberg et al. 1997). A problem with this study is that it requires the assump-
tion of unreasonably high synthesis rates of Lp(a) to compensate for the loss in the
kidneys. Further, Lp(a) binds with high affinity to megalin/gp330, a member of the
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 9
LDLR family expressed preferentially in kidneys (Niemeier et al. 1999) and frag-
ments of apo(a) were found in human urine (Oida et al. 1992; Mooser et al. 1996;
Kostner et al. 1996). None of this is direct evidence and at present the tissue(s) and
pathways of Lp(a) removal from the circulation remain unresolved. In contrast, the
liver as the site of synthesis and secretion of Lp(a) is undisputed. Hans-Georg Kraft
and colleagues from Innsbruck determined apo(a) isoform phenotypes (see below)
in plasma from patients undergoing liver transplantation and their organ donors.
They observed that genetic isoform phenotypes changed completely from recipients
to the donors phenotype following transplantation (Kraft et al. 1989). Apo(a) mRNA
was also most abundant in the liver from rhesus baboons and cynomolgus monkeys
(Tomlinson et al. 1989; Hixson et al. 1989; Azrolan et al. 1991).
Two important discoveries were made in 1987, the unique structure of apo(a)
(McLean et al. 1987) and the isoform polymorphism of apo(a) (Utermann et al. 1987).
The sequence of apo(a) had remained elusive for a long time and the reason
became clear when the sequence was finally resolved. Attempts to determine the
amino acids sequence of apo(a) by protein sequencing resulted in partial amino acids
sequences which demonstrated a high homology to plasminogen (Eaton et al. 1987;
Kratzin et al. 1987). Only by the breakthrough work of Richard Lawn and col-
leagues, at that time working at Genentech, the full sequence was elucidated. As a
pioneer in cloning technologies and DNA sequencing, Richard Lawn (Fig. 1.5) who
had previously sequenced hemoglobin loci from thalassemia patients started cloning
and cDNA sequencing of the LPA gene. This turned out to be much more compli-
cated than previous work. The result was unanticipated and astonishing. The deduced
amino acid sequence of apo(a) consisted of an array of so-called kringle domains
with a high internal homology and homology to kringle 4 from plasminogen. Ten
a b *
hedgehog 3’
3 3 3 3 3 3 3 3 3 3 3 3 3
apo (a)
72-75%
5’ 3’
plasminogen S P 1 2 3 4 5 PRO
75-85%
human 5’ 3’
a S 4 4 4 4 4 4 4 4 4 4 4 4 4 5 PRO
apo(a)
Fig. 1.5 Panel (a) Richard M. Lawn who published the first cDNA sequence of apo(a) (McLean
et al. 1987). Panel (b) Illustration of the convergent evolution of primate and insectivore apo(a).
The cDNA structures of plasminogen and human and hedgehog apo(a) are shown. Kringle types
are denoted by numbers and the protease domain by PRO. The stars indicate the sites of the
unpaired Cys residues which form the disulfide bridge with apoB in LDL. The percentages give the
degree of homology between plasminogen and apo(a). (Reproduced from Lawn et al. 1995b with
permission)
10 G. Utermann
kringle type IV different in sequence was identified, nine of them in single copy
(KIV-1, KIV-3 to KIV-10) whereas one (KIV-2) occurred in six identical copies in
the sequenced DNA. In addition, the protein contained one kringle with homology to
KV from PLG, a signal sequence and a plasminogen-like protease domain. The latter
was predicted to be inactive toward plasmin substrates due to mutation in the cata-
lytic triad. The findings showed that the LPA gene had been derived from the PLG
gene during evolution by a number of changes including duplication of PLG, dele-
tions and expansions of domains and mutations. The structure of all kringles is sta-
bilized by three internal disulfide bridges, which results in the typical appearance of
a Danish bretzel called “kringle.” One kringle (KIV-9) in addition contains one
unpaired cys residue, which turned out to be responsible for the covalent binding to
apoB of the LDL particle (Koschinsky et al. 1993; Brunner et al. 1993).
The sequence was so unusual that in an accompanying “News and Views” article
in Nature Joseph Goldstein and Michael Brown wrote that “the…. finding chal-
lenges the notion that evolution makes sense” (Brown and Goldstein 1987). The
enormous challenge which sequencing of LPA posed at the time becomes evident
when one considers that a successful search for mutations in the KIV-2 repeats of
LPA became possible only very recently (Coassin et al. 2019). It was certainly the
most heroic undertaking in Lp(a) research and opened new unexpected avenues.
Sequence analysis of PLG and human and rhesus LPA (Tomlinson et al. 1989)
had shown that LPA had evolved from PLG after the split of Old-world from New-
world monkeys some 40 million years ago and that Lp(a) existed only in Old-world
monkeys. Therefore, it was a surprise when Laplaud et al. in France reported pres-
ence of Lp(a) in the plasma of a hibernator, the hedgehog (Laplaud et al. 1988). The
surprise became even bigger when Lawns group sequenced the LPA of hedgehog.
Instead of KIV repeats, it contained multiple copies of KIII as the sole kringle type
and lacked the protease domain (Lawn et al. 1995a, 1997), but like the primate
counterpart hedgehog apo(a) formed a Lp(a) particle with LDL. Reports on the
presence of Lp(a) in guinea pigs (Rath and Pauling 1990) and the marmoset (a New-
world monkey) (Guo et al. 1991) were not confirmed. Hence, the occurrence of a
Lp(a)-like particle in the hedgehog by convergent evolution apparently remained a
solitary act (Lawn et al. 1995a, 1997).
Functional Studies
As shown by the work of Lawn and colleagues, the LPA gene had evolved from PLG
during primate evolution suggesting that the function of Lp(a) might be related to
the function of plasminogen and blood clotting. Already in the “News and Views”
article mentioned above, Goldstein and Brown had put forward the hypothesis that
Lp(a) through binding to fibrinogen might be involved in wound healing (Brown
and Goldstein 1987). The hypothesis—which still appears attractive, but was never
rigorously tested though—considers that Lp(a) is a macromolecular complex con-
taining two very different components, apo(a) and LDL. For most functions assigned
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 11
to Lp(a), the apo(a) alone is sufficient which leaves open the question, why the
particle exists. Beginning with the work of Harpel et al. (1989, 1995), numerous
studies demonstrated effects of Lp(a) on the blood clotting cascade and connected
thrombosis to atherosclerosis (Nachman 1992). Lp(a) was described as “an inter-
loper into the fibrinolytic system” (Miles and Plow 1990). Several interactions of
apo(a) with diverse ligands have been reported (Fig. 1.6) but whether any is of
physiological or pathophysiological relevance in humans remains unclear. A promi-
nent hypothesis explaining the atherogenic potential of Lp(a) was derived from the
finding that it is a “sink” for oxidized phospholipids (Tsimikas et al. 2005; Kiechl
et al. 2007; Bergmark et al. 2008). Lack in understanding of the function and patho-
physiological properties have recently been reviewed by an NHLBI working group
(Tsimikas et al. 2018). Existence of Lp(a) and the apo(a) size polymorphism in Old-
world monkeys implies that Lp(a) may have, or had, a function beyond one species.
The detection of many null mutations in the LPA gene (see below) may, however,
indicate that this function has been lost in modern humans with the possible excep-
tion of Africans.
Thrombolysis
Cells
PLG Matrix Platelets
A
tTP
Fibrin(ogen)
SMC Plasmin
OxPL
Tetranectin
C
TGFβ
N C
N Fibronectin
LDL
DANCE
Messangial
cell β2GP-I
Monocyte/ Decorin
Macrophage
Megalln/gp330
Anglogenesis
Endothelial cells VLDL-R
(LDL-R)
VCAM-1 PAI-1
MCA
E-selectin
Fig. 1.6 Model of Lp(a) and reported interactions of Lp(a)/apo(a) with components of the blood
clotting system, cell receptors, and other binding proteins. The binding of oxidized phospholipids
(OxPL in red) is considered as crucial for the pathogenicity of Lp(a). For explanation, see text and
Schmidt et al. (2016). (Modified from Utermann 1989, 2001 with permission)
12 G. Utermann
With the exception of the hedgehog, Lp(a) exists only in Old-world monkeys and
humans (Makino et al. 1989). The availability of natural animal models to study the
metabolism and pathophysiology of Lp(a) is limited. Rainwater and colleagues ana-
lyzed the genetics of Lp(a) extensively in baboons (Rainwater et al. 1986; White
et al. 1994a) and few studies were performed in rhesus monkeys (Rudel et al. 1977;
Williams-Blangero and Rainwater 1991; Enkhmaa et al. 2015) and more recently in
chimpanzees (Noureen et al. 2017). The isoform polymorphism and the inverse cor-
relation between isoform size and Lp(a) levels in plasma existed in all these pri-
mates. Chimpanzees from different West-African and Central-African habitats had
significantly different Lp(a) levels and isoform distributions in plasma (Noureen
et al. 2017). Experimental studies with these species are not allowed and unethical.
Therefore researchers started to generate mice transgenic for apo(a) immediately
following the cloning of apo(a) cDNA (Chiesa et al. 1992). The first animals gener-
ated had apo(a) free in plasma because mouse LDL apparently lacked the structural
requirement for binding apo(a) and forming the Lp(a) complex. Different approaches
were used to overcome this. Infusion of human LDL into apo(a) transgenic mice
resulted in the association of secreted apo(a) with circulating LDL and formation of
Lp(a) which could only be resolved by disulfide reduction (Chiesa et al. 1992). In
another study, human Lp(a) was infused into mice transgenic for the human LDL
receptor which confirmed cell culture studies which had shown high-affinity bind-
ing of Lp(a) to the receptor (Hofmann et al. 1990). With such short-term experi-
ments, it was not possible to investigate the pathophysiology and in particular the
atherogenic potential of Lp(a). This became possible when apo(a) transgenic mice
were crossed with mice strains transgenic for human apoB (Linton et al. 1993;
Callow et al. 1994). These mice strains were used to study the assembly (Callow
et al. 1994; Callow and Rubin 1995) and the atherogenic potential of Lp(a) (Callow
et al. 1995; Mancini et al. 1995a). To identify sequence elements that regulate liver-
specific tissue expression, sex hormone and diet response mice transgenic for yeast
artificial chromosomes (YACs) containing entire human apo(a) alleles were pro-
duced (Frazer et al. 1995; Acquati et al. 1999).
The second important finding in 1987 was the discovery of the size polymorphism
of apo(a). Beginning with its detection, it was clear that Lp(a) was a genetic trait.
Family and twin studies had shown that heritability of the trait is high. Morton et al.
(1985) concluded from a large family study that Lp(a) levels are controlled by one
major dominant gene and a residual heritable component. The gene(s) controlling
Lp(a) levels were unknown. This started to change when a group in Innsbruck/
Austria demonstrated that several genetic isoforms of apo(a), which differ in size,
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 13
occur in the population and that the size of isoforms correlated inversely with
plasma Lp(a) concentrations (Utermann et al. 1987; Utermann 1989), suggesting
that Lp(a) concentrations might be controlled by the LPA locus, which was con-
firmed by subsequent sib–pair linkage studies in European and North-American
White families (Boerwinkle et al. 1992; Kraft et al. 1992; Demeester et al. 1995;
Scholz et al. 1999) and in African Americans (Mooser et al. 1997). Sib–pair linkage
studies in families from South Africa and from Gabon demonstrated that the LPA
locus is the major locus determining Lp(a) levels also in autochthonous populations
from sub-Saharan Africa (Scholz et al. 1999; Schmidt et al. 2006). Compared to
populations of European descent, the KIV-2 VNTR explained less of the variation
in Lp(a) levels in Africans.
The size polymorphism was detected with the at that time new technique of
Western blotting. This method had, however, a drawback. The intensity of isoforms
varied widely depending on the associated Lp(a) concentrations. Many individuals
exhibited only one isoform upon Western blotting. For those individuals, it was
unclear whether they were homozygotes, i.e., expressed two isoforms of identical
size or whether one isoform was below detection limit or due to a none-expressed
allele (so-called null alleles). DNA technology was the way to overcome the prob-
lem. Already, the DNA sequence data demonstrating multiple identical copies of
kringle IV-2 had Lawn and colleagues led to speculate that differences in repeat
number might underlay the size polymorphism of apo(a). Semiquantitative data
from Southern blotting using a KIV-2-specific sequence as probe (Utermann 1989;
Lindahl et al. 1990) and differences in length of apo(a) mRNA from liver (Koschinsky
et al. 1990) supported this. The application of pulse-field gel electrophoresis/
Southern blotting, which had started as a collaboration and ended in a race, finally
allowed the group of Helen Hobbs in Dallas (Boerwinkle et al. 1992; Lackner et al.
1991) and Hans-Georg Kraft and colleagues in Innsbruck (Kraft et al. 1992) to
demonstrate the size polymorphism at the DNA level. By using appropriate nucle-
ases (e.g., KpnI), which cut the DNA only outside the KIV-2 sequence, allowed to
retain the entire repeat block in large DNA fragments of 20 to >200 kb. Its size
could be finally determined by PFGE/Southern blotting. It turned out that the pro-
tein size polymorphism resulted from a transcribed and translated copy number
variation. The genomic size of one KIV-2 copy was 5.6 kb. Today only a few protein
coding VNTRs have been characterized including in the PMU genes (Swallow et al.
1987), human proline-rich protein (Lyons et al. 1988), and the gene coding for
length variation in the keratin 10 chain (Korge et al. 1992). Very recently, these
transcribed and translated genes including LPA were identified in a genome- and
exomewide search (Mukamel et al. 2021). LPA is the most extensively studied with
a large impact on human health (Schmidt et al. 2016; Kronenberg 2016). In particu-
lar, Helen Hobbs and colleagues in Dallas characterized the LPA locus in detail at
the molecular level (Lackner et al. 1991, 1993).
The analysis by PFGE/Southern blotting alone, however, also resulted in an
incomplete picture. Whether and to which extend an allele was transcribed and
translated into protein could not be seen. Only the simultaneous application of
PFGE/Southern blotting of DNA and Western blotting of plasma allowed a
14 G. Utermann
with Lp(a) levels (Ogorelkova et al. 1999, 2001). With one exception, the SNPs
were not shared between populations (Fig. 1.7). The splice site SNP (Ogorelkova
et al. 1999) was shown by expression experiments in cell culture to result in a trun-
cated apo(a) protein unable to form the Lp(a) complex. This null allele had a fre-
quency of 0.053 in Tyrolians from Austria and 0.0635 in the Finnish population. It
was rediscovered in a large population genetic study in Finns (Lim et al. 2014)
without reference to the previous work. A high number of homozygotes was identi-
fied which had no associated clinical symptoms which led the authors to conclude
that Lp(a) has no essential function in vivo which is amazing for results from a
study analyzing an isolated population. Parson et al. (2004) identified a mutation in
the KIV-2 region of LPA which resulted in a stop codon (R21X) and extremely low
allele-associated Lp(a) levels. Later, large-scale studies showed that this variant has
a carrier frequency between 1.6% and 2.1% in European populations; 1000 Genome
data found that the R21X variant mostly occurs in Europeans and South Asians, is
absent in Africans, and shows varying frequencies in South American populations
(Di Maio et al. 2020).
A resequencing study of LPA was performed by the group of Crawford in indi-
viduals of non-Hispanic black and white ancestry from North America (Crawford
et al. 2008). Nineteen of the identified SNPs were then analyzed for an association
with plasma Lp(a) levels in >7000 participants of a population-based survey which
M75T
P71T W72R
K-IV K-IV K-IV K-IV K-IV K-IV K-IV K-IV K-IV K-IV K-V Protease
1 2 3 4 5 6 7 8 9 10
L101V
Khoi San donor splice
Africans (Khoi San and Blacks)
Europeans site mutation
Blacks and Europeans +1 G>A
Fig. 1.7 Figures illustrating differences in the genetic architecture of the Lp(a) trait between
human ethnic groups. The structure of the LPA gene with variants known until 2001 is shown. Data
compiled from Ogorelkova et al. (2001), Prins et al. (1999), Scanu et al. (1994). The color code is
only used for variants detected in the study including all four populations (Ogorelkova et al. 2001)
and illustrates that many variants in LPA occur only in one or few populations which is in agree-
ment with later larger studies (Mukamel et al. 2021; Dumitrescu et al. 2011). (Modified from
Utermann 1999 with permission)
16 G. Utermann
included three ethnic groups: Mexican Americans and non-Hispanic blacks and
whites (Dumitrescu et al. 2011). They found 15 SNPs which were associated with
Lp(a) levels in at least one ethnic group but none in all groups. They were not in
strong LD with the KIV-2 VNTR and explained from 7% to 11% of the variance of
Lp(a) levels in the respective ethnic group. Four of the variants were predicted by
PolyPhen to be possibly or probably damaging, but no functional studies were per-
formed. One variant KIV-8 Thr>Pro had also been described by Ogorelkova et al.
(2001). Hence, this large study extended and confirmed previous work. Very
recently, Sally McCormick’s group in New Zeeland performed a detailed functional
analysis of two non-synonymous variants R990Q and R1771C, which had been
detected in GWAS. They showed that both are causative for null Lp(a) phenotypes
and occur in positions homologous to positions in PLG which when mutated result
in PLG deficiency (Morgan et al. 2020). These are the first functionally character-
ized non-synonymous null mutations in the LPA gene.
With the exception of the one identified deleterious SNP R21X (Parson et al.
2004), the KIV-2 VNTR remained a black box for mutation detection until very
recently, when Asma Noureen and colleagues specifically amplified the KIV-2 tar-
get region by PCR from 90 PFGE-separated alleles from Asian, European, and four
different African populations and identified several SNPs in populations of African,
Asian, and European ancestry by Sanger sequencing (Noureen et al. 2015). As
reported for many other genes and from genome sequencing, they observed a higher
frequency of variable sites in Africans. Two previously unreported splice site vari-
ants were detected. One was a true null allele with no detectable Lp(a) associated
and the other had a high frequency (10–40%) in Africans. Their approach had the
advantage that SNPs could be assigned to KIV-2 copy number, but the disadvantage
that sensitivity was low and mutation detection was limited and depended on copy
number and on the number of KIV-2 repeats carrying the variant (intra-allelic fre-
quency). These problems were overcome when Stefan Coassin and colleagues in
Florian Kronenberg’s group in Innsbruck developed deep sequencing protocols,
which allowed systematic high-throughput mutation analysis of the KIV-2 VNTR
(Coassin et al. 2019). They identified a variety of new variants in LPA and analyzed
the effects on Lp(a) and associations with CHD of previously known (Di Maio et al.
2020) and newly identified variants (Coassin et al. 2017, 2020; Schachtl-Riess et al.
2021) in great detail. Their work and very recent genomic analysis (Mukamel et al.
2021) are presently at the cutting edge of Lp(a) genetics research. In particular by
the genomic analysis of Mukamel et al. (2021), many gaps in our knowledge of the
genetic architecture of the Lp(a) trait have been filled. They estimated KIV-2 VNTR
length from whole-genome sequencing data and defined VNTR alleles by imputa-
tion of SNP data which allowed to estimate frequencies and effects of VNTR hap-
lotypes on Lp(a) levels in populations of African, Asian, and European ancestry. A
total of 17 protein-altering variants each of which reduced Lp(a) levels significantly
as well as variants in the 5′ UTR which increased Lp(a) levels were observed.
Previously, the variants responsible for inter-population differences were largely
unknown. SNPs which had been claimed to explain level differences between ethnic
groups (Deo et al. 2011; Chretien et al. 2006) do this in a statistical sense only with
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 17
a few exceptions (Coassin et al. 2017; Schachtl-Riess et al. 2021). Mukamel et al.
(2021) now reported highly significant differences in the frequencies of variants
with causal effects between major human ethnic groups. This explains much of the
inter-ethnic differences in the genetic architecture of the Lp(a) trait between
these groups.
In addition to the major LPA locus, other genes have been identified which make
minor contributions to the variability of Lp(a) level variation including APOE (De
Knijff et al. 1991; Klausen et al. 1996) and APOH/ß2GPI (Hoekstra et al. 2021).
Genetic variation, which is restricted to an ethnic group, may also contribute. An
example is PCSK9. Loss-of-function mutations in this gene lower Lp(a) levels in
American blacks (Mefford et al. 2019).
A further category are genetic variants, which are rare or very rare, but have large
effects in carriers. Known examples are the genes for FH (Utermann et al. 1989; Van
der Hoek et al. 1997; Kraft et al. 2000), abetalipoproteinemia (Menzel et al. 1990),
lipoprotein lipase deficiency (Sandholzer et al. 1992a), and LCAT deficiency
(Steyrer et al. 1994).
The role of Lp(a) in cardiovascular disease has long been debated and the debate
followed an up and down parkour. The very first study reporting an association
observed a higher frequency of “Lp+” among patients with myocardial infarction
compared to controls (Renninger et al. 1965). This study of poor quality was pub-
lished in German language and largely ignored. The field started with the publica-
tions of Dahlen in Sweden (1974), who reported an association of
“pre-beta1-lipoprotein/Lp(a)” with CHD (Frick et al. 1974; Berg et al. 1974; Dahlén
et al. 1975). In a highly cited paper, Gerhard Kostner and colleagues reported
increased Lp(a) levels in patients with CHD over controls and defined 30 mg/dL as
the threshold for elevated Lp(a) in plasma (Kostner et al. 1981), a value which was
used in practice until recently. Histological demonstration and quantification of
Lp(a)/apo(a) in the aortic wall and atherosclerotic plaques strengthened the idea that
Lp(a) is a risk factor for cardiovascular disease (Költringer and Jürgens 1985; Rath
et al. 1989; Niendorf et al. 1990; Beisiegel et al. 1990). Further strong support
evolved from the homology of apo(a) with plasminogen (McLean et al. 1987) and
the functional studies based on this finding which assigned a dual role in the patho-
genesis of cardiovascular disease to Lp(a). As a particle composed of LDL and
apo(a), it was believed to be atherothrombotic (Loscalzo 1990). At the beginning of
the 1990th, the view of most researchers in the field was that Lp(a) is a risk factor
which was summarized in a popular paper by Richard Lawn in “Scientific American”
(Lawn 1992) with the title “Lipoprotein(a) in Heart Disease.”
Until then, all epidemiological studies relating Lp(a) to coronary risk were retro-
spective case–control studies. Circumstantial evidence for the pathogenicity of
Lp(a) was in addition deduced from functional studies and histology. To gain further
18 G. Utermann
Fig. 1.8 Panel (a) Histogram showing the distribution of binned short and long apo(a) isoform
frequencies in patients with CHD and controls in six populations (Data from Sandholzer et al.
1992c converted into graphic form. Adopted from Schmidt et al. 2016 with permission). Panel (b)
Schematic illustration of the principle of Mendelian Randomization as first applied for Lp(a) in the
studies of Sandholzer et al. (1992b, c). (From Kronenberg and Utermann 2013, used with
permission)
20 G. Utermann
which the principle of Mendelian randomization was clearly described. To cite from
these papers: “This is the first study which firmly establish a relationship between
genetic apo(a) isoforms, Lp(a) levels and CHD” and “The data demonstrate that
alleles at the apo(a) locus determine the risk for CHD through their effects on Lp(a)
levels and firmly establish the role of Lp(a) as a primary genetic risk factor”
(Sandholzer et al. 1992b).
For the time these studies were performed, they were large including 355 CHD
patients and 399 controls from China (Sandholzer et al. 1992b). The second study
(Sandholzer et al. 1992c) was even larger with more than 1.000 patients and con-
trols from six ethnic groups in which Lp(a) concentrations and apo(a) isoforms
were determined. In both studies, small isoforms (i.e., isoforms with fewer KIV-2
repeats) which determine higher Lp(a) levels were significantly more frequent in
CHD patients than in controls (Sandholzer et al. 1992c) (Fig. 1.8). In a further
smaller study, apo(a) KIV-2 genotypes were determined by PFGE/Southern blotting
together with apo(a) isoforms, Lp(a) levels, and disease status in patients that had
undergone coronary angiography. The results confirmed that apo(a) alleles with low
KIV-2 copy number and high associated Lp(a) concentration were significantly
overrepresented in the patients (Kraft et al. 1996a). Despite the small sample size,
the highly significant results reflected the fact that genotypes and expression level
of each allele was known.
These studies apparently were premature and at odds with some prospective
studies, which were the gold standard at the time. Though a first small prospective
study by Rosengren et al. (1990) reported serum Lp(a) as independent risk factor for
myocardial infarction in middle aged Swedish men, two subsequent studies, one
from Finland (Jauhiainen et al. 1991) and a large study from the US (Ridker et al.
1993), failed to find significant associations and concluded that Lp(a) is not an inde-
pendent risk factor. This provoked editorials in leading medical journals titled “Has
Lipoprotein ‘little’(a) Shrunk?” (Barnathan 1993). Subsequent prospective studies
(Schaefer et al. 1994; Cremer et al. 1994) and meta-analysis of a large number of
prospective studies published over the following years showed that these studies
were clearly outliers and found a strong association of Lp(a) concentration with
myocardial infarction and related phenotypes (Danesh et al. 2000; Bennet et al.
2008; Erqou et al. 2009). Today, it is known that the large influential study by Ridker
et al. (1993) was flawed by problems with Lp(a) quantification which was clarified
by the group in a later less prominently published paper (Suk et al. 2006). The
approach to relate apo(a) isoforms to CHD was also taken up by several groups and
meta-analysis of a series of 40 studies including 58.000 participants confirmed the
seminal studies of Sandholzer et al. (1992b, c) on the association of isoform size
with CHD (Erqou et al. 2010). Even this did not convince the entire community.
Only by the large Mendelian randomization studies of groups in Copenhagen/
Denmark (Kamstrup et al. 2009) and Oxford (Clarke et al. 2009) Lp(a) was finally
“…resurrected by genetics” (Kronenberg and Utermann 2013). Borge Nordestgaard’s
group determined Lp(a) levels and the sum of KIV-type-2 repeats from both apo(a)
alleles by quantitative PCR (qPCR) in relation to CHD in participants from the
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 21
Copenhagen City Heart Study. They demonstrated a strong relation between Lp(a)
levels, repeat number, and disease risk which was highest for individuals with high
Lp(a) concentration and low sum of repeat numbers (Kamstrup et al. 2009). The
conclusion from this study: “These data are consistent with a causal association
between elevated Lp(a) levels and increased risk of MI” (Kamstrup et al. 2009) was
almost identical with the one from the early isoform studies (Sandholzer et al.
1992b,c). Resurrection had happened twice but unlike in the bible, one was not
enough. The second resurrection again changed headlines in journals, e.g., in
“Lipoprotein(a): There’s life in the old dog yet” (Kronenberg 2014a) or
“Lipoprotein(a): the underestimated cardiovascular risk factor” (Thompson and
Seed 2014) and finally triggered the development of drugs to lower Lp(a) in people
with increased risk.
If genetically elevated Lp(a) levels increase risk for CVD as shown, geneti-
cally lowered Lp(a) should result in the opposite, i.e., risk reduction. This was in
fact shown in population-based Mendelian randomization study from Finland
(Lim et al. 2014) by the PROCARDIS study in Germany (Kyriakou et al. 2014)
and by Stefan Coassin, Florian Kronenberg, and colleagues in Innsbruck who
tested this hypothesis in two large studies (Coassin et al. 2017; Schachtl-Riess
et al. 2021): they discovered two common splice site variants (4925G>A and
4733G>A) newly detected by deep sequencing in the KIV-2 repeat which both
decreased Lp(a) concentrations tremendously (Coassin et al. 2017; Schachtl-
Riess et al. 2021). The 4925G>A variant is observed in about 22% of European
populations and is associated with smaller isoforms (mainly 19–25 K-IV repeats)
and decreases Lp(a) concentrations by roughly 30 mg/dL. Carriers of these
smaller isoforms who carry at the same time the 4925G>A splice site variant have
a decreased risk for CHD (Coassin et al. 2017) which has also been confirmed by
an Icelandic study (Gudbjartsson et al. 2019). The other splice site variant
4733G>A is with 38% even more frequent and occurs over a wide apo(a) isoform
range and lowers Lp(a) by 13.6 mg/dL and also the risk for CHD (Schachtl-Riess
et al. 2021). Using data from more than 440.000 participants from the UK Biobank
revealed that carriers of both variants have low Lp(a) concentrations and a 12%
decreased risk for CHD compared to non-carriers of the two mutations (Schachtl-
Riess et al. 2021).
Data from an Icelandic study confirmed that Lp(a) levels are associated in a
dose-dependent manner with risk for CAD, PVD, aortic valve stenosis, heart fail-
ure, and lifespan (Gudbjartsson et al. 2019). Short apo(a) alleles were also associ-
ated with risk but no additional residual association beyond the association with
Lp(a) levels was observed for the KIV-2 polymorphism when Lp(a) was at the same
time in the statistical model (Gudbjartsson et al. 2019). This can be explained by the
fact that the Lp(a) concentration is the measured biological exposure which is only
explained partially by the K-IV repeat polymorphism or other genetic variants.
The Mendelian randomization approach was further used to estimate the magni-
tude of a drug effect on Lp(a) to achieve a desired reduction of the risk for CHD
(Burgess et al. 2018; Lamina and Kronenberg 2019; Madsen et al. 2020).
22 G. Utermann
Estimates of the magnitude of the effect of the LPA locus on Lp(a) levels may be
misleading. The studies from which the estimates were derived were performed on
random population samples and healthy sib–pairs and families. The high heritability
of more than 90% does therefore not exclude that rare and common conditions, e.g.,
diseases not represented in the sample may have significant effects on Lp(a) in
affected individuals and groups and add to their health problems. Early studies on
the effects of environment and various disease states were small, definition, treat-
ments, and subtypes of disease differed between studies, and with few exceptions,
they were controversial.
It was early recognized that Lp(a) levels in an individual may not be stable over
time. Hormones and particularly changes in hormone levels during puberty and
pregnancy were recognized as a cause (reviewed in Kostner and Kostner 2004) but
slight fluctuations without apparent reason seem to be normal. Nutrition and physi-
cal activity have no effects.
Of clinical relevance are two associations of Lp(a) beyond the one with CAD. One
is the association with chronic kidney disease (CKD) and the other with type 2 dia-
betes mellitus (T2D). The relationship of Lp(a) with renal disease including CHD in
CKD is particularly complex. The first observation on elevated Lp(a) in hemodialy-
sis patients probably is by Papadopoulos et al. (1980) who described a high fre-
quency of an additional “pre-beta-lipoprotein” band visible in agarose gel
electrophoresis in the patients. This “pre-beta-lipoprotein” appears identical with
the “pre-beta1-lipoprotein” described by Dahlen to be associated with CAD (Dahlen
1974; Frick et al. 1974) and identified as Lp(a) (Berg et al. 1974). H. Parra in Jean-
Luis Fruchart’s group in Lille/France first reported Lp(a) elevation in patients with
chronic renal failure (Parra et al. 1987b). Numerous studies followed which con-
firmed the observation and further revealed that the extend of Lp(a) increase depends
on the type and treatment and that the increase in Lp(a) may have different causes,
i.e., impaired removal from the circulation (Frischmann et al. 2007) or increased
synthesis (Kronenberg et al. 1996). The increase is higher in patients with end-stage
renal disease treated by continuous ambulatory peritoneal dialysis compared to
those under hemodialysis (Kronenberg et al. 1996; Kronenberg 2014b). Patients
with nephrotic syndrome develop excessive elevations of Lp(a) (Kronenberg et al.
1996, 2004; Takegoshi et al. 1991; Wanner et al. 1993). Following renal transplanta-
tion Lp(a) levels decrease to almost normal concentrations (Kronenberg et al.
1994a) demonstrating that the increased Lp(a) levels are secondary to disease
(Kronenberg et al. 1994a; Black and Wilcken 1992).
Patients with ESRD have an increased risk for arterial vascular disease. Therefore,
the report of Cressman and colleagues (1992) describing elevated Lp(a) as indepen-
dent risk factor for CHD was noted with interest and triggered several follow-up
studies. Though confirmed by most studies, it turned out that it was not the full truth
but matters were more complicated. Apo(a) isoforms also play a role as demon-
strated by Florian Kronenberg, Hans Dieplinger, and colleagues in Innsbruck who
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 23
unrevealed the complex interplay between renal disease, Lp(a) levels, and athero-
sclerotic vascular disease (Kronenberg et al. 1994b, 1999a, b; Koch et al. 1997).
Despite higher Lp(a) levels in the patients, isoform frequencies were not different
from controls (Dieplinger et al. 1993; Kronenberg et al. 1995). The pronounced
increases in levels in patients were associated mainly with the longer isoforms and
much less so with the short isoforms (Kronenberg et al. 1995, 1996; Dieplinger
et al. 1993). The extend of Lp(a) increase with renal disease is mirrored by the rapid
reduction following renal transplantation which is also dependent on the genetic
phenotype (Kronenberg et al. 1994a). In heterozygous patients with one short and
one long isoform which were analyzed before and following transplantation the
change in concentration of the long, but not short isoform was impressively demon-
strated by Western blotting (Kronenberg et al. 2003).
As in the general population, apo(a) phenotypes predicted the risk for atheroscle-
rotic vascular disease in ESDR patients (Kronenberg et al. 1994b, 1999a) but sur-
prisingly and in contrast to the work of Cressman and colleagues (1992) Lp(a)
levels were found to be poor predictors. This paradoxical situation was explained by
the different timelines of events. The increase of Lp(a) in patients with high molecu-
lar weight phenotypes starts only with the onset of disease and therefore does not
last long enough for a significant pathogenic effect. In contrast, the exposure to high
Lp(a) is lifelong in patients with low molecular phenotypes resulting in more prein-
jury and rapid development of atherothrombotic vascular disease. Lp(a) levels lose
their predictive power whereas apo(a) types retain it. Kronenberg coined the term
“galloping” atherosclerosis for the rapidly progressive form of vascular disease in
ESDR patients with small apo(a) isoforms (Kronenberg et al. 1994b). As for most
disease associations in the Lp(a) field, the reported association of apo(a) isoforms
with ASVD in CKD patients was not confirmed by all and is controversial. In a
recent review (Hopewell et al. 2018), it was concluded that CKD patients with high
Lp(a) levels are at increased risk whereas it is unclear whether apo(a) isoforms are
predictive for ASVD.
The situation on the role of Lp(a) in T2D is far from clear and illustrates how in
some areas of Lp(a) research there was little progress over longer periods. Early
small case-control studies from the 1990s were controversial. Some reported ele-
vated (Bruckert et al. 1990; Heller et al. 1993), some lower (Rainwater et al. 1994),
and some no change in Lp(a) levels (Császár et al. 1993) compared to controls.
Rainwater et al. first reported decreased Lp(a) levels and larger apo(a) isoforms
(Rainwater et al. 1994). Prospective studies investigating the role of Lp(a) as risk
factor for incident or prevalent T2D observed an association of very low Lp(a) levels
with disease (Mora et al. 2010; Ye et al. 2014; Paige et al. 2017) but it was unclear
whether low Lp(a) is causally related to disease. To clarify this several groups initi-
ated Mendelian randomization studies. These were, however, controversial. In a
Mendelian randomization study from Copenhagen (Kamstrup and Nordestgaard
2013), no causal relation of low Lp(a) with T2D was observed and the role of long
KIV-2 repeats remained unclear. A study in Chinese patients with CHD in which
Lp(a) levels and KIV-2 repeats were determined, both low Lp(a) and high repeat
number were associated with T2D. This resulted in the conclusion that low Lp(a)
24 G. Utermann
predisposes to T2D (Mu-Han-Ha-Li et al. 2018). This view was challenged by the
work of Tolbus et al. (2017) who used SNPs tagging Lp(a) levels or KIV-2 repeat
number. They found high KIV-2 repeat numbers but not Lp(a) levels associated with
T2D. In the very large population-based Iceland study, Kari Steffansson’s group
(Gudbjartsson et al. 2019) analyzed the association of loss-of-function (LOF) muta-
tions in the LPA gene for a Mendelian randomization approach. These included
known “null” mutations in LPA which are associated with very low or no Lp(a) in
plasma. Presence of these mutations increased the risk for T2D in the Icelandic pop-
ulation. This to date is the most convincing evidence that very low Lp(a) predisposes
to T2D. The mechanism underlying this relation is unknown but if known could shed
new light on the still unresolved question for what reason evolution created Lp(a).
The recommendations for individuals with a high risk for CHD or which suffered
already from MI, angina pectoris, and related phenotypes and had elevated Lp(a) in
plasma was for a long time to reduce other risk factors more rigorously. There were
simply no drugs available. Niacin was the first recognized to lower Lp(a), but the
effect was small the drug affected also LDL and HDL metabolism and had unde-
sired side effects. An unexpected disappointment was that HMG-CoA-reductase
inhibitors which effectively lower LDL by increasing LDL receptors at the cell
surface did not lower Lp(a) (Kostner et al. 1989). The next surprise was that PCSK9
inhibitors, which also exert their effect on LDL by increasing LDL receptor-
mediated uptake of LDL did also decrease Lp(a) concentrations (Raal et al. 2014)
and are particularly effective in patients with familial hypercholesterolemia and
elevated Lp(a) (Vuorio et al. 2020). The Lp(a)-lowering effect of PCSK9 inhibitors
was shown in cell culture experiments to be mediated through an overexpression of
LDL receptors and increased internalization of Lp(a) by the receptors (Romagnuolo
et al. 2015). This is at odds with the lack of an effect of HMG-CoA-reductase inhib-
itors and presently unexplained. PCSK9 inhibitors are now a choice for treatment of
at-risk patients with high Lp(a), but do not selectively lower Lp(a) in patients with
very high Lp(a). Only recently, Lp(a) has been specifically targeted by antisense
therapy against apo(a) (Tsimikas et al. 2015; Graham et al. 2016) with ongoing
phase-III trials. A method, which has been very effective in reducing the risk for
MACE in patients with severe clinical CHD, is lipoprotein apheresis (Jaeger et al.
2009; Roeseler et al. 2016; Pottle et al. 2019; Schettler et al. 2017). This therapy had
been developed by the groups of Walter Stoffel in Cologne, which used antibody
columns (Stoffel et al. 1981) and Dietrich Seidel in Göttingen who used heparin
linked to columns (Eisenhauer et al. 1987; Armstrong et al. 1989) to absorb LDL
and Lp(a) from plasma. This therapy is presently still the only one with a proven
significant risk reduction in patients with severe CHD.
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 25
Outlook
A historical article describes the development of a scientific field from the past until
the present day but not beyond. But can we learn from history for the future? What
we certainly can learn is where the gaps in our knowledge are and what the impor-
tant questions for the future might be.
In Lp(a) research, one major open question is what the physiological function of
Lp(a) may be, a question which to the authors mind is tightly linked to evolutionary
genetic aspects. The rhetorical question of Brown and Goldstein “does evolution
make sense” in their News and Views article in Nature accompanying the cloning
and sequencing of the apo(a) cDNA (Brown and Goldstein 1987) may well be
extended to Lp(a).
It has been concluded that Lp(a) has no significant function since a large num-
ber of Europeans is heterozygous or compound homozygous for “null mutations”
in the LPA gene and consequently Lp(a) is absent in plasma; these individuals are
healthy and without any signs of disease (Schmidt et al. 2006). This conclusion is
not warranted. First, the conclusion can only be that Lp(a) has no health-related
function under present day conditions in the respective population (i.e., Finns).
Second and more importantly, it seems highly unlikely that a macromolecular
complex the assembly of which requires specific sites for non-covalent interac-
tion between two extremely different large components, a lipoprotein and a gly-
coprotein, and the formation of a covalent disulfide bridge between them being
present in all Old-world monkeys has survived for 40 Mio years when it has no
function.
Considering that Lp(a) concentrations on average are higher in South-East
Asians and severalfold higher in Africans compared to Europeans and that “null
mutations” are significantly less frequent in Africans though Africans had longer
time to accumulate deleterious mutations in none-essential genes, it may be specu-
lated that Lp(a) has a significant role in Africa and further Old-world tropical popu-
lations. If Lp(a) has an unknown health-related function, e.g., in Africans, lowering
Lp(a) by drugs for prevention of coronary disease may have undesired side effects.
Large-scale studies of the Lp(a)/apo(a) trait in relation to disease, e.g., infectious
disease in African populations may therefore elucidate the physiological role of
Lp(a) and also have practical consequences for Lp(a) lowering drug regimes in
these populations.
Acknowledgments I thank Anita Neuner for help with the literature, Eugen Preuss for preparing
figures, and Florian Kronenberg for his support and for critically reading the manuscript.
26 G. Utermann
References
Burgess S, Ference BA, Staley JR, Freitag DF, Mason AM, Nielsen SF, Willeit P, Young R,
Surendran P, Karthikeyan S, Bolton TR, Peters JE, Kamstrup PR, Tybjaerg-Hansen A, Benn
M, Langsted A, Schnohr P, Vedel-Krogh S, Kobylecki CJ, Ford I, Packard C, Trompet S,
Jukema JW, Sattar N, Di AE, Saleheen D, Howson JMM, Nordestgaard BG, Butterworth
AS, Danesh J. Association of LPA variants with risk of coronary disease and the implications
for lipoprotein(a)-lowering therapies: a Mendelian randomization analysis. JAMA Cardiol.
2018;3:619–27.
Cain WJ, Millar JS, Himebauch AS, Tietge UJ, Maugeais C, Usher D, Rader DJ. Lipoprotein [a]
is cleared from the plasma primarily by the liver in a process mediated by apolipoprotein [a]. J
Lipid Res. 2005;46:2681–91.
Callow MJ, Rubin EM. Site-specific mutagenesis demonstrates that cysteine 4326 of apo-
lipoprotein B is required for covalent linkage with apolipoprotein(a) in vivo. J Biol Chem.
1995;270:23914–7.
Callow MJ, Stoltzfus LJ, Lawn RM, Rubin EM. Expression of human apolipoprotein B and assem-
bly of lipoprotein(a) in transgenic mice. Proc Natl Acad Sci U S A. 1994;91:2130–4.
Callow MJ, Verstuyft J, Tangirala R, Palinski W, Rubin EM. Atherogenesis in transgenic mice with
human apolipoprotein B and lipoprotein (a). J Clin Invest. 1995;96:1639–46.
Chiesa G, Hobbs HH, Koschinsky ML, Lawn RM, Maika SD, Hammer RE. Reconstitution of
lipoprotein(a) by infusion of human low density lipoprotein into transgenic mice expressing
human apolipoprotein(a). J Biol Chem. 1992;267:24369–74.
Chretien JP, Coresh J, Berthier-Schaad Y, Kao WH, Fink NE, Klag MJ, Marcovina SM, Giaculli
F, Smith MW. Three single-nucleotide polymorphisms in LPA account for most of the increase
in lipoprotein(a) level elevation in African Americans compared with European Americans. J
Med Genet. 2006;43:917–23.
Clarke R, Peden JF, Hopewell JC, Kyriakou T, Goel A, Heath SC, Parish S, Barlera S, Franzosi
MG, Rust S, Bennett D, Silveira A, Malarstig A, Green FR, Lathrop M, Gigante B, Leander K,
de Faire U, Seedorf U, Hamsten A, Collins R, Watkins H, Farrall M. Genetic variants associ-
ated with Lp(a) lipoprotein level and coronary disease. N Engl J Med. 2009;361:2518–28.
Coassin S, Erhart G, Weissensteiner H, de Araújo MEG, Lamina C, Schönherr S, Forer L, Haun M,
Losso JL, Köttgen A, Schmidt K, Utermann G, Peters A, Gieger C, Strauch K, Finkenstedt A,
Bale R, Zoller H, Paulweber B, Eckardt KU, Hüttenhofer A, Huber LA, Kronenberg F. A novel
but frequent variant in LPA KIV-2 is associated with a pronounced Lp(a) and cardiovascular
risk reduction. Eur Heart J. 2017;38:1823–31.
Coassin S, Schönherr S, Weissensteiner H, Erhart G, Forer L, Losso JL, Lamina C, Haun M,
Utermann G, Paulweber B, Specht G, Kronenberg F. A comprehensive map of single-base
polymorphisms in the hypervariable LPA kringle IV type 2 copy number variation region. J
Lipid Res. 2019;60:186–99.
Coassin S, Hermann-Kleiter N, Haun M, Wahl S, Wilson R, Paulweber B, Kunze S, Meitinger
T, Strauch K, Peters A, Waldenberger M, Kronenberg F, Lamina C. A genome-wide analysis
of DNA methylation identifies a novel association signal for Lp(a) concentrations in the LPA
promoter. PLoS One. 2020;15:e0232073.
Crawford DC, Peng Z, Cheng JF, Boffelli D, Ahearn M, Nguyen D, Shaffer T, Yi Q, Livingston RJ,
Rieder MJ, Nickerson DA. LPA and PLG sequence variation and Kringle IV-2 copy number in
two populations. Hum Hered. 2008;66:199–209.
Cremer P, Nagel D, Labrot B, Mann H, Muche R, Elster H, Seidel D. Lipoprotein Lp(a) as pre-
dictor of myocardial infarction in comparison to fibrinogen, LDL cholesterol and other risk
factors: results from the prospective Göttingen Risk Incidence and Prevalence Study (GRIPS).
Eur J Clin Investig. 1994;24:444–53.
Cressman MD, Heyka RJ, Paganini EP, O’Neil J, Skibinski CI, Hoff HF. Lipoprotein(a) is an
independent risk factor for cardiovascular disease in hemodialysis patients. Circulation.
1992;86:475–82.
Császár A, Dieplinger H, Sandholzer C, Karádi I, Juhász E, Drexel H, Halmos T, Romics L, Patsch
JR, Utermann G. Plasma lipoprotein(a) concentration and phenotypes in diabetes mellitus.
Diabetologia. 1993;36:47–51.
28 G. Utermann
Dahlen G. The pre-beta1 lipoprotein phenomenon in relation to serum cholesterol and triglyceride
levels, the Lp(a) lipoprotein and coronary heart disease. Acta Med Scand Suppl. 1974;570:
1–45.
Dahlén G, Berg K, Gillnäs T, Ericson C. Lp(a) lipoprotein/pre-beta1-lipoprotein in Swedish
middle-aged males and in patients with coronary heart disease. Clin Genet. 1975;7:334–41.
Danesh J, Collins R, Peto R. Lipoprotein(a) and coronary heart disease: meta-analysis of prospec-
tive studies. Circulation. 2000;102:1082–5.
Davey Smith G, Ebrahim S. Mendelian randomization: prospects, potentials, and limitations. Int J
Epidemiol. 2004;33:30–42.
De Knijff P, Kaptein A, Boomsma D, Princen HMG, Frants RR, Havekes LM. Apolipoprotein E
polymorphism affects plasma levels of lipoprotein(a). Atherosclerosis. 1991;90:169–74.
Demeester CA, Bu X, Gray RJ, Lusis AJ, Rotter JI. Genetic variation in lipoprotein (a) levels in
families enriched for coronary artery disease is determined almost entirely by the apolipopro-
tein (a) gene locus. Am J Hum Genet. 1995;56:287–93.
Deo RC, Wilson JG, Xing C, Lawson K, Kao WH, Reich D, Tandon A, Akylbekova E, Patterson
N, Mosley TH Jr, Boerwinkle E, Taylor HA Jr. Single-nucleotide polymorphisms in LPA
explain most of the ancestry-specific variation in Lp(a) levels in African Americans. PLoS
One. 2011;6:e14581.
Di Maio S, Grüneis R, Streiter G, Lamina C, Maglione M, Schoenherr S, Öfner D, Thorand B,
Peters A, Eckardt K-U, Köttgen A, Kronenberg F, Coassin S. Investigation of a nonsense muta-
tion located in the complex KIV-2 copy number variation region of apolipoprotein(a) in 10,910
individuals. Genome Med. 2020;12:74.
Dieplinger H, Lackner C, Kronenberg F, Sandholzer C, Lhotta K, Hoppichler F, Graf H, König
P. Elevated plasma concentrations of lipoprotein(a) in patients with end-stage renal disease are
not related to the size polymorphism of apolipoprotein(a). J Clin Invest. 1993;91:397–401.
Dumitrescu L, Glenn K, Brown-Gentry K, Shephard C, Wong M, Rieder MJ, Smith JD, Nickerson
DA, Crawford DC. Variation in LPA is associated with Lp(a) levels in three populations from
the Third National Health and Nutrition Examination Survey. PLoS One. 2011;6:e16604.
Eaton DL, Fless GM, Kohr WJ, McLean JW, Xu Q-T, Miller CG, Lawn RM, Scanu AM. Partial
amino acid sequence of apolipoprotein[a] shows that it is homologous to plasminogen. Proc
Natl Acad Sci U S A. 1987;84:3224–8.
Ehnholm C, Garoff H, Simons K, Aro H. Purification and quantitation of the human plasma lipo-
protein carrying the Lp(a) antigen. Biochim Biophys Acta. 1971;236:431–9.
Ehnholm C, Garoff H, Renkonen O, Simons K. Protein and carbohydrate composition of Lp(a)
lipoprotein from human plasma. Biochemistry. 1972;11:3229–32.
Eisenhauer T, Schuff-Werner P, Armstrong VW, Talartschik J, Scheler F, Seidel D. Long-term
experience with the HELP system for treatment of severe familial hypercholesterolemia.
ASAIO Trans. 1987;33:395–7.
Enkhmaa B, Abbuthalha A, Anuurad E, Zhang W, Tarantal AF, Berglund L. Rhesus mon-
key (Macaca mulatta) lipoprotein(a) and apolipoprotein(a): high frequency of small size
apolipoprotein(a) isoforms. J Med Primatol. 2015;44:117–24.
Erqou S, Kaptoge S, Perry PL, Di AE, Thompson A, White IR, Marcovina SM, Collins R,
Thompson SG, Danesh J. Lipoprotein(a) concentration and the risk of coronary heart disease,
stroke, and nonvascular mortality. JAMA. 2009;302:412–23.
Erqou S, Thompson A, Di AE, Saleheen D, Kaptoge S, Marcovina S, Danesh J. Apolipoprotein(a)
isoforms and the risk of vascular disease: systematic review of 40 studies involving 58,000
participants. J Am Coll Cardiol. 2010;55:2160–7.
Frazer KA, Narla G, Zhang JL, Rubin EM. The apolipoprotein(a) gene is regulated by sex hor-
mones and acute-phase inducers in YAC transgenic mice. Nat Genet. 1995;9:424–31.
Frick MH, Dahlén G, Furbery C, Ericson C, Wiljasalo M. Serum pre-beta-1 lipoprotein fraction in
coronary atherosclerosis. Acta Med Scand. 1974;195:337–40.
Frischmann ME, Kronenberg F, Trenkwalder E, Schaefer J, Schweer H, Dieplinger B, König
P, Ikewaki K, Dieplinger H. In vivo turnover study demonstrates diminished clearance of
lipoprotein(a) in hemodialysis patients. KI. 2007;71:1036–43.
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 29
Gaubatz JW, Heideman C, Gotto AM Jr, Morrisett JD, Dahlen GH. Human plasma lipoprotein[a].
Structural properties. J Biol Chem. 1983;258:4582–9.
Graham MJ, Viney N, Crooke RM, Tsimikas S. Antisense inhibition of apolipoprotein (a) to lower
plasma lipoprotein (a) levels in humans. J Lipid Res. 2016;57:340–51.
Gudbjartsson DF, Thorgeirsson G, Sulem P, Helgadottir A, Gylfason A, Saemundsdottir J, Bjornsson
E, Norddahl GL, Jonasdottir A, Jonasdottir A, Eggertsson HP, Gretarsdottir S, Thorleifsson G,
Indridason OS, Palsson R, Jonasson F, Jonsdottir I, Eyjolfsson GI, Sigurdardottir O, Olafsson
I, Danielsen R, Matthiasson SE, Kristmundsdottir S, Halldorsson BV, Hreidarsson AB,
Valdimarsson EM, Gudnason T, Benediktsson R, Steinthorsdottir V, Thorsteinsdottir U, Holm
H, Stefansson K. Lipoprotein(a) concentration and risks of cardiovascular disease and diabetes.
J Am Coll Cardiol. 2019;74:2982–94.
Guo HC, Michel JB, Blouquit Y, Chapman MJ. Lipoprotein(a) and apolipoprotein(a) in a
New World monkey, the common marmoset (Callithrix jacchus). Association of variable
plasma lipoprotein(a) levels with a single apolipoprotein(a) isoform. Arterioscler Thromb.
1991;11:1030–41.
Harpel PC, Gordon BR, Parker TS. Plasmin catalyzes binding of lipoprotein(a) to immobilized
fibrinogen and fibrin. Proc Natl Acad Sci U S A. 1989;86:3847–51.
Harpel PC, Hermann A, Zhang XX, Ostfeld I, Borth W. Lipoprotein(a), plasmin modulation, and
atherogenesis. Thromb Haemost. 1995;74:382–6.
Harvie NR, Schultz JS. Studies of Lp-lipoprotein as a quantitative genetic trait. Proc Natl Acad Sci
U S A. 1970;66:99–103.
Havekes L, Vermeer BJ, Brugman T, Emeis J. Binding of Lp (a) to the low density lipoprotein
receptor of human fibroblast. FEBS Lett. 1981;132:169–73.
Hegele RA, Breslow JL. Apolipoprotein genetic variation in the assessment of atherosclerosis
susceptibility. Genet Epidemiol. 1987;4:163–84.
Hegele RA, Huang LS, Herbert PN, Blum CB, Buring JE, Hennekens CH, Breslow
JL. Apolipoprotein B-gene DNA polymorphisms associated with myocardial infarction. N
Engl J Med. 1986;315:1509–15.
Heller FR, Jamart J, Honore P, Derue G, Novik V, Galanti L, Parfonry A, Hondekijn J-C, Buysschaert
M. Serum lipoprotein(a) in patients with diabetes mellitus. Diabetes Care. 1993;16:819–23.
Helmhold M, Bigge J, Muche R, Mainoo J, Thiery J, Seidel D, Armstrong VW. Contribution of the
apo[a] phenotype to plasma Lp[a] concentrations shows considerable ethnic variation. J Lipid
Res. 1991;32:1919–28.
Hixson JE, Britten ML, Manis GS, Rainwater DL. Apolipoprotein(a) (Apo(a)) glycoprotein iso-
forms result from size differences in Apo(a) mRNA in baboons. J Biol Chem. 1989;264:6013–6.
Hoekstra M, Chen HY, Rong J, Dufresne L, Yao J, Guo X, Tsai MY, Tsimikas S, Post WS, Vasan
RS, Rotter JI, Larson MG, Thanassoulis G, Engert JC. Genome-wide association study
highlights APOH as a novel locus for lipoprotein(a) levels-brief report. Arterioscler Thromb
Vasc Biol. 2021;41:458–64.
Hofmann SL, Eaton DL, Brown MS, McConathy WJ, Goldstein JL, Hammer RE. Overexpression
of human low density lipoprotein receptors leads to accelerated catabolism of Lp(a) lipoprotein
in transgenic mice. J Clin Invest. 1990;85:1542–7.
Hopewell JC, Haynes R, Baigent C. The role of lipoprotein (a) in chronic kidney disease. J Lipid
Res. 2018;59:577–85.
Humphries SE, Dunning A, Xu CF, Peacock R, Talmud P, Hamsten A. DNA polymorphism stud-
ies. Approaches to elucidating multifactorial ischaemic heart disease: the apo B gene as an
example. Ann Med. 1992;24:349–56.
Jaeger BR, Richter Y, Nagel D, Heigl F, Vogt A, Roeseler E, Parhofer K, Ramlow W, Koch M,
Utermann G, Labarrere CA, Seidel D. Longitudinal cohort study on the effectiveness of lipid
apheresis treatment to reduce high lipoprotein(a) levels and prevent major adverse coronary
events. Nat Clin Pract Cardiovasc Med. 2009;6:229–39.
Jauhiainen M, Koskinen P, Ehnholm C, Frick MH, Mänttäri M, Manninen V, Huttunen
JK. Lipoprotein(a) and coronary heart disease risk: a nested case-control study of the Helsinki
Heart Study participants. Atherosclerosis. 1991;89:59–67.
30 G. Utermann
Kamstrup PR, Nordestgaard BG. Lipoprotein(a) concentrations, isoform size, and risk of type 2
diabetes: a Mendelian randomisation study. Lancet Diabetes Endocrinol. 2013;1:220–7.
Kamstrup PR, Tybjaerg-Hansen A, Steffensen R, Nordestgaard BG. Genetically elevated
lipoprotein(a) and increased risk of myocardial infarction. JAMA. 2009;301:2331–9.
Katan MB. Apolipoprotein E isoforms, serum cholesterol, and cancer. Lancet. 1986;1:507–8.
Katan MB. Commentary: Mendelian randomization, 18 years on. Int J Epidemiol. 2004;33:
10–1.
Kiechl S, Willeit J, Mayr M, Viehweider B, Oberhollenzer F, Kronenberg F, Wiedermann CJ,
Oberthaler S, Xu Q, Witztum JL, Tsimikas S. Oxidized phospholipids, lipoprotein(a),
lipoprotein-associated phospholipase A2 activity and 10-year cardiovascular outcomes: pro-
spective results from the Bruneck study. Arterioscler Thromb Vasc Biol. 2007;27:1788–95.
Klausen IC, Gerdes LU, Hansen PS, Lemming L, Gerdes C, Faergeman O. Effects of apoE gene
polymorphism on Lp(a) concentrations depend on the size of apo(a): a study of 466 white men.
J Mol Med. 1996;74:685–90.
Knight BL, Perombelon YFN, Soutar AK, Wade DP, Seed M. Catabolism of lipoprotein(a) in
familial hypercholesterolaemic subjects. Atherosclerosis. 1991;87:227–37.
Koch M, Kutkuhn B, Trenkwalder E, Bach D, Grabensee B, Dieplinger H, Kronenberg
F. Apolipoprotein B, fibrinogen, HDL cholesterol and apolipoprotein(a) phenotypes predict
coronary artery disease in hemodialysis patients. J Am Soc Nephrol. 1997;8:1889–98.
Költringer P, Jürgens G. A dominant role of lipoprotein(a) in the investigation and evalua-
tion of parameters indicating the development of cervical atherosclerosis. Atherosclerosis.
1985;58:187–98.
Korge BP, Gan SQ, McBride OW, Mischke D, Steinert PM. Extensive size polymorphism of the
human keratin 10 chain resides in the C-terminal V2 subdomain due to variable numbers and
sizes of glycine loops. Proc Natl Acad Sci U S A. 1992;89:910–4.
Koschinsky ML, Beisiegel U, Henne-Bruns D, Eaton DL, Lawn RM. Apolipoprotein(a) size
heterogeneity is related to variable number of repeat sequences in its mRNA. Biochemistry.
1990;29:640–4.
Koschinsky ML, Côté GP, Gabel B, Van der Hoek YY. Identification of the cysteine residue in
apolipoprotein(a) that mediates extracellular coupling with apolipoprotein B-100. J Biol Chem.
1993;268:19819–25.
Kostner KM, Kostner GM. Factors affecting plasma lipoprotein(a) levels: role of hormones and
other nongenetic factors. Semin Vasc Med. 2004;4:211–4.
Kostner GM, Avogaro P, Cazzolato G, Marth E, Bittolo-Bon G, Qunici GB. Lipoprotein Lp(a) and
the risk for myocardial infarction. Atherosclerosis. 1981;38:51–61.
Kostner GM, Gavish D, Leopold B, Bolzano K, Weintraub MS, Breslow JL. HMG CoA reduc-
tase inhibitors lower LDL cholesterol without reducing Lp(a) levels. Circulation. 1989;80:
1313–9.
Kostner KM, Maurer G, Huber K, Stefenelli T, Dieplinger H, Steyrer E, Kostner GM. Urinary
excretion of apo(a) fragments. Role in apo(a) catabolism. Arterioscler Thromb Vasc Biol.
1996;16:905–11.
Kraft HG, Menzel HJ, Hoppichler F, Vogel W, Utermann G. Changes of genetic apolipoprotein
phenotypes caused by liver transplantation. Implications for apolipoprotein synthesis. J Clin
Invest. 1989;83:137–42.
Kraft HG, Köchl S, Menzel HJ, Sandholzer C, Utermann G. The apolipoprotein(a) gene: a tran-
scribed hypervariable locus controlling plasma lipoprotein(a) concentration. Hum Genet.
1992;90:220–30.
Kraft HG, Lingenhel A, Köchl S, Hoppichler F, Kronenberg F, Abe A, Mühlberger V, Schönitzer
D, Utermann G. Apolipoprotein(a) Kringle IV repeat number predicts risk for coronary heart
disease. Arterioscler Thromb Vasc Biol. 1996a;16:713–9.
Kraft HG, Lingenhel A, Pang RWC, Delport R, Trommsdorff M, Vermaak H, Janus ED, Utermann
G. Frequency distributions of apolipoprotein(a) kringle IV repeat alleles and their effects on
lipoprotein(a) levels in Caucasian, Asian, and African populations: the distribution of null
alleles is non-random. Eur J Hum Genet. 1996b;4:74–87.
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 31
Kraft HG, Windegger M, Menzel HJ, Utermann G. Significant impact of the +93 C/T polymor-
phism in the apolipoprotein(a) gene on Lp(a) concentrations in Africans but not in Caucasians:
confounding effect of linkage disequilibrium. Hum Mol Genet. 1998;7:257–64.
Kraft HG, Lingenhel A, Raal FJ, Hohenegger M, Utermann G. Lipoprotein(a) in homozygous
familial hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2000;20:522–8.
Kratzin H, Armstrong VW, Niehaus M, Hilschmann N, Seidel D. Structural relationship of an
apolipoprotein(a) phenotype (570 kDa) to plasminogen: homologous kringle domains are
linked by carbohydrate-rich regions. Biol Chem Hoppe Seyler. 1987;368:1533–44.
Krempler F, Kostner G, Bolzano K, Sandhofer F. Lipoprotein (a) is not a metabolic product of
other lipoproteins containing apolipoprotein B. Biochim Biophys Acta. 1979;575:63–70.
Krempler F, Kostner GM, Bolzano K, Sandhofer F. Turnover of lipoprotein(a) in man. J Clin
Invest. 1980;65:1483–90.
Krempler F, Kostner GM, Roscher A, Haslauer F, Bolzano K, Sandhofer F. Studies on the role
of specific cell surface receptors in the removal of lipoprotein(a) in man. J Clin Invest.
1983;71:1431–41.
Kronenberg F. Lipoprotein(a): there’s life in the old dog yet. Circulation. 2014a;129:619–21.
Kronenberg F. Causes and consequences of lipoprotein(a) abnormalities in kidney disease. Clin
Exp Nephrol. 2014b;18:234–7.
Kronenberg F. Human genetics and the causal role of lipoprotein(a) for various diseases. Cardiovasc
Drugs Ther. 2016;30:87–100.
Kronenberg F, Utermann G. Lipoprotein(a)—resurrected by genetics. J Intern Med. 2013;273:6–30.
Kronenberg F, König P, Lhotta K, Öfner D, Sandholzer C, Margreiter R, Dosch E, Utermann G,
Dieplinger H. Apolipoprotein(a) phenotype-associated decrease in lipoprotein(a) plasma con-
centrations after renal transplantation. Arterioscler Thromb. 1994a;14:1399–404.
Kronenberg F, Kathrein H, König P, Neyer U, Sturm W, Lhotta K, Gröchenig E, Utermann G,
Dieplinger H. Apolipoprotein(a) phenotypes predict the risk for carotid atherosclerosis in
patients with end-stage renal disease. Arterioscler Thromb. 1994b;14:1405–11.
Kronenberg F, König P, Neyer U, Auinger M, Pribasnig A, Lang U, Reitinger J, Pinter G, Utermann
G, Dieplinger H. Multicenter study of lipoprotein(a) and apolipoprotein(a) phenotypes in
patients with end-stage renal disease treated by hemodialysis or continuous ambulatory perito-
neal dialysis. J Am Soc Nephrol. 1995;6:110–20.
Kronenberg F, Utermann G, Dieplinger H. Lipoprotein(a) in renal disease. Am J Kidney Dis.
1996;27:1–25.
Kronenberg F, Trenkwalder E, Lingenhel A, Friedrich G, Lhotta K, Schober M, Moes N, König P,
Utermann G, Dieplinger H. Renovascular arteriovenous differences in Lp(a) plasma concen-
trations suggest removal of Lp(a) from the renal circulation. J Lipid Res. 1997;38:1755–63.
Kronenberg F, Neyer U, Lhotta K, Trenkwalder E, Auinger M, Pribasnig A, Meisl T, König P,
Dieplinger H. The low molecular weight apo(a) phenotype is an independent predictor for
coronary artery disease in hemodialysis patients: a prospective follow-up. J Am Soc Nephrol.
1999a;10:1027–36.
Kronenberg F, Kronenberg MF, Kiechl S, Trenkwalder E, Santer P, Oberhollenzer F, Egger G,
Utermann G, Willeit J. Role of lipoprotein(a) and apolipoprotein(a) phenotype in atherogen-
esis: prospective results from the Bruneck Study. Circulation. 1999b;100:1154–60.
Kronenberg F, Lhotta K, König P, Margreiter R, Dieplinger H, Utermann G. Apolipoprotein(a)
isoform-specific changes of lipoprotein(a) after kidney transplantation. Eur J Hum Genet.
2003;11:693–9.
Kronenberg F, Lingenhel A, Lhotta K, Rantner B, Kronenberg MF, König P, Thiery J, Koch M,
Von Eckardstein A, Dieplinger H. The apolipoprotein(a) size polymorphism is associated with
nephrotic syndrome. Kidney Int. 2004;65:606–12.
Kyriakou T, Seedorf U, Goel A, Hopewell JC, Clarke R, Watkins H, Farrall M. A common LPA null
allele associates with lower lipoprotein(a) levels and coronary artery disease risk. Arterioscler
Thromb Vasc Biol. 2014;34:2095–9.
32 G. Utermann
Lackner C, Boerwinkle E, Leffert CC, Rahmig T, Hobbs HH. Molecular basis of apolipoprotein
(a) isoform size heterogeneity as revealed by pulsed-field gel electrophoresis. J Clin Invest.
1991;87:2153–61.
Lackner C, Cohen JC, Hobbs HH. Molecular definition of the extreme size polymorphism in
apolipoprotein(a). Hum Mol Genet. 1993;2:933–40.
Lamina C, Kronenberg F. Estimation of the required lipoprotein(a)-lowering therapeutic effect size
for reduction in coronary heart disease outcomes: a Mendelian randomization analysis. JAMA
Cardiol. 2019;4:575–9.
Laplaud PM, Beaubatie L, Rall SC Jr, Luc G, Saboureau M. Lipoprotein[a] is the major apoB-
containing lipoprotein in the plasma of a hibernator, the hedgehog (Erinaceus europaeus). J
Lipid Res. 1988;29:1157–70.
Lawn RM. Lipoprotein(a) in heart disease. Sci Am. 1992;266:54–60.
Lawn RM, Wade DP, Hammer RE, Chiesa G, Verstuyft JG, Rubin EM. Atherogenesis in trans-
genic mice expressing human apolipoprotein(a). Nature. 1992;360:670–2.
Lawn RM, Boonmark NW, Schwartz K, Lindahl GE, Wade DP, Byrne CD, Fong KJ, Meer K,
Patthy L. The recurring evolution of lipoprotein(a). Insights from cloning of hedgehog
apolipoprotein(a). J Biol Chem. 1995a;270:24004–9.
Lawn RM, Boonmark NW, Schwartz K, Lindahl GE, Wade DP, Byrne CD, Fong KJ, Meer K,
Patthy L. The recurring evolution of lipoprotein(a)—insights from cloning of hedgehog
apolipoprotein(a). J Biol Chem. 1995b;270:24004–9.
Lawn RM, Schwartz K, Patthy L. Convergent evolution of apolipoprotein(a) in primates and
hedgehog. Proc Natl Acad Sci U S A. 1997;94:11992–7.
Lim ET, Wurtz P, Havulinna AS, Palta P, Tukiainen T, Rehnstrom K, Esko T, Magi R, Inouye
M, Lappalainen T, Chan Y, Salem RM, Lek M, Flannick J, Sim X, Manning A, Ladenvall C,
Bumpstead S, Hamalainen E, Aalto K, Maksimow M, Salmi M, Blankenberg S, Ardissino D,
Shah S, Horne B, McPherson R, Hovingh GK, Reilly MP, Watkins H, Goel A, Farrall M, Girelli
D, Reiner AP, Stitziel NO, Kathiresan S, Gabriel S, Barrett JC, Lehtimaki T, Laakso M, Groop
L, Kaprio J, Perola M, McCarthy MI, Boehnke M, Altshuler DM, Lindgren CM, Hirschhorn
JN, Metspalu A, Freimer NB, Zeller T, Jalkanen S, Koskinen S, Raitakari O, Durbin R,
MacArthur DG, Salomaa V, Ripatti S, Daly MJ, Palotie A. Distribution and medical impact of
loss-of-function variants in the Finnish founder population. PLoS Genet. 2014;10:e1004494.
Lindahl G, Gersdorf E, Menzel HJ, Seed M, Humphries S, Utermann G. Variation in the
size of human apolipoprotein(a) is due to a hypervariable region in the gene. Hum Genet.
1990;84:563–7.
Lingenhel A, Kraft H-G, Kotze M, Peeters AV, Kronenberg F, Kruse R, Utermann G. Concentrations
of the atherogenic Lp(a) are elevated in FH. Eur J Hum Genet. 1998;6:50–60.
Linton MF, Farese RV, Chiesa G, Grass DS, Chin P, Hammer RE, Hobbs HH, Young SG. Transgenic
mice expressing high plasma concentrations of human apolipoprotein B100 and lipoprotein(a).
J Clin Invest. 1993;92:3029–37.
Lobentanz E-M, Krasznai K, Gruber A, Brunner C, Müller H-J, Sattler J, Kraft H-G, Utermann G,
Dieplinger H. Intracellular metabolism of human apolipoprotein(a) in stably transfected Hep
G2 cells. Biochemistry. 1998;37:5417–25.
Loscalzo J. Lipoprotein(a). A unique risk factor for atherothrombotic disease. Arteriosclerosis.
1990;10:672–9.
Lyons KM, Stein JH, Smithies O. Length polymorphisms in human proline-rich protein genes
generated by intragenic unequal crossing over. Genetics. 1988;120:267–78.
Madsen CM, Kamstrup PR, Langsted A, Varbo A, Nordestgaard BG. Lp(a) (Lipoprotein[a])-
lowering by 50 mg/dL (105 nmol/L) may be needed to reduce cardiovascular disease 20% in sec-
ondary prevention: a population-based study. Arterioscler Thromb Vasc Biol. 2020;40:255–66.
Makino K, Abe A, Maeda S, Noma A, Kawade M, Takenaka O. Lipoprotein(a) in nonhuman pri-
mates. Presence and characteristics of Lp(a) immunoreactive materials using anti-human Lp(a)
serum. Atherosclerosis. 1989;78:81–5.
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 33
Mancini FP, Newland DL, Mooser V, Murata J, Marcovina S, Young SG, Hammer RE, Sanan
DA, Hobbs HH. Relative contributions of apolipoprotein(a) and apolipoprotein-B to the
development of fatty lesions in the proximal aorta of mice. Arterioscler Thromb Vasc Biol.
1995a;15:1911–6.
Mancini FP, Mooser V, Guerra R, Hobbs HH. Sequence microheterogeneity in apolipoprotein(a)
gene repeats and the relationship to plasma Lp(a) levels. Hum Mol Genet. 1995b;4:1535–42.
McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors.
Pathology. 2019;51:155–64.
McLean JW, Tomlinson JE, Kuang W-J, Eaton DL, Chen EY, Fless GM, Scanu AM, Lawn
RM. cDNA sequence of human apolipoprotein(a) is homologous to plasminogen. Nature.
1987;330:132–7.
Mefford MT, Marcovina SM, Bittner V, Cushman M, Brown TM, Farkouh ME, Tsimikas S,
Monda KL, Lopez JAG, Muntner P, Rosenson RS. PCSK9 loss-of-function variants and Lp(a)
phenotypes among black US adults. J Lipid Res. 2019;60:1946–52.
Menzel HJ, Kladetzky RG, Assmann G. Apolipoprotein E polymorphism and coronary artery dis-
ease. Arteriosclerosis. 1983;3:310–5.
Menzel H-J, Dieplinger H, Lackner C, Hoppichler F, Lloyd JK, Muller DR, Labeur C, Talmud PJ,
Utermann G. Abetalipoproteinemia with an apoB-100-lipoprotein(a) glycoprotein complex in
plasma: indication for an assembly defect. J Biol Chem. 1990;265:981–6.
Miles LA, Plow EF. Lp(a): an interloper into the fibrinolytic system? Thromb Haemost.
1990;63:331–5.
Mooser V, Mancini FP, Bopp S, Pethö-Schramm A, Guerra R, Boerwinkle E, Müller H-J, Hobbs
HH. Sequence polymorphisms in the apo(a) gene associated with specific levels of Lp(a) in
plasma. Hum Mol Genet. 1995;4:173–81.
Mooser V, Seabra MC, Abedin M, Landschulz KT, Marcovina S, Hobbs HH. Apolipoprotein(a)
kringle 4-containing fragments in urine. Relationship to plasma levels of lipoprotein(a). J Clin
Invest. 1996;97:858–64.
Mooser V, Scheer D, Marcovina SM, Wang JP, Guerra R, Cohen J, Hobbs HH. The Apo(a) gene
is the major determinant of variation in plasma Lp(a) levels in African Americans. Am J Hum
Genet. 1997;61:402–17.
Mora S, Kamstrup PR, Rifai N, Nordestgaard BG, Buring JE, Ridker PM. Lipoprotein(a) and risk
of type 2 diabetes. Clin Chem. 2010;56:1252–60.
Morgan BM, Brown AN, Deo N, Harrop TWR, Taiaroa G, Mace PD, Wilbanks SM, Merriman TR,
Williams MJA, McCormick SPA. Nonsynonymous SNPs in LPA homologous to plasminogen
deficiency mutants represent novel null apo(a) alleles. J Lipid Res. 2020;61:432–44.
Morton NE, Berg K, Dahlen G, Ferrell RE, Rhoads GG. Genetics of the Lp lipoprotein in Japanese-
Americans. Genet Epidemiol. 1985;2:113–21.
Mu-Han-Ha-Li D-L-D-E, Zhai T-Y, Ling Y, Gao X. LPA kringle IV type 2 is associated with
type 2 diabetes in a Chinese population with very high cardiovascular risk. J Lipid Res.
2018;59:884–91.
Mukamel RE, Handsaker RE, Sherman MA, Barton AR, Zheng Y, McCarroll SA, Loh
PR. Protein-coding repeat polymorphisms strongly shape diverse human phenotypes. Science.
2021;373:1499–505.
Nachman RL. Review: Stratton Lecture. Thrombosis and atherogenesis: molecular connections.
Blood. 1992;79:1897–906.
Niemeier A, Willnow T, Dieplinger H, Jacobsen C, Meyer N, Hilpert J, Beisiegel U. Identification
of Megalin/gp330 as a receptor for lipoprotein(a) in vitro. Arterioscler Thromb Vasc Biol.
1999;19:552–61.
Niendorf A, Rath M, Wolf K, Peters S, Arps H, Beisiegel U, Dietel M. Morphological detection
and quantification of lipoprotein(a) deposition in atheromatous lesions of human aorta and
coronary arteries. Virchows Arch [A]. 1990;417:105–11.
34 G. Utermann
Noureen A, Fresser F, Utermann G, Schmidt K. Sequence variation within the KIV-2 copy number
polymorphism of the human LPA gene in African, Asian, and European populations. PLoS
One. 2015;10:e0121582.
Noureen A, Ronke C, Khalifa M, Halbwax M, Fischer A, André C, Atencia R, Garriga R,
Mugisha L, Ceglarek U, Thiery J, Utermann G, Schmidt K. Significant differentiation in the
apolipoprotein(a)/lipoprotein(a) trait between chimpanzees from Western and Central Africa.
Am J Primatol. 2017;79 https://doi.org/10.1002/ajp.22683.
Ogorelkova M, Gruber A, Utermann G. Molecular basis of congenital Lp(a) deficiency: a frequent
apo(a) ‘null’ mutation in Caucasians. Hum Mol Genet. 1999;8:2087–96.
Ogorelkova M, Kraft HG, Ehnholm C, Utermann G. Single nucleotide polymorphisms in exons of
the apo(a) kringles IV types 6 to 10 domain affect Lp(a) plasma concentrations and have differ-
ent patterns in Africans and Caucasians. Hum Mol Genet. 2001;10:815–24.
Oida K, Takai H, Maeda H, Takahashi S, Shimada A, Suzuki J, Tamai T, Nakai T, Miyabo
S. Apolipoprotein(a) is present in urine and its excretion is decreased in patients with renal
failure. Clin Chem. 1992;38:2244–8.
Paige E, Masconi KL, Tsimikas S, Kronenberg F, Santer P, Weger S, Willeit J, Kiechl S, Willeit
P. Lipoprotein(a) and incident type-2 diabetes: results from the prospective Bruneck study and
a meta-analysis of published literature. Cardiovasc Diabetol. 2017;16:38.
Papadopoulos NM, Borer WZ, Elin RJ. An abnormal lipoprotein in the serum of uremic patients
maintained on chronic hemodialysis. Ann Intern Med. 1980;92:634–5.
Parra HJ, Luyéyé I, Bouramoué C, Demarquilly C, Fruchart JC. Black-white differences in serum
Lp(a) lipoprotein levels. Clin Chim Acta. 1987a;168:27–31.
Parra HJ, Mezdour H, Cachera C, Dracon M, Tacquet A, Fruchart JC. Lp(a) lipoprotein in patients
with chronic renal failure treated by hemodialysis. Clin Chem. 1987b;33:721.
Parson W, Kraft HG, Niederstatter H, Lingenhel AW, Kochl S, Fresser F, Utermann G. A common
nonsense mutation in the repetitive Kringle IV-2 domain of human apolipoprotein(a) results in
a truncated protein and low plasma Lp(a). Hum Mutat. 2004;24:474–80.
Paulweber B, Friedl W, Krempler F, Humphries SE, Sandhofer F. Genetic variation in the apo-
lipoprotein AI-CIII-AIV gene cluster and coronary heart disease. Atherosclerosis. 1988;73:
125–33.
Pottle A, Thompson G, Barbir M, Bayly G, Cegla J, Cramb R, Dawson T, Eatough R, Kale V,
Neuwirth C, Nicholson K, Payne J, Scott J, Soran H, Walji S, Watkins S, Weedon H, Nath Datta
DB. Lipoprotein apheresis efficacy, challenges and outcomes: a descriptive analysis from the
UK Lipoprotein Apheresis Registry, 1989-2017. Atherosclerosis. 2019;290:44–51.
Prassl R, Schuster B, Abuja PM, Zechner M, Kostner GM, Laggner P. A comparison of structure
and thermal behavior in human plasma lipoprotein(a) and low-density lipoprotein. Calorimetry
and small-angle X-ray scattering. Biochemistry. 1995;34:3795–801.
Prins J, Leus FR, Van der Hoek YY, Kastelein JJP, Bouma BN, Van Rijn HJM. The identifica-
tion and significance of a Thr–>Pro polymorphism in Kringle IV type 8 of apolipoprotein(a).
Thromb Haemost. 1997;77:949–54.
Prins J, Leus FR, Bouma BN, van Rijn HJ. The identification of polymorphisms in the coding
region of the apolipoprotein (a) gene—association with earlier identified polymorphic sites and
influence on the lipoprotein (a) concentration. Thromb Haemost. 1999;82:1709–17.
Raal FJ, Giugliano RP, Sabatine MS, Koren MJ, Langslet G, Bays H, Blom D, Eriksson M, Dent R,
Wasserman SM, Huang F, Xue A, Albizem M, Scott R, Stein EA. Reduction in lipoprotein(a)
with PCSK9 monoclonal antibody evolocumab (AMG 145): a pooled analysis of more than
1,300 patients in 4 phase II trials. J Am Coll Cardiol. 2014;63:1278–88.
Rader DJ, Mann WA, Cain W, Kraft H-G, Usher D, Zech LA, Hoeg JM, Davignon J, Lupien P,
Grossman M, Wilson JM, Brewer HB Jr. The low density lipoprotein receptor is not required
for normal catabolism of Lp(a) in humans. J Clin Invest. 1995;95:1403–8.
Rainwater DL, Manis GS, Kushwaha RS. Characterization of an unusual lipoprotein similar to
human lipoprotein a isolated from the baboon, Papio sp. Biochim Biophys Acta. 1986;877:75–8.
Rainwater DL, Maccluer JW, Stern MP, VandeBerg JL, Haffner SM. Effects of NIDDM on
lipoprotein(a) concentration and apolipoprotein(a) size. Diabetes. 1994;43:942–6.
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 35
Rath M, Pauling L. Immunological evidence for the accumulation of lipoprotein(a) in the athero-
sclerotic lesion of the hypoascorbemic guinea pig. Proc Natl Acad Sci U S A. 1990;87:9388–90.
Rath M, Niendorf A, Reblin T, Dietel M, Krebber H-J, Beisiegel U. Detection and quantifica-
tion of lipoprotein(a) in the arterial wall of 107 coronary bypass patients. Arteriosclerosis.
1989;9:579–92.
Renninger W, Wendt GG, Nawrocki P, Weigand H. [Contribution to problems of the Lp-system].
Humangenetik. 1965;1:658–67.
Ridker PM, Hennekens CH, Stampfer MJ. A prospective study of lipoprotein(a) and the risk of
myocardial infarction. JAMA. 1993;270:2195–9.
Roeseler E, Julius U, Heigl F, Spitthoever R, Heutling D, Breitenberger P, Leebmann J, Lehmacher
W, Kamstrup PR, Nordestgaard BG, Maerz W, Noureen A, Schmidt K, Kronenberg F, Heibges
A, Klingel R. Lipoprotein apheresis for lipoprotein(a)-associated cardiovascular disease: pro-
spective 5 years of follow-up and apolipoprotein(a) characterization. Arterioscler Thromb Vasc
Biol. 2016;36:2019–27.
Romagnuolo R, Scipione CA, Boffa MB, Marcovina SM, Seidah NG, Koschinsky
ML. Lipoprotein(a) catabolism is regulated by proprotein convertase subtilisin/kexin type 9
through the low density lipoprotein receptor. J Biol Chem. 2015;290:11649–62.
Rosengren A, Wilhelmsen L, Eriksson E, Risberg B, Wedel H. Lipoprotein(a) and coronary heart
disease: a prospective case-control study in a general population sample of middle aged men.
BMJ. 1990;301:1248–51.
Rudel LL, Greene DG, Shah R. Separation and characterization of plasma lipoproteins of rhesus
monkeys (Macaca mulatta). J Lipid Res. 1977;18:734–44.
Sandholzer C, Hallman DM, Saha N, Sigurdsson G, Lackner C, Császár A, Boerwinkle E,
Utermann G. Effects of the apolipoprotein(a) size polymorphism on the lipoprotein(a) concen-
tration in 7 ethnic groups. Hum Genet. 1991;86:607–14.
Sandholzer C, Feussner G, Brunzell J, Utermann G. Distribution of apolipoprotein(a) in the
plasma from patients with lipoprotein lipase deficiency with type III hyperlipoproteinemia: no
evidence for a triglyceride-rich precursor of lipoprotein(a). J Clin Invest. 1992a;90:1958–65.
Sandholzer C, Boerwinkle E, Saha N, Tong MC, Utermann G. Apolipoprotein(a) phenotypes,
Lp(a) concentration and plasma lipid levels in relation to coronary heart disease in a Chinese
population: evidence for the role of the apo(a) gene in coronary heart disease. J Clin Invest.
1992b;89:1040–6.
Sandholzer C, Saha N, Kark JD, Rees A, Jaross W, Dieplinger H, Hoppichler F, Boerwinkle E,
Utermann G. Apo(a) isoforms predict risk for coronary heart disease: a study in six popula-
tions. Arterioscler Thromb. 1992c;12:1214–26.
Scanu AM, Pfaffinger D, Lee JC, Hinman J. A single point mutation (Trp72–>Arg) in human
apo(a) kringle 4-37 associated with a lysine binding defect in Lp(a). Biochim Biophys Acta.
1994;1227:41–5.
Schachtl-Riess JF, Kheirkhah A, Grüneis R, Di Maio S, Schoenherr S, Streiter G, Losso JL,
Paulweber B, Eckardt K-U, Köttgen A, Lamina C, Kronenberg F, Coassin S. Frequent LPA
KIV-2 variants lower lipoprotein(a) concentrations and protect against coronary artery disease.
J Am Coll Cardiol. 2021;78:437–49.
Schaefer EJ, Lamon-Fava S, Jenner JL, McNamara JR, Ordovas JM, Davis CE, Abolafia JM,
Lippel K, Levy RI. Lipoprotein(a) levels and risk of coronary heart disease in men—the lipid
research clinics coronary primary prevention trial. JAMA. 1994;271:999–1003.
Schettler VJJ, Neumann CL, Peter C, Zimmermann T, Julius U, Roeseler E, Heigl F, Grutzmacher
P, Blume H. Current insights into the German Lipoprotein Apheresis Registry (GLAR)—
almost 5 years on. Atheroscler Suppl. 2017;30:50–5.
Schmidt K, Kraft HG, Parson W, Utermann G. Genetics of the Lp(a)/apo(a) system in an autoch-
thonous Black African population from the Gabon. Eur J Hum Genet. 2006;14:190–201.
Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of
lipoprotein(a). J Lipid Res. 2016;57:1339–59.
Scholz M, Kraft HG, Lingenhel A, Delport R, Vorster EH, Bickeböller H, Utermann G. Genetic
control of lipoprotein(a) concentrations is different in Africans and Caucasians. Eur J Hum
Genet. 1999;7:169–78.
36 G. Utermann
Schultz JS, Shreffler DC, Harvie NR. Genetic and antigenic studies and partial purification of
a human serum lipoprotein carrying the Lp antigenic determinant. Proc Natl Acad Sci U S
A. 1968;61:963–70.
Seed M, Hoppichler F, Reaveley D, McCarthy S, Thompson GR, Boerwinkle E, Utermann
G. Relation of serum lipoprotein(a) concentration and apolipoprotein(a) phenotype to coronary
heart disease in patients with familial hypercholesterolemia. N Engl J Med. 1990;322:1494–9.
Simons K, Ehnholm C, Renkonen O, Bloth B. Characterization of the Lp(a) lipoprotein in human
plasma. Acta Pathol Microbiol Scand B: Microbiol Immunol. 1970;78:459–66.
Sines J, Rothnagel R, Vanheel M, Gaubatz JW, Morrisett JD, Chiu W. Electron cryomicroscopy
and digital image processing of lipoprotein(A). Chem Phys Lipids. 1994;67-8:81–9.
Soutar AK, McCarthy SN, Seed M, Knight BL. Relationship between apolipoprotein(a) pheno-
type, lipoprotein(a) concentration in plasma, and low density lipoprotein receptor function in a
large kindred with familial hypercholesterolemia due to the pro664 -> leu mutation in the LDL
receptor gene. J Clin Invest. 1991;88:483–92.
Steyrer E, Durovic S, Frank S, Giessauf W, Burger A, Dieplinger H, Zechner R, Kostner GM. The
role of lecithin: cholesterol acyltransferase for lipoprotein(a) assembly. Structural integrity of
low density lipoproteins is a prerequisite for Lp(a) formation in human plasma. J Clin Invest.
1994;94:2330–40.
Stoffel W, Borberg H, Greve V. Application of specific extracorporeal removal of low density
lipoprotein in familial hypercholesterolaemia. Lancet. 1981;2:1005–7.
Suk DJ, Rifai N, Buring JE, Ridker PM. Lipoprotein(a), measured with an assay independent of
apolipoprotein(a) isoform size, and risk of future cardiovascular events among initially healthy
women. JAMA. 2006;296:1363–70.
Swallow DM, Gendler S, Griffiths B, Corney G, Taylor-Papadimitriou J, Bramwell ME. The
human tumour-associated epithelial mucins are coded by an expressed hypervariable gene
locus PUM. Nature. 1987;328:82–4.
Takegoshi T, Kitoh C, Haba T, Hirai J, Wakasugi T, Saga T, Yamazaki Y, Mabuchi H. A study of
the clinical significance of lipoprotein(a) in nephrotic syndrome. Jpn J Med. 1991;30:21–5.
Thompson GR, Seed M. Lipoprotein(a): the underestimated cardiovascular risk factor. Heart.
2014;100:534–5.
Tolbus A, Mortensen MB, Nielsen SF, Kamstrup PR, Bojesen SE, Nordestgaard BG. Kringle IV
type 2, not low lipoprotein(a), as a cause of diabetes: a novel genetic approach using SNPs
associated selectively with lipoprotein(a) concentrations or with Kringle IV type 2 repeats. Clin
Chem. 2017;63:1866–76.
Tomlinson JE, McLean JW, Lawn RM. Rhesus monkey apolipoprotein(a). Sequence, evolution,
and sites of synthesis. J Biol Chem. 1989;264:5957–65.
Trommsdorff M, Köchl S, Lingenhel A, Kronenberg F, Delport R, Vermaak H, Lemming L,
Klausen IC, Faergeman O, Utermann G, Kraft H-G. A pentanucleotide repeat polymorphism
in the 5′ control region of the apolipoprotein(a) gene is associated with lipoprotein(a) plasma
concentrations in Caucasians. J Clin Invest. 1995;96:150–7.
Tsimikas S, Brilakis ES, Miller ER, McConnell JP, Lennon RJ, Kornman KS, Witztum JL, Berger
PB. Oxidized phospholipids, Lp(a) lipoprotein, and coronary artery disease. N Engl J Med.
2005;353:46–57.
Tsimikas S, Viney NJ, Hughes SG, Singleton W, Graham MJ, Baker BF, Burkey JL, Yang
Q, Marcovina SM, Geary RS, Crooke RM, Witztum JL. Antisense therapy targeting
apolipoprotein(a): a randomised, double-blind, placebo-controlled phase 1 study. Lancet.
2015;386:1472–83.
Tsimikas S, Fazio S, Ferdinand KC, Ginsberg HN, Koschinsky ML, Marcovina SM, Moriarty PM,
Rader DJ, Remaley AT, Reyes-Soffer G, Santos RD, Thanassoulis G, Witztum JL, Danthi S,
Olive M, Liu L. NHLBI working group recommendations to reduce lipoprotein(a)-mediated
risk of cardiovascular disease and aortic stenosis. J Am Coll Cardiol. 2018;71:177–92.
Utermann G. The mysteries of lipoprotein(a). Science. 1989;246:904–10.
Utermann G. Genetic architecture and evolution of the lipoprotein(a) trait. Curr Opin Lipidol.
1999;10(2):133–41.
1 60 Years of Lp(a) Research: From Ouchterlonys Double Diffusion to Copy Number… 37
Utermann G. Lipoprotein(a). In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The metabolic
& molecular bases of inherited disease. 8th ed. New York: McGraw-Hill; 2001. p. 2753–87.
Utermann G, Weber W. Protein composition of Lp(a) lipoprotein from human plasma. FEBS Lett.
1983;154:357–61.
Utermann G, Wiegandt H. [Isolation and characterization of a lipoprotein with antigenic activity in
the Lp system]. Humangenetik. 1969;8:39–46.
Utermann G, Lipp K, Wiegandt H. Studies on the Lp(a)-lipoprotein of human serum. IV. The dis-
aggregation of the Lp(a)-lipoprotein. Humangenetik. 1972;14:142–50.
Utermann G, Hees M, Steinmetz A. Polymorphism of apolipoprotein E and occurrence of dysbet-
alipoproteinaemia in man. Nature. 1977;269:604–7.
Utermann G, Pruin N, Steinmetz A. Polymorphism of apolipoprotein E. III. Effect of a single poly-
morphic gene locus on plasma lipid levels in man. Clin Genet. 1979;15:63–72.
Utermann G, Kindermann I, Kaffarnik H, Steinmetz A. Apolipoprotein E phenotypes and hyper-
lipidemia. Hum Genet. 1984;65:232–6.
Utermann G, Menzel HJ, Kraft HG, Duba HC, Kemmler HG, Seitz C. Lp(a) glycoprotein phe-
notypes: inheritance and relation to Lp(a)-lipoprotein concentrations in plasma. J Clin Invest.
1987;80:458–65.
Utermann G, Hoppichler F, Dieplinger H, Seed M, Thompson G, Boerwinkle E. Defects in the low
density lipoprotein receptor gene affect lipoprotein (a) levels: multiplicative interaction of two
gene loci associated with premature atherosclerosis. Proc Natl Acad Sci U S A. 1989;86:4171–4.
Van der Hoek YY, Lingenhel A, Kraft HG, Defesche JC, Kastelein JJP, Utermann G. Sib-pair
analysis detects elevated Lp(a) levels and large variation of Lp(a) concentration in subjects
with familiar defective ApoB. J Clin Invest. 1997;99:2269–73.
Vuorio A, Watts GF, Schneider WJ, Tsimikas S, Kovanen PT. Familial hypercholesterolemia and
elevated lipoprotein(a): double heritable risk and new therapeutic opportunities. J Intern Med.
2020;287:2–18.
Wanner C, Rader D, Bartens W, Krämer J, Brewer HB, Schollmeyer P, Wieland H. Elevated plasma
lipoprotein(a) in patients with the nephrotic syndrome. Ann Intern Med. 1993;119:263–9.
Weisel JW, Nagaswami C, Woodhead JL, Higazi AA, Cain WJ, Marcovina SM, Koschinsky
ML, Cines DB, Bdeir K. The structure of lipoprotein(a) and ligand-induced conformational
changes. Biochemistry. 2001;40:10424–35.
Wendt GG. International Lp-workshop. Humangenetik. 1967;3:269–72.
White AL, Hixson JE, Rainwater DL, Lanford RE. Molecular basis for null lipoprotein(a) pheno-
types and the influence of apolipoprotein(a) size on plasma lipoprotein(a) level in the baboon.
J Biol Chem. 1994a;269:9060–6.
White AL, Rainwater DL, Hixson JE, Estlack LE, Lanford RE. Intracellular processing of Apo(A)
in primary baboon hepatocytes. Chem Phys Lipids. 1994b;67-8:123–33.
Wiegandt H, Lipp K, Wendt GG. [Identification of a lipoprotein with antigenic activity in the
Lp-system]. Hoppe Seylers Z Physiol Chem. 1968;349:489–94.
Williams-Blangero S, Rainwater DL. Variation in Lp(a) levels and apo(a) isoform frequencies in
five baboon subspecies. Hum Biol. 1991;63:65–76.
Ye Z, Haycock PC, Gurdasani D, Pomilla C, Boekholdt SM, Tsimikas S, Khaw KT, Wareham NJ,
Sandhu MS, Forouhi NG. The association between circulating lipoprotein(a) and type 2 diabe-
tes: is it causal? Diabetes. 2014;63:332–42.
Zysow BR, Lindahl GE, Wade DP, Knight BL, Lawn RM. C/T polymorphism in the 5′ untrans-
lated region of the apolipoprotein(a) gene introduces an upstream ATG and reduces in vitro
translation. Arterioscler Thromb. 1995;15:58–64.
Chapter 2
Lp(a) Biochemistry, Composition,
and Structure
Gerhard M. Kostner
Abbreviations
Historical Developments
In the early days, atherosclerosis research was dominated among others by two
patho-mechanisms, one related to lipids and lipoproteins and the other to hemostasis
and fibrinolysis. At that time, no one knew that Lp(a) constitutes a connection
between them. Since lipids are mostly water insoluble, they have to be transported in
blood complexed with amphipathic compounds such as phospholipids and apo-
lipoproteins. The qualitative and quantitative separation of lipoproteins (Lp) was
performed by (1) electrophoresis, (2) ultracentrifugation, and (3) by immune-affinity
methods such as ELISA or immune-specific adsorbers. The nomenclature of lipo-
proteins reflected the separation methods and there were basically three classifica-
tion systems: (1) based on the electrophoretic mobility yielding alpha-, ß-, and pre-ß
Lp; (2) density classes with the main fractions VLDL, LDL, and HDL; and (3) Lp
families with the main fractions Lp-A, Lp-B, and Lp-C (reviewed in Kostner 1983).
G. M. Kostner (*)
Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
e-mail: [email protected]
1
HDL 26 36
4 Lp(a) 31 42
VLDL 4 5
2
LDL 39 52
TChol 135
TG 121
100
50
0
0 5 10 mm 15
Fig. 2.1 Separation of Lp(a) by electrophoresis: Lp(a) migrates in gel electrophoresis as extra
pre-ß1 band and may be quantitated either after staining for lipids with Sudan black, or by
staining with a cholesterol reagent. Here, Lp(a) is separated by the Helena® Electrophoretic
system https://www.helena.com/. The concentration in mg/dL refers to Lp(a)-cholesterol as
staining was performed with a cholesterol dye. Since Lp(a) consists of some 25–30% of cho-
lesterol, Lp(a) mass in mg/dL may be calculated by multiplication with a factor of 3–4. (1 and
2): Plasma with Lp(a) of <30 mg/dL; (4) plasma with a Lp(a) concentration of 140 mg/dL. (From:
Kostner, K.M.; Kostner, G.M. Lp(a) and the Risk for Cardiovascular Disease: Focus on the
Lp(a) Paradox in Diabetes Mellitus. Int. J. Mol. Sci. 2022, 23, 3584. https://doi.org/10.3390/
ijms23073584)
In the first reports by K. Berg and his collaborators, Lp(a) was denominated “pre-
ß1” and/or “sinking pre-ß lipoprotein” (Berg 1963). In paper or agarose gel electro-
phoresis, Lp(a) migrated somewhat faster than pre-ß-Lp (VLDL) but slower than
alpha-Lp (HDL). This is depicted in Fig. 2.1. It must, however, be mentioned at this
point that the actual position of Lp(a) by electrophoretic methods depends on the
carrier material, the type, and pH of the electrophoresis buffer and the presence of
anti-coagulants such as heparin.
42 G. M. Kostner
% Distribution of Lp(a)
Density Gradient Ultracentrifugation
%
VLDL 5,8
2,7
LDL
10,9
20,1
Lp(a) 44,6
HDL2 3,2
1,0
8,2
HDL3
BOTTOM 3,7
Fig. 2.2 Separation of Lp(a) by density gradient ultracentrifugation that highlights the heteroge-
nous nature of Lp(a). After prestaining all serum constituents with Coomassie blue, lipoproteins
were separated by ultracentrifugation in the SW-41 Rotor, Beckmann® for 24 h at 40,000 rpm. In
the particular plasma, some 75% of Lp(a) was found in the HDL1 region, the rest distributed
between the top up to the bottom fraction. (From: Kostner, K.M.; Kostner, G.M. Lp(a) and the Risk
for Cardiovascular Disease: Focus on the Lp(a) Paradox in Diabetes Mellitus. Int. J. Mol. Sci.
2022, 23, 3584. https://doi.org/10.3390/ijms23073584)
2 Lp(a) Biochemistry, Composition, and Structure 43
94
67
43
30
20.1
14.4
Fig. 2.3 Western blot of Apo(a) isolated from plasma or urine. Plasma Lp(a) was purified as
described in Kostner et al. (1999) from a donor containing 90 mg/dL of Lp(a). The urine of the
same individual was concentrated 50-fold and both fractions were separated by SDS–polyacryl-
amide gel electrophoresis followed by W-blotting. St: protein molecular weight standard, the num-
bers indicating the mass in kDa. P refers to plasma and U refers to urine. Alb: Plasma albumin used
as a reference. (From: Kostner, K.M.; Kostner, G.M. Lp(a) and the Risk for Cardiovascular
Disease: Focus on the Lp(a) Paradox in Diabetes Mellitus. Int. J. Mol. Sci. 2022, 23, 3584. https://
doi.org/10.3390/ijms23073584)
situation is even more complex and much larger amounts of apo(a) are found in
VLDL or IDL. The exact morphology of Lp(a) outside of HDL1 has not been eluci-
dated in detail. Whether they are artifacts or true metabolic entities is not fully clear.
We characterized the apo(a) immune reactivity found in the bottom fraction of
human plasma and found that they consist of fragments created by Ca2+-dependent
proteases that are abundant on cell surfaces from several organs (Frank et al. 2001;
Gries et al. 1987). These fragments are not bound to lipoproteins and have masses
of some 50–150 kDa. Similar fragments of apo(a) are found in urine despite their
rather large size (Kostner et al. 2001) (Fig. 2.3). The amount of apo(a) fragments in
urine correlates significantly with the parent Lp(a) plasma concentration and we
therefore proposed the use of urinary apo(a) as a clinical chemical risk parameter
for atherosclerotic diseases (Kostner et al. 1996).
The preparation of pure Lp(a) with high yield from plasma with low concentra-
tion—mostly corresponding to large apo(a) isoforms is not an easy task. Therefore,
most investigators use plasma from donors with high Lp(a) values, that usually
contain small apo(a) isoforms. These Lp(a) specimens have a density not much dif-
ferent from that of LDL and therefore by ultracentrifugation used mostly as a first
step, large amounts of Lp(a) may be contaminated with LDL or may be lost in the
LDL fraction. There are two more hassles that must be considered by purification of
Lp(a) from various donors: (1) Lp(a) with large apo(a) isoforms in pure form are
prone to spontaneous precipitation and (2) Lp(a) associates with numerous other
44 G. M. Kostner
apo-Lp such as for example apoE, apoH (ß2-glycoprotein), and many kinds of other
serum constituents. These Lp(a) complexes may be dissociated by the addition of
amino acids such as Lys, Pro, hydroxy-Pro, and others. We therefore elaborated a
purification procedure that yielded Lp(a) with a purity of some 98%. It was sug-
gested that this material might be suitable for use as a “gold standard” in value
assignment for clinical chemical analyses (Kostner et al. 1999).
In short, blood is harvested from donors with Lp(a) concentrations of >30 mg/dL
and in the first step either citrate plasma or serum is prepared after coagulation.
After adding some preservatives (EDTA, BHT, PMSF, thiomersal), all lipoproteins
with d < 1.060 g/mL are separated by ultracentrifugation. Next, the density fraction
>1.060 < 1.125 g/mL is obtained by ultracentrifugation. After concentration to
approx. 10–20 mg/mL Lp(a)-cholesterol, proline at a final concentration of
0.1 mol/L is added. This step is essential as it dissociates all proteins from Lp(a)
other than apo(a) and apoB. In the next step, lipoproteins are separated by size
exclusion chromatography over Biogel A-15 m and the Lp(a) peak is harvested and
concentrated by pressure dialysis. For storage over a longer time period, pure Lp(a)
may be frozen at −20 to −70 °C in the presence of stabilizers. We tried several ones
including saccharose, polyethylene glycol, and others and at the end found out that
the 1:1 admixture of pure glycerol gave the best results. If prepared according to this
procedure, Lp(a) shows one band in agarose gel electrophoresis and is also virtually
pure by SDS–PAGE.
There is quite some variation in the composition of purified Lp(a) that is donor spe-
cific reflecting the isoform size, the lipid status of the plasma, and more. In Table 2.1.,
some average values are shown of Lp(a) isolated from fasting healthy donors in
comparison with LDL.
It must be emphasized here that the composition of Lp(a) shown in Table 2.1. is
just a snapshot of a fraction isolated from fasting plasma of normolipemic healthy
individuals. Lp(a) outside of the HDL1 density fraction or Lp(a) isolated from dys-
or hyperlipemic plasma may have quite significantly deviant structures and
compositions.
Fig. 2.4 Hypothetical model of Lp(a) showing the LDL core with apoB-100 as the major surface
protein and apo(a). 41 refers to K-IVT-1, 42 to K-IV-T2, and so one. 5 = kringle V and P = the
protease domain. (The figure was drawn by Timo Speer, Med. University of Homburg/Saar,
Germany. From: Kostner, K.M.; Kostner, G.M. Lp(a) and the Risk for Cardiovascular Disease:
Focus on the Lp(a) Paradox in Diabetes Mellitus.Int. J. Mol. Sci. 2022, 23, 3584. https://doi.
org/10.3390/ijms23073584)
46 G. M. Kostner
Fig. 2.5 cDNA structure of apo(a) in comparison to plasminogen. In apo(a), kringle-IV’s (K-IV)
homologous to plasminogen are repeated several times. There is also one K-V like domain and a
protease domain in apo(a) (P) with a homology of 94 to that of plasminogen. The Arg of plasmino-
gen is replaced in apo(a) by Ser and thus the protease cannot by activated in Lp(a) by t-PA or
urokinase. (From: Kostner, K.M.; Kostner, G.M. Lp(a) and the Risk for Cardiovascular Disease:
Focus on the Lp(a) Paradox in Diabetes Mellitus.Int. J. Mol. Sci. 2022, 23, 3584. https://doi.
org/10.3390/ijms23073584)
2 Lp(a) Biochemistry, Composition, and Structure 47
High-resolution structures of whole apo(a) have not been published, but there
exist a few crystal structures of recombinant apo(a)-kringle-IV’s (Ye et al. 2001).
The crystal structure refined to a resolution of 1.45 Å revealed important structural
features of kringle-IV-T-7 that are postulated to be responsible for the interaction
with Lysine groups.
The K-IV domains are connected by linker regions that are highly glycosylated by
N- and O-linked sugars. The best characterization of the apo(a) carbohydrate
arrangement has been published by Garner et al. (2001) who demonstrated that
approx. 20% of the oligosaccharide structures consist of two major Asp-linked
N-oligosaccharides. N-glycans are complex biantennary structures in either a mono-
or disialylated state. The remaining 80% of the sugars are Ser/Thr O-linked oligo-
saccharides and are present in all apo(a) isoforms. The majority consist of the
mono-sialylated core type-I structure, NeuNAcα2-3Galβ1-3GalNAc, and the
remaining consist of disialylated and non-sialylated O-glycans. The latter finding
prompted us to elucidate the possibility that Lp(a) might bind to the asialo-
glycoprotein receptor (ASGPR) on liver cells.
It is well known that the sialic acid content of glycoproteins regulates their fate in
circulating blood. Aged protein may be devoid of their terminal sialic acid due to the
action of specific sialidases. There are specific asialo-glycoprotein receptors on liver
cells that very effectively bind and catabolize such asialo-glycoproteins. This is sche-
matically displayed in Fig. 2.6. ASGPR may be specific for galactose or mannose.
In a study we published in 2003 (Hrzenjak et al. 2003), it was important that
antimicrobial agents, among other preservatives, were added immediately after
drawing blood and also throughout the different isolation steps, in order to obtain
“native Lp(a)” as far as possible. The metabolic experiments were carried out in
hedgehogs, the only animal species with the exception of old-world monkeys that
synthesize an Lp(a) like lipoprotein. In vivo experiments in these animals revealed
that desialylated Lp(a) is catabolized 25 times faster than native Lp(a) and is almost
exclusively taken up by the liver. Concomitant injection of asialo-Lp(a) with asialo-
orosomucoide to hedgehogs reduced the half-life of asialo-Lp(a) to values observed
for native Lp(a). Mannan, the competitive inhibitor for the mannose-specific
ASGPR, had no effect. Similar results were observed in wild-type mice where desi-
alylated Lp(a) is catabolized 50–100 times faster than sialylated Lp(a). Here also,
only asialo-orosomucoide, but not mannan, acted as a competitive inhibitor.
48 G. M. Kostner
Fig. 2.6 The sugar moiety Does not Binds to ASGPR receptor
of glycoproteins consists bind to ASGPR on liver cells
of complex antennary and is internalized
structures, many of them
containing sialic acid (Sia)
as terminal sugar. Aged
proteins may be targets of
sialidases that cleave sialic
acid off, and in turn,
Gal Gal
galactose (Gal) or
mannoses may be exposed
to the surface. These latter
sugars are avidly bound to GlcNAc GlcNAc
asialo-glycoprotein
receptors (ASGPR) on core sugars core sugars
liver cells and catabolized
protein protein
Sialoglycoprotein Asialoglycoprotein
Whether or not the results of these experiments might be transferable to the situ-
ation in humans cannot be answered from these experiments. Any Lp(a) preparation
isolated from human plasma may contain only a small fraction of the non-sialylated
Lp(a), because of its very short half-life in the circulating blood of the donor.
Whether such putative non-sialylated Lp(a) is directly secreted from human liver or
is generated during circulation in blood or might be an artifact generated during
Lp(a) preparation remains to be investigated. It is, however, important to note that
we also carried out similar metabolic studies with native Lp(a) in wild-type mice
and in ASGPR knockout mice. In the ko-mice, the HL-2 subunit of the ASGPR was
absent. When native freshly isolated Lp(a) was injected in either mice, we observed
a measurable retardation of the Lp(a) catabolism in the KO-mice as compared to
wild-type mice. The amount of Lp(a) labeled with the non-degradable isotope
[125I]
tyramine cellobiose accumulating in the liver of knockout mice was signifi-
cantly lower compared with wild-type mice (Hrzenjak et al. 2003). We believe this
is a strong argument for the ASGPR pathway being indeed involved in Lp(a) catab-
olism in humans.
We also observed that some 87% of intravenously injected asialo-Lp(a) was
cleared by ASGPR-negative mice within 1 h. Previous studies by Roos et al. (1983)
demonstrated two galactose-specific receptors in rat liver: The Kupffer cell-specific
glycoprotein receptor readily interacts with galactose-exposing particles of the size
of LDL (Fadden et al. 2003). We assume that this receptor might be responsible for
the clearance of asialo-Lp(a) in our ASGPR ko mouse model. Yet, the Kupffer cell-
specific glycoprotein receptor is not expressed in humans, as human genome analy-
sis reveals a pseudogene that is not translated into a protein (Van Berkel et al. 1985).
Taken together, the role of the ASGPR for the catabolism of Lp(a) in humans is far
from being clear and requires further investigation.
2 Lp(a) Biochemistry, Composition, and Structure 49
This topic is thoroughly covered in the article by G. Utermann in this book, and
thus, I mention in this paragraph only a few aspects that might be relevant for struc-
tural considerations of Lp(a). Among the numerous polymorphisms and mutations
described elsewhere in this book, there is in particular one mutation relevant here,
the truncated form of apo(a) expressed by the so-called null allele. The first report
on truncated apo(a) was published by M. Ogorelkova from the laboratory of
G. Utermann in Innsbruck (Ogorelkova et al. 1999). Gene sequencing of APOA in
Caucasian individuals with almost zero Lp(a) levels revealed a G→A substitution at
the 1+ donor splice site of K-IV type 8 introns. This nonsense mutation led to the
expression of a truncated form of apo(a) that consisted of a N-terminal fragment
lacking all entities after kringle-IV-T7. Since K-IV T9 in apo(a) contains the single
free –SH group that is necessary for the covalent binding to apoB-100, such trun-
cated apo(a) are not able to stably assemble with LDL. Interestingly, there are small
amounts of free truncated apo(a) found in plasma indicating that the liver secretes
such apo(a) mutants, but it seems that they are very rapidly catabolized. This opens
up the question whether plasma Lp(a) levels might be drastically reduced in general
by inhibition of Lp(a) assembly.
We addressed this question in in vivo and in vitro studies using tranexamic acid for
the inhibition of Lp(a) assembly (Frank et al. 1999). In vivo studies were performed
with single transgenic apo(a) mice or double transgenic apo(a):apoB mice both of
them carrying the relevant human genes. The assembly of apo(a) in the test tube
may be fully inhibited by Lys or analogs thereof such as delta-amino valeric d-AVA
acid or tranexamic acid TXA, the latter being the strongest inhibitor that is also
known to inhibit fibrinolysis by plasmin (displayed in a cartoon in Fig. 2.7). The
mice were fed 150 mg/dL of d-AVA or TXA for 1–2 weeks and the plasma apo(a)
and Lp(a) concentrations were followed over time. In the double transgenic Lp(a)
mice, in contrast to what we expected, the concentration of Lp(a) rose after 1 week
of feeding to almost twice the value observed in the absence of d-AVA or TNX feed-
ing. This was a transient effect since after omitting the inhibitors from the chow, the
Lp(a) concentration returned to the pretreatment values. We also revealed that the
increase of plasma Lp(a) was fully accounted for by the presence of genuine Lp(a)
and not by free apo(a). When single transgenic apo(a) mice were treated in a similar
protocol apo(a) rose after 1 week by 57% and returned to pretreatment values as
well. Similar results were obtained by d-AVA feeding, yet they were less pro-
nounced. We then harvested the livers of mice treated with TNX or d-AVA and
found that their concentration in the liver was significantly lower than without
treatment.
50 G. M. Kostner
Fig. 2.7 Possible models of Lp(a) assembly and inhibition by tranexamic acid. There are currently
two models of Lp(a) assembly discussed: (1) apo(a) is biosynthesized in the liver and after passage
through the Golgi apparatus it binds to the surface of liver cells. Bypassing LDL associate with
apo(a) and both components are covalently linked by a disulfide bridge. The first step of assembly,
the interaction of kringles-4 with Lys groups of apoB-100 may be competed for by Lys analogs
such as Tranexamic acid. Free apo(a) not complexed to LDL might be degraded by hydrolytic
enzymes. Alternatively, the assembly may take place intracellularly in the liver cells. (From:
Kostner, K.M. and Kostner, K.M. Lipoprotein(a): a historical appraisal. J Lipid Res. 2017, 58,
1–14. https://doi.org/10.1194/jlr.R071571)
to liver cell surfaces and are partly degraded before assembling with LDL to intact
circulating Lp(a).
Given the fact that Lp(a) is a strong independent risk factor for atherosclerosis and
CAD it was speculated that individuals with apo(a) mutations or polymorphisms
that cause reduced plasma Lp(a) levels might be at a lower risk for CAD. This was
addressed in a cohort of the PROCARDIS study published by Kyriakou (Kyriakou
et al. 2014). Indeed, it was found that the LPA null allele as identified by the
rs41272114 SNP not only is associated with reduced plasma Lp(a) concentrations
but also with a significantly reduced CAD risk.
Among all the mutations in the whole apo(a) gene including the promoter region
and regulatory cis-acting regions described in the literature, there are two publica-
tions from the Innsbruck laboratory that need attention (Noureen et al. 2015;
Coassin et al. 2019). It was known since the first published report on the LPA gene
sequence by McLean and Lawn (McLean et al. 1987) that there are silent mutations
in the repetitive gene region coding for the K-IV T2. These variations were called
K-IV2 A and K-IV2 B. Despite of the difference in the gene sequence, the A and B
alleles translated into the same apo(a) protein sequence. Sequencing the apo(a) gene
is not an easy task because of the repetitive structure caused by K-IV T2 and the
homology of the non-repetitive kringles K-IV-1 and K-IV-3 to K-IV 10. Nevertheless,
Stefan Coassin and his colleagues succeeded to elaborate a sophisticated protocol
that allowed apo(a) sequencing in larger quantities. In these studies, gene variants in
the K-IV-T2 region were identified that translated drastically into plasma Lp(a) con-
centrations. The exact mechanisms on a molecular level are not fully clear, but the
results highlight the importance of the polymorphic APOA gene sequence for the
apo(a) protein expression and plasma concentration of Lp(a).
The reason why apo(a) only assembles with LDL and not with other serum proteins
that may have Lys groups exposed to the surface has never been fully explored. As
a matter of fact, the greatest portion of full length apo(a)—if not all is found on
apoB-100 containing lipoproteins in human plasma. This led us to assume that the
composition and morphology of LDL have just the right prerequisites for this
assembly. Two observations published earlier by our laboratory strongly support
this concept.
Numerous reports in the literature demonstrate that Lp(a) hardly binds directly to
the LDL receptor (Hofer et al. 1997). This is at a first consideration surprising, as
some 50% of the Lp(a) moiety consist of apoB-100. The most plausible explanation
that Lp(a) is not bound to the LDL-R would be that the large glycoprotein apo(a)
52 G. M. Kostner
masks the epitope in apoB-100 responsible for LDL receptor binding. This concept
is strongly supported by our studies of patients suffering from familial defective
apoB-100 (FDB) (Durovic et al. 1994). FDB patients express a mutant apoB-100
where Arg at position 3500 is substituted by Gln. This substitution causes the bio-
synthesis of an LDL particle that has a strongly reduced binding affinity to the LDL
receptor and in turn patients with FDB are hypercholesterolemic.
In the investigations published by Ernst Steyrer et al. (1994), we studied the in vitro
assembly of Lp(a) by mixing purified LDL with recombinant apo(a) and followed the
covalent linkage of both components. Whereas wild-type LDL mixed with apo(a)
complexed under the given experimental conditions between 15% and 44% with
apo(a), LDL from a homozygous FDB patient showed only 2–16% association. The
corresponding figure using LDL from heterozygous FDB individuals was 2–30%.
Moreover, we found that in heterozygous FDB patients the ratio of defective to wild-
type apoB100 in Lp(a) is significantly lower than in LDL from the same patients.
These results strongly suggest that the epitope in apoB-100 that is involved in LDL
receptor binding is also highly relevant for apo(a) binding and covalent linkage.
Another example for the importance of the right morphology of LDL to warrant
an ideal assembly has been published in 1994 by our group. It is known that patients
suffering from LCAT deficiency (LCAT-D) have a grossly altered plasma lipopro-
tein pattern. We made also the observation that 9 heterozygous LCAT-D patients
had only 2–13 mg/dL of Lp(a) whereas the Lp(a) concentration in the non-affected
siblings was significantly higher (Ernst Steyrer et al. 1994). Eleven of the studied
homozygous LCAT-D patients exhibited plasma Lp(a) levels of virtually zero. The
morphology of lipoproteins in the LDL region isolated from homozygous LCAT-D
patients was grossly altered with large vesicles and small spheres and there was an
almost complete lack of cholesteryl esters (Fig. 2.8). When LCAT-D LDL were
incubated with recombinant apo(a) for 20 h at 37 °C, no complex of
Fig. 2.8 Negative-stain electron microscopy of LDL (density 1.030–1.063 g/mL) and Lp(a) iso-
lated from a healthy control individual and from a homozygous patient suffering from LCAT
deficiency. (From: Kostner, K.M.; Kostner, G.M. Lp(a) and the Risk for Cardiovascular Disease:
Focus on the Lp(a) Paradox in Diabetes Mellitus. Int. J. Mol. Sci. 2022, 23, 3584. https://doi.
org/10.3390/ijms23073584)
2 Lp(a) Biochemistry, Composition, and Structure 53
apoB100:r-apo(a) had been formed (i.e., no assembly to Lp(a) took place). Normal
LDL under the same conditions showed complete assembly to Lp(a). We concluded
that the integrity of the LDL structure is a prerequisite for the biosynthesis of genu-
ine Lp(a). Furthermore, it appears that the lack of complexing apo(a) to bona fide
LDL leads to a fast catabolism or degradation of the expressed apo(a). Thus, not
only is an intact LCAT activity necessary, but there must also be an abundance of
“normal” native LDL. These factors substantially regulate plasma Lp(a) metabo-
lism and serum Lp(a) levels.
References
Alaupovic P, Kostner G, Lee DM, McConnathy WJ, Magnani HN. Peptide composition of human
plasma lipoproteins A, B and C. Exp Ann Biochim Med. 1972;31:145–60.
Allison AC, Blumberg BS. An isoprecipitation reaction distinguishing human serum protein types.
Lancet. 1961;1:634–7.
Berg K. A new serum type system in man—the Lp system. Acta Path Microbiol Scand.
1963;59:369–82.
Bundschuh A, Vogt A. Die Häufigkeit des Merkmals Lp(a) in der Berliner Bevölkerung.
Humangenetik. 1965;1:379–82.
Coassin S, Schönherr S, Weissensteiner H, Erhart G, Forer L, Lee Losso J, Lamina C, Haun M,
Utermann G, Paulweber B, Specht G, Kronenberg F. A comprehensive map of single-base
polymorphisms in the hypervariable LPA kringle IV type 2 copy number variation region. J
Lipid Res. 2019;60:186–99.
Durovic S, März W, Frank S, Scharnagl H, Baumstark MW, Zechner R, Kostner GM. Decreased
binding of apolipoprotein(a) to familial defective apolipoprotein B-100 (Arg3500→Gln). J Biol
Chem. 1994;269:30320–5.
Ernst Steyrer E, Durovic S, Frank S, Gissauf W, Burger A, Dieplinger H, Zechner R, Kostner
GM. The role of lecithin: cholesterol acyltransferase for lipoprotein(a) assembly structural
integrity of low density lipoproteins is a prerequisite for Lp(a) formation in human plasma. J
Clin Invest. 1994;9:2330–40.
Fadden AJ, Holt OJ, Drickamer K. Molecular characterization of the rat Kupffer cell glycoprotein
receptor. Glycobiology. 2003;13:529–37.
Fless G. Heterogeneity of particles containing the apo-apo(a) complex. In: Scanu AM, editor.
Lipoprotein(a). New York: Academic Press; 1990. p. 41–51.
Fless GM, ZumMallen ME, Scanu AM. Isolation of apolipoprotein(a) from lipoprotein(a). J Lipid
Res. 1985;26:1224–9.
Frank S, Hrazenjak A, Blaschitz G, Dohr G, Kostner GM. Role of various tissues in apo(a) frag-
mentation and excretion of fragments by the kidney. Eur J Clin Invest. 2001;31:504–412.
Fredrickson DS, Levy RI, Lees RS. Fat transport in lipoproteins—an integrated approach to mech-
anisms and disorders. N Engl J Med. 1967;276:148–56.
Garner B, Merry AH, Royle L, Harvey DJ, Rudd PM, Thillet J. Structural elucidation of the N- and
O-glycans of human apolipoprotein(a). J Biol Chem. 2001;276:22200–8.
Gofman JW, Lindgren FT, Elliott H. Ultracentrifugal studies of lipoproteins of human serum. J
Biol Chem. 1949;179:937–9.
Gries A, Nimpf J, Nimpf M, Wurm H, Kostner GM. Free and apoB-associated Lp(a) specific pro-
tein in human serum. Clin Chim Acta. 1987;164:93–100.
54 G. M. Kostner
Hofer G, Steyrer E, Kostner GM, Hermetter A. LDL-mediated interaction of Lp[a] with HepG2
cells: a novel fluorescence microscopy approach. J Lipid Res. 1997;38(241):1–2421.
Frank S, Hrzenjak A, Kostner K, Sattler W, Kostner GM. Effect of tranexamic acid and
N-aminovaleric acid on lipoprotein(a)metabolism in transgenic mice. Biochim Biophys Acta.
1999;1438:99–110.
Hrzenjak A, Frank S, Wo X, Zhou Y, Van Berkel T, Kostner GM. Galactose specific asialo glyco-
protein receptor is involved in lipoprotein(a) catabolism. Biochem J. 2003;376:765–71.
Kostner GM. Lp(a) lipoproteins and the genetic polymorphisms of lipoprotein B. In: Day CE,
Levy RS, editors. Low density lipoproteins. New York: Plenum Press; 1976. p. 229–69.
Kostner GM. Apolipoproteins and lipoproteins of human plasma: significance in health and dis-
ease. Adv Lipid Res. 1983;20:1–41.
Kostner GM, Avogaro P, Cazzolato G, Marth E, Bittolo-Bon G, Quinci GB. Lipoprotein Lp(a) and
the risk for myocardial infarction. Atherosclerosis. 1981;38:51–61.
Kostner KM, Maurer G, Huber K, Stefenelli T, Dieplinger H, Steyrer E, Kostner GM. Urinary
excretion of apo(a) fragments. Role in apo(a) catabolism. Arteriosc Thromb Vasc Biol.
1996;16:905–11.
Kostner GM, Ibovnik A, Holzer H, Grillhofer H. Preparation of a stable fresh frozen primary
lipoprotein[a] standard. J Lipid Res. 1999;40:2255–63.
Kostner K, Spitzauer S, Rumpold H, Maurer G, Knipping G, Hrzenjak A, Frank S, Kostner
GM. Urinary excretion of apolipoprotein(a): relation to other plasma proteins. Clin Chim Acta.
2001;304:29–37.
Kyriakou T, Seedorf U, Goel A, Hopewell JC, Clarke R, Watkins H, Farrall M, on behalf of the
PROCARDIS Consortium. A common LPA null allele associates with lower lipoprotein(a)
levels and coronary artery disease risk. Arterioscler Thromb Vasc Biol. 2014;34:2095–9.
McLean JW, Tomlinson JE, Kuang WJ, Eaton DL, Chen EY, Fless GM, Scanu AM, Lawn
RM. cDNA sequence of human apolipoprotein(a) is homologous to plasminogen. Nature.
1987;330:132–27.
Morrisett JD, Gaubatz JW, Knapp RD, Guevara JG Jr. Structural properties of apo(a): a major apo-
protein of human lipoprotein(a). In: Scanu AM, editor. Lipoprotein(a). New York: Academic
Press; 1990. p. 41–51.
Noureen A, Fresser F, Utermann G, Schmidt K. Sequence variation within the KIV-2 copy number
polymorphism of the human LPA gene in African, Asian, and European populations. PLoS
One. 2015:1–23. https://doi.org/10.1371/journal.pone.0121582.
Ogorelkova M, Gruber A, Utermann G. Molecular basis of congenital Lp(a) deficiency: a frequent
apo(a) null mutation in Caucasians. Hum Mol Genet. 1999;8:2087–96.
Roos PH, Kolb-Bachofen V, Schlepper-Schafer J, Monsigny M, Stockert RJ, Kolb H. Two
galactose-specific receptors in the liver with different function. FEBS Lett. 1983;157:253–6.
Utermann G. The mysteries of lipoprotein(a). Science. 1989;246:904–10.
Van Berkel TJ, Kruijt JK, Spanjer HH, Nagelkerke JF, Harkes L, Kempen HJ. The effect of a
water-soluble Tris-galactoside-terminated cholesterol derivative on the fate of low density lipo-
proteins and liposomes. J Biol Chem. 1985;260:2694–9.
Weisel JW, Nagaswami C, Woodhead JL, Higazi AA-R, Cain WJ, Marcovina SM, Koschinsky
ML, Cines DB, Bdeir K. The structure of lipoprotein(a) and ligand-induced conformational
changes. Biochemistry. 2001;40:10424–35.
Ye Q, Rahman MN, Koschinsky ML, JIA Z. High-resolution crystal structure of apolipoprotein(a)
kringle IV type 7: insights into ligand binding. Protein Sci. 2001;10:1124–9.
Chapter 3
Genetics of Lipoprotein(a)
Gerd Utermann
Introduction
The genetics of lipoprotein(a) [Lp(a] is both, simple and complex at the same time.
Initially described as a dominant trait (Berg 1963; Berg and Mohr 1963) with two
immunologically defined phenotypes, Lp+ and Lp−, where Lp+ is dominant over
Lp−, it is now well established that Lp(a) is a quantitative trait with a very broad
distribution in all studied populations (Schmidt et al. 2016). Lp(a) concentrations
vary more than 1000-fold between individuals in the same population and range
from undetectable to >200 mg/dL in healthy individuals (Utermann 1989). In all
populations, the distribution of Lp(a) levels is skewed and far from Gaussian. In
Europeans, most individuals have low and few have very high Lp(a) concentrations.
Mean and median Lp(a) concentrations, the distribution of Lp(a) concentrations
(e.g., skewness) vary widely between human ethnic groups. Sub-Saharan Africans
have by far the highest levels and lowest skewness. Compared to Europeans, they
are two to fourfold higher. The highest concentrations were reported for Black
Sudanese (Sandholzer et al. 1991) and Gabonese Bantu (Schmidt et al. 2006).
Differences exist also within populations from major ethnic groups. In Europe,
Finns have the lowest reported concentrations (Erhart et al. 2018; Waldeyer et al.
2017). Asian populations are even more heterogeneous. Most studies report very
low levels and highly skewed Lp(a) distributions in East Asians (Japanese and
Chinese). South-East Asians, e.g., Indians and Thais, have concentrations between
Europeans and East Asians and Africans (Schmidt et al. 2016; Sandholzer et al.
1991; Helmhold et al. 1991; Enkhmaa et al. 2016) (Fig. 3.1). Notably different
Lp(a) concentrations have also been reported for the same or similar population,
e.g., Chinese (Sandholzer et al. 1991; Helmhold et al. 1991; Enkhmaa et al. 2016).
G. Utermann (*)
Institute for Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
e-mail: [email protected]
Gabonese
Khoisan
Egyptians
Black South Africans
Asian Indians North
Asian Indians South
Chinese
Inuits
Thai
Indonesians
Russians
Austrians
Japanese
Danes
Finns
In Lp(a) concentraion
Fig. 3.1 Distribution of Lp(a) concentrations in 15 populations. Median, range, and 95% confi-
dence intervals are given (in ln mg/dL). Colors denote continental groups. (Data from Sandholzer
et al. 1991; Schmidt et al. 2006; Kraft et al. 1996; Khalifa et al. 2015; Scholz et al. 1999;
Trommsdorff et al. 1995)
For understanding the genetics of the Lp(a) trait, a brief description of the structure
of Lp(a) is necessary. Lp(a) is a complex, assembled from one low-density lipopro-
tein (LDL) and the high molecular weight glycoprotein apolipoprotein(a) [apo(a)]
which confers the immunological specificity to the particle. Both are held together
by non-covalent binding of domains in apo(a) to apoB in LDL and by covalent bind-
ing through a single disulfide bridge between apo(a) and apoB (Schmidt et al. 2016;
Brunner et al. 1993; Koschinsky et al. 1993; McCormick et al. 1995; Callow and
Rubin 1995; Ernst et al. 1995; Gabel and Koschinsky 1998). Apo(a) is highly gly-
cosylated, does not bind lipids, and is not a true apolipoprotein. The protein has a
high homology to plasminogen from which it has evolved during primate evolution
by a series of gene duplication, deletions, domain duplications, and point mutations
(McLean et al. 1987; Tomlinson et al. 1989). The result is an odd protein consisting
of a signal sequence, ten different domains with homology to PLG kringle type IV
(KIV-1 to KIV-10), one PLG-derived kringle type V(KV), and a protease domain
with AA-substitutions rendering it inactive toward plasmin substrates.
One of the kringles, KIV-2 occurs in multiple identical copies and varying copy
numbers in the gene (Utermann 1989; Lackner et al. 1991, 1993; Boerwinkle et al.
1992; Kraft et al. 1992) (Fig. 3.2). The genomic size of one KIV-2 unit is 5.6 kb.
This variable number of repeats (VNTR) is translated and transcribed into protein
resulting in a size polymorphism of apo(a) (Schmidt et al. 2016; Utermann 1989;
Utermann et al. 1987). The size polymorphism of LPA/apo(a) has been demon-
strated at the protein level by polyacrylamide- or agarose-gel-electrophoresis/Western
blotting of plasma using antibodies against apo(a)/Lp(a) (Fig. 3.3a) or at the DNA
level by enzymatic digestion of genomic DNA using appropriate DNAses followed
by pulsed-field-gel electrophoresis and Southern blotting with KIV-2-specific
probes (Fig. 3.3b). Using enzymes, e.g., KpnI that cut the genomic DNA only out-
side the KIV-2 repeats (Fig. 3.3), the complete block of DNA can be cut out and the
number of repeats determined from its size (Lackner et al. 1991; Boerwinkle et al.
1992; Kraft et al. 1992). A technique, which has been used only in one single pub-
lication and needs special skills in molecular cytogenetics, is fiber-FISH. This
enabled to visualize and count the number of KIV-2 repeats of single alleles under
the microscope (Erdel et al. 1999) (Fig. 3.3). The frequency of KIV-2 alleles varies
significantly between different ethnic groups (Fig. 3.4a).
The KIV-2 VNTR held the key to the understanding of the genetics of the Lp(a)-
trait. The size of apo(a) isoforms was shown to be inversely correlated with Lp(a)
concentration in plasma (Fig. 3.4b). On average, small isoforms corresponding to
low KIV-2 copy number were associated with high Lp(a) in plasma and large
58 G. Utermann
Fig. 3.2 Panel (a): Exon-Intron structure of the human LPA gene. Domains are represented in dif-
ferent colors (KIV-2 = red; KIV-1 and KIV-3 to KIV-10 = black; KV = green; protease domain
purple). Indicated are the KIV-2 VNTR, cutting sites for KpnI (Lackner et al. 1991) and rs3798200
(Clarke et al. 2009) (Adapted from Noureen et al. 2015 with permission). Panel (b): Fibre-FISH
image of LPA alleles with four and nineteen KIV-2 repeats (colored in red-yellow; count yellow
dots flanked by red). (Copy of Fig. 1b in Erdel M et al. Nat. Genet 1999; 21:357–358)
isoforms (high KIV-2 copy numbers) with low concentrations (Schmidt et al. 2016;
Utermann et al. 1987). There is, however, wide variation within alleles defined by
copy number (allele-associated Lp(a) levels) especially for low copy number alleles
(Fig. 3.5).
The analytical techniques agarose-gel-electrophoresis/Western blotting and
PFGE/Southern blotting achieve a similar resolution and with both >30 alleles of
different sizes have been demonstrated (Schmidt et al. 2016). They are, however, not
equivalent but rather complement each other. By PFGE/Southern blotting the KIV-2
genotype can be precisely determined and >95% of individuals were found to be
heterozygotes. It can, however, not be measured which concentration of Lp(a) is
associated with each allele. By contrast, agarose-gel-electrophoresis/Western blot-
ting allows assignment of Lp(a) concentration to both alleles if total Lp(a) concentra-
tion is known. Due to the extremely wide range of concentrations and the sensitivity
limits of Western blotting, apo(a) alleles associated with very low or absent Lp(a) in
plasma cannot be seen (so-called “null” alleles). Hence, in a considerable number of
samples, only one isoform is seen on the blot. Further for such samples, it cannot be
distinguished whether they are from a rare homozygote or from individuals with one
“null” allele. The frequency of “null” alleles has been estimated from 1% to 29%
depending on the population and sensitivity of Western blotting (Kraft et al. 1996;
Marcovina et al. 1996; Gaw et al. 1994) which is interesting in view of the frequency
of true “null” alleles defined today at the molecular level (Ogorelkova et al. 1999; Di
Maio et al. 2020; Morgan et al. 2020; Mukamel et al. 2021).
3 Genetics of Lipoprotein(a) 59
a b
35
27
26
23
22
20
19
15
14
K5 K5 marker
Southern blot Western blot
Fig. 3.3 Determination of the Size of Alleles of the KIV-2 VNTR by PFGE/Southern Blotting
(Panel a) and Western Blotting (Panel b). The same four samples from one family were analyzed.
The allele with 15 KIV repeats (corresponding to 6 KIV-2 repeats) in the lane denoted K5 in the
Southern blot by a black arrow is not expressed (=null allele). The corresponding isoform is miss-
ing in the Western blot (red arrow). (Modified from Noureen et al. 2015 with permission)
Fig. 3.4 Panel (a): Frequency distribution of binned KIV-2 VNTR alleles (numbers denote KIV
repeats including KIV-1 and KIV-3 to 10) in pooled data from three continental groups (Adapted
from Schmidt et al. 2016 with permission). Panel (b): Lp(a) concentrations by ancestry associated
with binned KIV-2 VNTR alleles. Note large differences in concentrations of Lp(a) associated with
KIV-2 alleles of the same binned size category between the major continental groups. (Data from
Schmidt et al. 2006; Kraft et al. 1996; Scholz et al. 1999)
PCR (qPCR) (Kamstrup et al. 2009). This allowed neither identification of the gen-
otype nor assignment of allele-associated Lp(a) concentrations. Second, SNPs in
the LPA gene (see below) in LD with the KIV-2 repeats were used as proxies (Clarke
et al. 2009). Both approaches allow to detect strong associations. A serious caveat
is, however, that LDs of SNPs with KIV-2 alleles may differ significantly between
ethnic groups and use as proxy for Lp(a) concentration may lead to grossly false
3 Genetics of Lipoprotein(a) 61
150
allele associated Lp(a) [mg/dl]
125
100
75
50
25
0
10 15 20 25 30 35 40 45
KIV-CNV size [number of KIV repeats]
Fig. 3.5 Illustration of the inverse correlation of KIV-2 VNTR allele size with Lp(a) concentration
(allele-associated Lp(a) concentration) in Gabonese Bantu. (Data from Schmidt et al. 2006.
Adapted from Schmidt et al. 2016 with permission)
results. This has been demonstrated for rs3798220 which is associated with low
KIV-2 copy number and high Lp(a) in Europeans (Clarke et al. 2009) and median/
high copy numbers and low Lp(a) in East and South East Asians (Khalifa et al.
2015) (Fig. 3.6).
The apo(a) VNTR explains about 40–70% of the heritability of the quantitative
Lp(a) trait depending on study design and population (Schmidt et al. 2006;
Boerwinkle et al. 1992; Kraft et al. 1992). The KIV-2 VNTR was further used in
family and sib-pair linkage studies to estimate the heritability of Lp(a) explained by
the LPA locus. This demonstrated that heritability ranged from about 70% to >95%
in populations of European descent (Boerwinkle et al. 1992; Kraft et al. 1992). LPA
is also the major locus determining Lp(a) levels in Africans but explained heritabil-
ity is lower in Africans than in populations of European descent (Schmidt et al.
2006; Mooser et al. 1997; Scholz et al. 1999; Enkhmaa et al. 2019). Hence, it
appears that both the KIV-2 VNTR and the LPA locus explain less of the genetic
variability of Lp(a) in Africans suggesting that other loci or environmental factors
have a larger impact on Lp(a) levels in Africans. Several GWAS confirmed that LPA
is the major locus determining Lp(a) levels in Europeans (Mack et al. 2017; Li et al.
2015; Lu et al. 2015; Ober et al. 2009). Minor loci detected by GWAS are the genes
coding for apo E (Mack et al. 2017) and apo H (Hoekstra et al. 2021). ApoH codes
for beta-2 glycoprotein 1 which has been shown to physically interact with apo(a) in
human plasma (Köchl et al. 1997) and which has been implicated in the pathogen-
esis of anti-phospholipid syndrome. A candidate association study implicated TLR2
as a gene modulating Lp(a) levels (Mack et al. 2017). The genomic heritability, i.e.,
the heritability explained by the measured genetic variation in a GWAS was esti-
mated to account for 49.5% of the total variability of Lp(a) levels (Mack et al. 2017).
62 G. Utermann
China
rs3798220
TT TC
55 55
50 50
45 45
40 40
35 35
KIV
KIV
30 30
25 25
20 20
15 15
10 10
50 45 40 35 30 25 20 15 10 5 0 5 10 15 20 25 30 35 40 45 50
Japan
rs3798220
TT TC
50 50
45 45
40 40
35 35
KIV
KIV
30 30
25 25
20 20
15 15
10 10
50 45 40 35 30 25 20 15 10 5 0 5 10 15 20 25 30 35 40 45 50
Frequency Frequency
Fig. 3.6 Graphic representation of the distribution of SNP rs3798220 which is associated with
short KIV-2 alleles in Europeans (Clarke et al. 2009) in Chinese and Japanese were it is associated
with long KIV-2 alleles. (Calculated from the data of Khalifa et al. 2015)
3 Genetics of Lipoprotein(a) 63
The discrepancy between the heritability explained by the VNTR and by the locus
which exists in all studied populations needed an explanation and suggested further
genetic variation at the LPA locus or nearby beyond the KIV-2 VNTR with effects
on Lp(a) levels. Several polymorphisms including SNPs, simple repeats, or restric-
tion site polymorphisms in LPA were shown to explain some of the “missing
64 G. Utermann
the 1000 Genomes project, the variant is present in Europeans and South-East
Asians, occurs with varying frequency in South Americans, and is absent in Africans
(Di Maio et al. 2020).
Surprisingly, the R21X variant was found to be present on the same allele as the
null mutation KIV-8+1G>A (rs41272114). All alleles carrying the R21X mutation
also carried the null mutation KIV-8+1G>A but not vice versa suggesting that
KIV-8+1G>A is the older mutation and R21X occurred on an allele with the splic-
ing defect in KIV-8 generating a “double null” variant (Di Maio et al. 2020).
Only three non-synonymous variants have been demonstrated by functional
studies to have effects on plasma Lp(a) levels. The variant I4399M (rs3798220) in
the protease domain of LPA has been associated with elevated Lp(a) and CHD risk
(Shiffman et al. 2008; Luke et al. 2007) and a benefit from aspirin therapy was
reported (Chasman et al. 2009). An effect of the variant on fibrin clot architecture
and fibrinolysis has been suggested (Shiffman et al. 2008; Luke et al. 2007) but this
was not confirmed in all populations and a dependency from ethnicity was postu-
lated (Rowland et al. 2014). McCormick and coworkers (Morgan et al. 2020) care-
fully characterized two non-synonymous SNPs R990Q and R1771C, which both
result in a null phenotype. They occur in positions of LPA which are homologue to
positions in PLG where mutations result in PLG deficiency. The positions are
important for proper folding of the protein and variants poorly transit to the Golgi
and are not secreted (Morgan et al. 2020).
With the exception of the R21X variant, which had been detected by analysis of
a single family, the KIV-2 VNTR had remained a black box for mutation detection
and was not accessible by standard sequencing nor next-generation sequencing
(NGS). Depending on the number of identical KIV-2 repeats, this region can include
up to 70% of the coding sequence of the LPA gene. This region therefore may sig-
nificantly contribute to functionally relevant variation in the gene. A single study
using a laborious cloning- and a protocol for specific batch-wise PCR-amplification
of KIV-2 repeats from alleles separated by PFGE detected several previously unre-
ported variants in the KIV-2 repeats including a donor splice site mutation desig-
nated K421+1G>A which was associated with a “null allele” (Noureen et al. 2015)
(Fig. 3.3a). This variant occurred in two African and one European alleles. A puta-
tive acceptor splice site variant K422-6T>G associated with short alleles was pres-
ent with high frequency (10% in Khoi San to 40% in Egyptians) only in African
samples. Due to the small total number of only 90 alleles from six ethnic groups, the
study was limited and exact population frequencies and data on effects of variants
on Lp(a) levels in populations were not provided.
Coassin et al. (2017) used the batch amplification of KIV-2 in combination with
NGS. Starting with a discovery set of samples from individuals with discordance
between KIV-2 copy number and Lp(a) levels, they identified a novel frequent
splice site variant G4925A. The variant results in a reduction in splicing activity in
66 G. Utermann
an in vitro assay but not a “null allele.” 4925G>A has a carrier frequency of 21% in
Europeans is associated with short repeats (mainly 19–25 K-IV repeats). It reduced
Lp(a) levels by 31.8 mg/dL and coronary risk significantly (Coassin et al. 2017).
Only recently, a pipeline for ultradeep sequencing of the KIV-2 repeat domain of
LPA (Coassin et al. 2019) and methods to measure KIV2 VNTR length from whole-
exome sequencing data has been developed and allowed for the systematic investi-
gation of variation in this genomic region. The effect of a splice site variant
4733G>A detected in this study on Lp(a) levels and CVD was studied in detail
(Schachtl-Riess et al. 2021) together with the previously reported splice site variant
4925G>A (Coassin et al. 2017). The 4733G>A allele had a high carrier frequency
of 38% and occurred on KIV-2 repeats of all sizes. Overall, it reduced Lp(a) levels
by 13.6 mg/mL. The two splice site variants cooperate in their effect on Lp(a) levels
and CHD risk reduction (Schachtl-Riess et al. 2021).
A further possible level of complexity of the genetic architecture of the Lp(a)
trait is the presence of cis-epistatic effects of variants on Lp(a) levels. A GWAS of
DNA methylation identified a novel association signal associated with elevated
Lp(a) levels in the LPA promoter (Coassin et al. 2020). The effect turned out to be
caused by a non-methylated SNP (rs10455872) which is in LD with short KIV-2
alleles (Coassin et al. 2020). A cis-epistatic effect on Lp(a) levels and coronary risk
was recently demonstrated for variants rs1800769 and rs9458001 which are jointly
associated with elevated Lp(a) levels and with risk for CHD (OR 1.37). Most of this
effect was however explained by rs140570886 (Zeng et al. 2022) known to be asso-
ciated with Lp(a) levels (Mack et al. 2017).
An epistatic effect of two SNPs and the KIV-2 VNTR was also noted in the study
by Mukamel et al. (2021). These authors estimated KIV-2 VNTR copy number from
whole genome sequencing data. Fusing these data by imputation with SNP data,
they were able to define KIV-2 haplotypes and estimate their effects on Lp(a) con-
centrations. They identified 17 protein altering variants. Six of the variants abol-
ished splice sites totally or partially and six were missense variants, all of which
greatly reduced Lp(a) levels. Most of these variants were detected in the KIV-2
VNTR. Variants resulting in increased Lp(a) concentration were found in the 5′UTR
of the LPA gene (Mukamel et al. 2021).
The work of Mukamel et al. (2021) also provided new insights into the genetic
basis underlying the differences in Lp(a) levels between human ethnic groups in
particular between sub-Saharan Africans and Europeans. Analysis by ancestry dem-
onstrated that these differences are largely explained by a significantly lower fre-
quency of Lp(a) decreasing variants and higher frequency of Lp(a) increasing SNPs
in Africans compared to Europeans (Fig. 3.7).
3 Genetics of Lipoprotein(a) 67
Summary
Fig. 3.8 Illustration of the genetic determination of Lp(a) concentrations in plasma by the com-
bined effects of the KIV-2 VNTR and SNPs in the LPA gene. The number of KIV-2 repeats deter-
mines apo(a) isoform size and correlates inversely with the rate of synthesis and with Lp(a)
concentration in plasma. Allele 1 in subject A codes for a long isoform and moderately low Lp(a)
and allele 3 in subject B for a short isoform and high Lp(a). This basic situation is modulated by
SNPs. As examples allele 2 in subject A carries the Lp(a) decreasing SNPs KIV-8 IVS+1G>A
(Ogorelkova et al. 1999) and KIV-2 R21X (Parson et al. 2004) which are in strong LD (Di Maio
et al. 2020) and result in a null allele and allele 4 in subject B which codes for an isoform of inter-
mediate size and carries the variant 4733G>A in KIV-2 which affects splicing and moderately
decreases Lp(a) (Schachtl-Riess et al. 2021). The total plasma Lp(a) concentration in a subject is
the sum of the two allele-associated concentrations (cis-epistatic effects are not considered). Other
loci may have minor effects by unknown mechanisms
3 Genetics of Lipoprotein(a) 69
but rather by different types, allele distributions, and LDs with KIV-2 alleles
between them. Other gene loci beyond LPA, i.e., APOE and APOH have only small
effects on Lp(a) concentrations.
Acknowledgments I thank Anita Neuner for help with the literature, Eugen Preuss for preparing
figures, and Florian Kronenberg for his support and for critically reading the manuscript.
References
Austin MA, Sandholzer C, Selby JV, Newman B, Krauss RM, Utermann G. Lipoprotein(a) in
women twins: heritability and relationship to apolipoprotein(a) phenotypes. Am J Hum Genet.
1992;51:829–40.
Berg K. A new serum type system in man—the Lp system. Acta Pathol Microbiol Scand.
1963;59:369–82.
Berg K, Mohr J. Genetics of the LP system. Acta Genet Stat Med. 1963;13:349–60.
Boerwinkle E, Leffert CC, Lin J, Lackner C, Chiesa G, Hobbs HH. Apolipoprotein(a) gene
accounts for greater than 90% of the variation in plasma lipoprotein(a) concentrations. J Clin
Invest. 1992;90:52–60.
Boomsma DI, Kaptein A, Kempen HJM, Gevers Leuven JA, Princen HMG. Lipoprotein(a): rela-
tion to other risk factors and genetic heritability. Results from a Dutch parent-twin study.
Atherosclerosis. 1993;99:23–33.
Bopp S, Kochl S, Acquati F, Magnaghi P, Petho-Schramm A, Kraft HG, Utermann G, Muller HJ,
Taramelli R. Ten allelic apolipoprotein[a] 5′ flanking fragments exhibit comparable promoter
activities in HepG2 cells. J Lipid Res. 1995;36:1721–8.
Brunner C, Kraft HG, Utermann G, Müller HJ. Cys4057 of apolipoprotein(a) is essential for
lipoprotein(a) assembly. Proc Natl Acad Sci U S A. 1993;90(24):11643–7.
Brunner C, Lobentanz EM, Pethö-Schramm A, Ernst A, Kang C, Dieplinger H, Müller HJ,
Utermann G. The number of identical kringle IV repeats in apolipoprotein(a) affects its pro-
cessing and secretion by HepG2 cells. J Biol Chem. 1996;271:32403–10.
Callow MJ, Rubin EM. Site-specific mutagenesis demonstrates that cysteine 4326 of apo-
lipoprotein B is required for covalent linkage with apolipoprotein(a) in vivo. J Biol Chem.
1995;270:23914–7.
Chasman DI, Shiffman D, Zee RY, Louie JZ, Luke MM, Rowland CM, Catanese JJ, Buring JE,
Devlin JJ, Ridker PM. Polymorphism in the apolipoprotein(a) gene, plasma lipoprotein(a),
cardiovascular disease, and low-dose aspirin therapy. Atherosclerosis. 2009;203:371–6.
Clarke R, Peden JF, Hopewell JC, Kyriakou T, Goel A, Heath SC, Parish S, Barlera S, Franzosi
MG, Rust S, Bennett D, Silveira A, Malarstig A, Green FR, Lathrop M, Gigante B, Leander K,
de Faire U, Seedorf U, Hamsten A, Collins R, Watkins H, Farrall M. Genetic variants associ-
ated with Lp(a) lipoprotein level and coronary disease. N Engl J Med. 2009;361:2518–28.
Coassin S, Erhart G, Weissensteiner H, de Araújo MEG, Lamina C, Schönherr S, Forer L, Haun M,
Losso JL, Köttgen A, Schmidt K, Utermann G, Peters A, Gieger C, Strauch K, Finkenstedt A,
Bale R, Zoller H, Paulweber B, Eckardt KU, Hüttenhofer A, Huber LA, Kronenberg F. A novel
but frequent variant in LPA KIV-2 is associated with a pronounced Lp(a) and cardiovascular
risk reduction. Eur Heart J. 2017;38:1823–31.
Coassin S, Schönherr S, Weissensteiner H, Erhart G, Forer L, Losso JL, Lamina C, Haun M,
Utermann G, Paulweber B, Specht G, Kronenberg F. A comprehensive map of single-base
polymorphisms in the hypervariable LPA kringle IV type 2 copy number variation region. J
Lipid Res. 2019;60(1):186–99.
Coassin S, Hermann-Kleiter N, Haun M, Wahl S, Wilson R, Paulweber B, Kunze S, Meitinger
T, Strauch K, Peters A, Waldenberger M, Kronenberg F, Lamina C. A genome-wide analysis
70 G. Utermann
of DNA methylation identifies a novel association signal for Lp(a) concentrations in the LPA
promoter. PLoS One. 2020;15(4):e0232073.
Crawford DC, Peng Z, Cheng JF, Boffelli D, Ahearn M, Nguyen D, Shaffer T, Yi Q, Livingston RJ,
Rieder MJ, Nickerson DA. LPA and PLG sequence variation and kringle IV-2 copy number in
two populations. Hum Hered. 2008;66:199–209.
Di Maio S, Grüneis R, Streiter G, Lamina C, Maglione M, Schoenherr S, Öfner D, Thorand B,
Peters A, Eckardt K-U, Köttgen A, Kronenberg F, Coassin S. Investigation of a nonsense muta-
tion located in the complex KIV-2 copy number variation region of apolipoprotein(a) in 10,910
individuals. Genome Med. 2020;12(1):74.
Dumitrescu L, Glenn K, Brown-Gentry K, Shephard C, Wong M, Rieder MJ, Smith JD, Nickerson
DA, Crawford DC. Variation in LPA is associated with Lp(a) levels in three populations from
the Third National Health and Nutrition Examination Survey. PLoS One. 2011;6:e16604.
Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and
medical conditions. J Lipid Res. 2016;57(7):1111–25.
Enkhmaa B, Anuurad E, Zhang W, Kim K, Berglund L. Heritability of apolipoprotein (a) traits
in two-generational African-American and Caucasian families. J Lipid Res. 2019;60:1603–9.
Erdel M, Hubalek M, Lingenhel A, Kofler K, Duba HC, Utermann G. Counting the repetitive
kringle-IV repeats in the gene encoding human apolipoprotein(a) by fibre-FISH. Nat Genet.
1999;21:357–8.
Erhart G, Lamina C, Lehtimäki T, Marques-Vidal P, Kähönen M, Vollenweider P, Raitakari OT,
Waeber G, Thorand B, Strauch K, Gieger C, Meitinger T, Peters A, Kronenberg F, Coassin
S. Genetic factors explain a major fraction of the 50% lower lipoprotein[a] concentrations in
Finns. Arterioscler Thromb Vasc Biol. 2018;38:1230–41.
Ernst A, Helmhold M, Brunner C, Pethö-Schramm A, Armstrong VW, Müller H-J. Identification
of two functionally distinct lysine-binding sites in kringle 37 and in kringles 32-36 of human
apolipoprotein(a). J Biol Chem. 1995;270:6227–34.
Gabel BR, Koschinsky ML. Sequences within apolipoprotein(a) kringle IV types 6-8 bind directly
to low-density lipoprotein and mediate noncovalent association of apolipoprotein(a) with apo-
lipoprotein B-100. Biochemistry. 1998;37:7892–8.
Gaw A, Boerwinkle E, Cohen JC, Hobbs HH. Comparative analysis of the apo(a) gene, apo(a) gly-
coprotein, and plasma concentrations of Lp(a) in three ethnic groups: evidence for no common
“Null” allele at the apo(a) locus. J Clin Invest. 1994;93:2526–34.
Helmhold M, Bigge J, Muche R, Mainoo J, Thiery J, Seidel D, Armstrong VW. Contribution of the
apo[a] phenotype to plasma Lp[a] concentrations shows considerable ethnic variation. J Lipid
Res. 1991;32:1919–28.
Hoekstra M, Chen HY, Rong J, Dufresne L, Yao J, Guo X, Tsai MY, Tsimikas S, Post WS, Vasan
RS, Rotter JI, Larson MG, Thanassoulis G, Engert JC. Genome-wide association study high-
lights APOH as a novel locus for lipoprotein(a) levels-brief report. Arterioscler Thromb Vasc
Biol. 2021;41(1):458–64.
Kamstrup PR, Tybjaerg-Hansen A, Steffensen R, Nordestgaard BG. Genetically elevated
lipoprotein(a) and increased risk of myocardial infarction. JAMA. 2009;301:2331–9.
Khalifa M, Noureen A, Ertelthalner K, Bandegi AR, Delport R, Firdaus WJ, Geethanjali FS,
Luthra K, Makemaharn O, Pang RW, Salem AH, Sasaki J, Schiefenhoevel W, Lingenhel
A, Kronenberg F, Utermann G, Schmidt K. Lack of association of rs3798220 with small
apolipoprotein(a) isoforms and high lipoprotein(a) levels in East and Southeast Asians.
Atherosclerosis. 2015;242:521–8.
Köchl S, Fresser F, Lobentanz E, Baier G, Utermann G. Novel interaction of apolipoprotein(a)
with b-2 glycoprotein I mediated by the kringle IV domain. Blood. 1997;90:1482–9.
Koschinsky ML, Côté GP, Gabel B, Van der Hoek YY. Identification of the cysteine residue in
apolipoprotein(a) that mediates extracellular coupling with apolipoprotein B-100. J Biol Chem.
1993;268:19819–25.
Kostner KM, Kostner GM. Factors affecting plasma lipoprotein(a) levels: role of hormones and
other nongenetic factors. Semin Vasc Med. 2004;4:211–4.
3 Genetics of Lipoprotein(a) 71
Kraft HG, Köchl S, Menzel HJ, Sandholzer C, Utermann G. The apolipoprotein(a) gene: a tran-
scribed hypervariable locus controlling plasma lipoprotein(a) concentration. Hum Genet.
1992;90:220–30.
Kraft HG, Lingenhel A, Pang RWC, Delport R, Trommsdorff M, Vermaak H, Janus ED, Utermann
G. Frequency distributions of apolipoprotein(a) kringle IV repeat alleles and their effects on
lipoprotein(a) levels in Caucasian, Asian, and African populations: The distribution of null
alleles is non-random. Eur J Hum Genet. 1996;4:74–87.
Krempler F, Kostner GM, Bolzano K, Sandhofer F. Turnover of lipoprotein(a) in man. J Clin
Invest. 1980;65:1483–90.
Kyriakou T, Seedorf U, Goel A, Hopewell JC, Clarke R, Watkins H, Farrall M. A common LPA null
allele associates with lower lipoprotein(a) levels and coronary artery disease risk. Arterioscler
Thromb Vasc Biol. 2014;34:2095–9.
Lackner C, Boerwinkle E, Leffert CC, Rahmig T, Hobbs HH. Molecular basis of apolipoprotein
(a) isoform size heterogeneity as revealed by pulsed-field gel electrophoresis. J Clin Invest.
1991;87:2153–61.
Lackner C, Cohen JC, Hobbs HH. Molecular definition of the extreme size polymorphism in
apolipoprotein(a). Hum Mol Genet. 1993;2:933–40.
Li J, Lange LA, Sabourin J, Duan Q, Valdar W, Willis MS, Li Y, Wilson JG, Lange EM. Genome-
and exome-wide association study of serum lipoprotein (a) in the Jackson Heart Study. J Hum
Genet. 2015;60:755–61.
Lim ET, Wurtz P, Havulinna AS, Palta P, Tukiainen T, Rehnstrom K, Esko T, Magi R, Inouye
M, Lappalainen T, Chan Y, Salem RM, Lek M, Flannick J, Sim X, Manning A, Ladenvall C,
Bumpstead S, Hamalainen E, Aalto K, Maksimow M, Salmi M, Blankenberg S, Ardissino D,
Shah S, Horne B, McPherson R, Hovingh GK, Reilly MP, Watkins H, Goel A, Farrall M, Girelli
D, Reiner AP, Stitziel NO, Kathiresan S, Gabriel S, Barrett JC, Lehtimaki T, Laakso M, Groop
L, Kaprio J, Perola M, McCarthy MI, Boehnke M, Altshuler DM, Lindgren CM, Hirschhorn
JN, Metspalu A, Freimer NB, Zeller T, Jalkanen S, Koskinen S, Raitakari O, Durbin R,
MacArthur DG, Salomaa V, Ripatti S, Daly MJ, Palotie A. Distribution and medical impact of
loss-of-function variants in the finnish founder population. PLoS Genet. 2014;10:e1004494.
Lobentanz E-M, Krasznai K, Gruber A, Brunner C, Müller H-J, Sattler J, Kraft H-G, Utermann G,
Dieplinger H. Intracellular metabolism of human apolipoprotein(a) in stably transfected Hep
G2 cells. Biochemistry. 1998;37:5417–25.
Lu W, Cheng YC, Chen K, Wang H, Gerhard GS, Still CD, Chu X, Yang R, Parihar A, O’Connell
JR, Pollin TI, Angles-Cano E, Quon MJ, Mitchell BD, Shuldiner AR, Fu M. Evidence for sev-
eral independent genetic variants affecting lipoprotein (a) cholesterol levels. Hum Mol Genet.
2015;24:2390–400.
Luke MM, Kane JP, Liu DM, Rowland CM, Shiffman D, Cassano J, Catanese JJ, Pullinger CR,
Leong DU, Arellano AR, Tong CH, Movsesyan I, Naya-Vigne J, Noordhof C, Feric NT,
Malloy MJ, Topol EJ, Koschinsky ML, Devlin JJ, Ellis SG. A polymorphism in the protease-
like domain of apolipoprotein(a) is associated with severe coronary artery disease. Arterioscler
Thromb Vasc Biol. 2007;27:2030–6.
Mack S, Coassin S, Rueedi R, Yousri NA, Seppala I, Gieger C, Schoenherr S, Forer L, Erhart G,
Marques-Vidal P, Ried JS, Waeber G, Bergmann S, Daehnhardt D, Stoeckl A, Raitakari OT,
Khahonen M, Peters A, Meitinger T, Strauch K, Kedenko L, Paulweber B, Lehtimaki T, Hunt
SC, Vollenweider P, Lamina C, Kronenberg F, Grp K-S. A genome-wide association meta-
analysis on lipoprotein (a) concentrations adjusted for apolipoprotein (a) isoforms. J Lipid Res.
2017;58(9):1834–44.
Marcovina SM, Albers JJ, Wijsman E, Zhang ZH, Chapman NH, Kennedy H. Differences in
Lp[a] concentrations and apo[a] polymorphs between black and white Americans. J Lipid Res.
1996;37:2569–85.
McCormick SPA, No JK, Taylor S, Flynn LM, Hammer RE, Young SG. Mutagenesis of the human
apolipoprotein B gene in a yeast artificial chromosome reveals the site of attachment for
apolipoprotein(a). Proc Natl Acad Sci U S A. 1995;92:10147–51.
72 G. Utermann
McLean JW, Tomlinson JE, Kuang W-J, Eaton DL, Chen EY, Fless GM, Scanu AM, Lawn
RM. cDNA sequence of human apolipoprotein(a) is homologous to plasminogen. Nature.
1987;330:132–7.
Mooser V, Scheer D, Marcovina SM, Wang JP, Guerra R, Cohen J, Hobbs HH. The Apo(a) gene
is the major determinant of variation in plasma Lp(a) levels in African Americans. Am J Hum
Genet. 1997;61:402–17.
Morgan BM, Brown AN, Deo N, Harrop TWR, Taiaroa G, Mace PD, Wilbanks SM, Merriman TR,
Williams MJA, McCormick SPA. Nonsynonymous SNPs in LPA homologous to plasminogen
deficiency mutants represent novel null apo(a) alleles. J Lipid Res. 2020;61(3):432–44.
Mukamel RE, Handsaker RE, Sherman MA, Barton AR, Zheng Y, McCarroll SA, Loh
PR. Protein-coding repeat polymorphisms strongly shape diverse human phenotypes. Science.
2021;373(6562):1499–505.
Noureen A, Fresser F, Utermann G, Schmidt K. Sequence variation within the KIV-2 copy number
polymorphism of the human LPA gene in African, Asian, and European populations. PLoS
One. 2015;10:e0121582.
Ober C, Nord AS, Thompson EE, Pan L, Tan Z, Cusanovich D, Sun Y, Nicolae R, Edelstein C,
Schneider DH, Billstrand C, Pfaffinger D, Phillips N, Anderson RL, Philips B, Rajagopalan
R, Hatsukami TS, Rieder MJ, Heagerty PJ, Nickerson DA, Abney M, Marcovina S, Jarvik GP,
Scanu AM, Nicolae DL. Genome-wide association study of plasma lipoprotein(a) levels identi-
fies multiple genes on chromosome 6q. J Lipid Res. 2009;50:798–806.
Ogorelkova M, Gruber A, Utermann G. Molecular basis of congenital Lp(a) deficiency: a frequent
apo(a) ‘null’ mutation in Caucasians. Hum Mol Genet. 1999;8:2087–96.
Ogorelkova M, Kraft HG, Ehnholm C, Utermann G. Single nucleotide polymorphisms in exons of
the apo(a) kringles IV types 6 to 10 domain affect Lp(a) plasma concentrations and have differ-
ent patterns in Africans and Caucasians. Hum Mol Genet. 2001;10:815–24.
Parson W, Kraft HG, Niederstatter H, Lingenhel AW, Kochl S, Fresser F, Utermann G. A common
nonsense mutation in the repetitive Kringle IV-2 domain of human apolipoprotein(a) results in
a truncated protein and low plasma Lp(a). Hum Mutat. 2004;24:474–80.
Prins J, Leus FR, Van der Hoek YY, Kastelein JJP, Bouma BN, Van Rijn HJM. The identifica-
tion and significance of a Thr–>Pro polymorphism in Kringle IV type 8 of apolipoprotein(a).
Thromb Haemost. 1997;77:949–54.
Prins J, Leus FR, Bouma BN, van Rijn HJ. The identification of polymorphisms in the coding
region of the apolipoprotein (a) gene-association with earlier identified polymorphic sites and
influence on the lipoprotein (a) concentration. Thromb Haemost. 1999;82:1709–17.
Rader DJ, Cain W, Zech LA, Usher D, Brewer HB Jr. Variation in lipoprotein(a) concentra-
tions among individuals with the same apolipoprotein(a) isoform is determined by the rate of
lipoprotein(a) production. J Clin Invest. 1993;91:443–7.
Rowland CM, Pullinger CR, Luke MM, Shiffman D, Green L, Movsesyan I, Devlin JJ, Malloy
MJ, Kane JP, Undas A. Lipoprotein (a), LPA Ile4399Met, and fibrin clot properties. Thromb
Res. 2014;133:863–7.
Sandholzer C, Hallman DM, Saha N, Sigurdsson G, Lackner C, Császár A, Boerwinkle E,
Utermann G. Effects of the apolipoprotein(a) size polymorphism on the lipoprotein(a) concen-
tration in 7 ethnic groups. Hum Genet. 1991;86:607–14.
Schachtl-Riess JF, Kheirkhah A, Grüneis R, Di Maio S, Schoenherr S, Streiter G, Losso JL,
Paulweber B, Eckardt K-U, Köttgen A, Lamina C, Kronenberg F, Coassin S. Frequent LPA
KIV-2 variants lower lipoprotein(a) concentrations and protect against coronary artery disease.
J Am Coll Cardiol. 2021;78(5):437–49.
Schmidt K, Kraft HG, Parson W, Utermann G. Genetics of the Lp(a)/apo(a) system in an autoch-
thonous Black African population from the Gabon. Eur J Hum Genet. 2006;14:190–201.
Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of
lipoprotein(a). J Lipid Res. 2016;57:1339–59.
Scholz M, Kraft HG, Lingenhel A, Delport R, Vorster EH, Bickeböller H, Utermann G. Genetic
control of lipoprotein(a) concentrations is different in Africans and Caucasians. Eur J Hum
Genet. 1999;7:169–78.
3 Genetics of Lipoprotein(a) 73
Shiffman D, O’Meara ES, Bare LA, Rowland CM, Louie JZ, Arellano AR, Lumley T, Rice K,
Iakoubova O, Luke MM, Young BA, Malloy MJ, Kane JP, Ellis SG, Tracy RP, Devlin JJ, Psaty
BM. Association of gene variants with incident myocardial infarction in the Cardiovascular
Health Study. Arterioscler Thromb Vasc Biol. 2008;28:173–9.
Tomlinson JE, McLean JW, Lawn RM. Rhesus monkey apolipoprotein(a). Sequence, evolution,
and sites of synthesis. J Biol Chem. 1989;264:5957–65.
Trommsdorff M, Köchl S, Lingenhel A, Kronenberg F, Delport R, Vermaak H, Lemming L,
Klausen IC, Faergeman O, Utermann G, Kraft H-G. A pentanucleotide repeat polymorphism
in the 5′ control region of the apolipoprotein(a) gene is associated with lipoprotein(a) plasma
concentrations in Caucasians. J Clin Invest. 1995;96:150–7.
Utermann G. The mysteries of lipoprotein(a). Science. 1989;246:904–10.
Utermann G, Menzel HJ, Kraft HG, Duba HC, Kemmler HG, Seitz C. Lp(a) glycoprotein phe-
notypes: inheritance and relation to Lp(a)-lipoprotein concentrations in plasma. J Clin Invest.
1987;80:458–65.
Waldeyer C, Makarova N, Zeller T, Schnabel RB, Brunner FJ, Jorgensen T, Linneberg A, Niiranen
T, Salomaa V, Jousilahti P, Yarnell J, Ferrario MM, Veronesi G, Brambilla P, Signorini SG,
Iacoviello L, Costanzo S, Giampaoli S, Palmieri L, Meisinger C, Thorand B, Kee F, Koenig W,
Ojeda F, Kontto J, Landmesser U, Kuulasmaa K, Blankenberg S. Lipoprotein(a) and the risk of
cardiovascular disease in the European population: results from the BiomarCaRE consortium.
Eur Heart J. 2017;38:2490–8.
Wendt GG. International Lp-workshop. Humangenetik. 1967;3(3):269–72.
White AL, Rainwater DL, Lanford RE. Intracellular maturation of apolipoprotein[a] and assembly
of lipoprotein[a] in primary baboon hepatocytes. J Lipid Res. 1993;34:509–17.
White AL, Rainwater DL, Hixson JE, Estlack LE, Lanford RE. Intracellular processing of Apo(A)
in primary baboon hepatocytes. Chem Phys Lipids. 1994;67-8:123–33.
Zeng L, Moser S, Mirza-Schreiber N, Lamina C, Coassin S, Nelson CP, Annilo T, Franzen O,
Kleber ME, Mack S, Andlauer TFM, Jiang B, Stiller B, Li L, Willenborg C, Munz M, Kessler
T, Kastrati A, Laugwitz KL, Erdmann J, Moebus S, Nothen MM, Peters A, Strauch K, Muller-
Nurasyid M, Gieger C, Meitinger T, Steinhagen-Thiessen E, Marz W, Metspalu A, Bjorkegren
JLM, Samani NJ, Kronenberg F, Muller-Myhsok B, Schunkert H. Cis-epistasis at the LPA
locus and risk of cardiovascular diseases. Cardiovasc Res. 2022;118(4):1088–102.
Chapter 4
Lp(a) Metabolism
Introduction
J. S. Millar
Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
Institute for Diabetes Obesity and Metabolism, Perelman School of Medicine, University of
Pennsylvania, Philadelphia, PA, USA
e-mail: [email protected]
D. J. Rader (*)
Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
Institute for Diabetes Obesity and Metabolism, Perelman School of Medicine, University of
Pennsylvania, Philadelphia, PA, USA
Department of Genetics, Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA, USA
Institute for Translational Medicine and Therapeutics, Perelman School of Medicine,
University of Pennsylvania, Philadelphia, PA, USA
Smilow Center for Translational Research, University of Pennsylvania,
Philadelphia, PA, USA
e-mail: [email protected]
et al. 2016). The apo(a) peptide consists of a series of domains that are highly
homologous to several domains of plasminogen (McLean et al. 1987). The
N-terminal portion of apo(a) consists of a variable number of repeating domains
that are homologous to kringle IV of plasminogen. This is followed by a non-
variable portion that consists of two additional domains that are homologous to the
kringle V and the serine protease domains of plasminogen. The number of kringle
IV repeats in the variable portion of apo(a) range from 2 to more than 40 resulting
in over 30 different isoforms ranging in size from approximately 300 to 800 kDa
(Kronenberg and Utermann 2013).
While the vast majority of apo(a) in blood is found covalently bound to apoB100
on Lp(a) particles that overlap the LDL–HDL density range (Rainwater et al. 1995),
it has been noted that a proportion of apo(a) can be found non-covalently associated
with triglyceride-rich lipoproteins (Bersot et al. 1986). It is unclear if these com-
plexes develop into mature Lp(a) particles. Such a model suggests that mature Lp(a)
particles are formed extracellularly in plasma consistent with findings from some
in vitro studies examining apo(a) secretion from hepatocytes (Koschinsky et al.
1991; White et al. 1993). However, there is a report showing evidence for apo(a)
binding to apoB-containing lipoproteins intracellularly (Bonen et al. 1997) leading
to an alternative model whereby Lp(a) can be formed intracellularly and secreted as
an intact particle.
In addition to the apoB100 and apo(a) components, proteomic analysis of highly
purified Lp(a) has identified 35 additional proteins that are associated with Lp(a)
(von Zychlinski et al. 2011). In addition to proteins known to be involved in lipid
metabolism (such as apoE and apoC-III), other proteins found associated with Lp(a)
include those involved in wound healing (coagulation [fibrinogen], complement
activation [complement C3 and C4A]), and inflammatory response (platelet activat-
ing factor acetyl hydrolase). However, biological significance of these additional
proteins associated with Lp(a) is unknown.
While the Lp(a) resembles LDL containing a covalently linked apo(a) peptide,
analysis of the lipid portion of Lp(a) has revealed that unlike LDL, Lp(a) is enriched
in oxidized phospholipids (Tsimikas and Witztum 2008), both in the lipoprotein por-
tion of the particle as well as being bound to the apo(a) peptide (Leibundgut et al.
2013). It has been shown that oxidized phospholipids are transferred from LDL to
Lp(a) in vitro and proposed that Lp(a) is the preferential carrier of oxidized phospho-
lipids in plasma (Bergmark et al. 2008). The presence of large amounts of oxidized
lipids on Lp(a) could contribute to the atherogenicity of this lipoprotein. A genome-
wide association study designed to identify risk factors for aortic stenosis identified
variants at the LPA locus encoding apo(a) as the most significantly associated
genomic locus. This has led to the hypothesis that oxidized phospholipids on Lp(a)
contribute to the progression of aortic calcification and stenosis (Yeang et al. 2016).
The apo(a) peptide is encoded by the LPA gene located on chromosome 6q27
(Scanu et al. 1991). The gene is primarily expressed in liver with minor expression
in kidney. The length of each LPA allele is variable due to there being variability in
the copy number of domains that encode kringle IV type 2 (KIV-2) (Lanktree et al.
2010). The number of KIV-2 domain repeats in LPA has been estimated to range
4 Lp(a) Metabolism 77
from 2 to >40 copies (Kronenberg and Utermann 2013). Null alleles of LPA that
encode a truncated apo(a) protein that is unable to bind covalently to apoB have
been reported (Ogorelkova et al. 1999). In addition, LPA alleles with a large number
of the KIV-2 domain repeats are unable to be secreted, presumably due to being
unstable intracellularly, and are therefore also considered null alleles (White et al.
1994). In vitro studies suggest that the number of KIV-2 domain repeats are inversely
associated with circulating Lp(a) levels due to the more efficient intracellular pro-
cessing and secretion of smaller apo(a) isoforms (White et al. 1994).
A number of genome-wide association studies have been conducted that provide
insight into the genes that regulate Lp(a) levels. Quantitively, genetic variation at the
LPA locus itself is, by far, the most important factor influencing Lp(a) levels. Clarke
et al. identified two SNPs (rs10455872 and rs3798220) at the LPA locus, which
were associated with reduced KIV-2 copy number, small Lp(a) size, and increased
Lp(a) levels (Clarke et al. 2009). Mack et al. identified 30 single nucleotide poly-
morphisms (SNPs) in the LPA gene, which either increased or decreased Lp(a) lev-
els (Mack et al. 2017). They confirmed the two SNPs identified by Clarke et al. as
well as other SNPs that were associated with the number of KIV-2 domain repeats
that are inversely associated with Lp(a) levels. There was also an association with
Lp(a) levels and the apolipoprotein E (APOE) gene, specifically with the APOE2
allele being associated with decreased Lp(a) levels. Li et al. also found an associa-
tion between a SNP in the APOE gene with Lp(a) levels (Li et al. 2015). Since apoE
is an exchangeable apolipoprotein that binds to multiple lipoprotein receptors, it is
possible that apoE on Lp(a) contributes to the clearance of Lp(a) from the circula-
tion. There is also an association between the Toll-like receptor 2 (TLR2) gene and
Lp(a) levels (Mack et al. 2017) leading to speculation that TLR2 may participate in
Lp(a) clearance from plasma. TLR2 is known to bind lipopolysaccharide but, thus
far, there have been no reports regarding the interaction between Lp(a) and TLR2.
Lp(a) Metabolism
The metabolism of Lp(a) is complex and has been the subject of intensive investiga-
tion. Here we review the major aspects of what has been reported regarding Lp(a)
metabolism.
The first study to examine the metabolism of autologous Lp(a) in humans was con-
ducted by Krempler et al. (1978) to characterize the clearance and metabolic fate of
Lp(a) in plasma. Lp(a) was isolated using a combination of ultracentrifugation, and
78 J. S. Millar and D. J. Rader
Intracellular assembly
A and secretion of -S-S- -S-S-
mature Lp(a)
Intracellular assembly
of disulfide-linked apo(a)
B to VLDL, secretion, and -S-S- VLDL -S-S- VLDL -S-S-
extracellular conversion
to mature Lp(a)
Fig. 4.1 The biosynthesis of mature Lp(a) may occur through diverse pathways. Possible path-
ways (not mutually exclusive) include: (a) intracellular assembly and secretion of the mature Lp(a)
particle; (b) intracellular assembly of disulfide-linked apo(a) to VLDL, secretion, and extracellular
conversion to mature Lp(a); (c) secretion of free apo(a) and extracellular association with VLDL/
IDL/LDL and conversion to mature Lp(a)
allowed for formation of Lp(a) in both the liver (pre-formed) or in plasma. They
calculated that about 50% of Lp(a) was formed in plasma from LDL with the
remainder being secreted into plasma directly from liver as a preformed Lp(a) par-
ticle. They also compared the clearance rates of the apo(a) and apoB100 compo-
nents of Lp(a) and found that they were cleared from plasma at similar rates. The
potential pathways by which Lp(a) is formed from apo(a) and apoB-containing
lipoproteins are shown in Fig. 4.1.
Krempler et al. (1980) addressed the question of whether circulating Lp(a) levels
were controlled primarily by production or by clearance. They measured Lp(a) pro-
duction and clearance in subjects with a wide range of Lp(a) levels and found that
there was a significant correlation between circulating Lp(a) levels and the Lp(a)
production rate. There was no relationship between Lp(a) levels and the Lp(a) frac-
tional clearance rate. These results indicated that Lp(a) levels are primarily con-
trolled by production.
While circulating Lp(a) levels were known to be associated with apo(a) isoform
size, it had been noted that there was a considerable variation in Lp(a) levels in
subjects with the same apo(a) isoform size (Utermann et al. 1987). However, the
mechanism responsible for these differences was unknown. Rader et al. (1993)
examined the metabolism of Lp(a) in humans to determine the mechanism respon-
sible for the differences in Lp(a) levels seen in individuals with the same sized
80 J. S. Millar and D. J. Rader
apo(a) isoform. Lp(a) was isolated from donors with a single apo(a) isoform size by
sequential ultracentrifugation followed by density gradient ultracentrifugation.
Isolated Lp(a) was then radiolabeled and injected into study subjects with a range of
Lp(a) levels but having a single apo(a) isoform size. The results showed that there
was no difference in the clearance of Lp(a) from plasma in subjects with the same
apo(a) isoform size. However, there were substantial differences in the production
rate of Lp(a) among individuals with the same sized apo(a) isoform, perhaps due to
variants in the LPA gene that are independent of isoform size but which affect apo(a)
production (White et al. 1994). They concluded that Lp(a) production is the most
important determinant of the plasma level of Lp(a) independent of apo(a) iso-
form size.
It is well established that plasma Lp(a) levels are inversely correlated with apo(a)
isoform size, but the mechanism behind this correlation was unknown. Rader et al.
(1994) examined the physiology responsible for differences in Lp(a) levels based on
apo(a) isoform size. The goal of the study was to determine if there were differences
in the fractional clearance rates of Lp(a) particles containing different sized apo(a)
isoforms. Healthy normolipidemic subjects were injected with either autologous or
homologous radiolabeled Lp(a) isolated from plasma by sequential followed by
density gradient ultracentrifugation. Subjects with different apo(a) isoform pheno-
types were injected with radiolabeled Lp(a) preparations containing different sized
apo(a) isoforms. They found that Lp(a) containing different sized apo(a) isoforms
had similar clearance rates consistent with what they had observed in their previous
study (Rader et al. 1993). However, there were substantial differences in the produc-
tion rate of Lp(a) containing different sized apo(a) isoforms. The production rate of
Lp(a) containing small apo(a) isoforms was considerably higher than that for Lp(a)
containing large apo(a) isoforms. They concluded that the inverse association of
plasma Lp(a) concentrations with apo(a) isoform size is not due to differences in the
fractional clearance rates of Lp(a) containing different sized isoforms but rather the
production rate. These studies provided in vivo evidence that supported earlier
in vitro observations that smaller apo(a) isoforms are more readily secreted from
hepatocytes, likely due to more efficient intracellular processing (White et al. 1994).
The relationship between apo(a) isoform size and apo(a) production is shown in
Fig. 4.2.
The receptor(s) responsible for Lp(a) clearance from plasma are currently unknown.
Since Lp(a) contains apoB100, the ligand for the LDL receptor, the potential role of
the LDL receptor in mediating Lp(a) clearance has been of great interest. One
approach to address this question has been the use of genetics. In studies with
patients with familial hypercholesterolemia (FH), Kraft et al. found a gene dosage
effect of the LDL receptor gene (LDLR) on Lp(a) levels when controlling for LPA
alleles (Kraft et al. 2000). A study conducted using the UK Biobank found that
4 Lp(a) Metabolism 81
* Minority Secreted
Large K-IV2-1 K-IV2-20 * Majority Unstable
Fig. 4.2 The LPA gene encodes the apo(a) protein which is of highly variable length due to varia-
tion in the number of kringle IV2 domain repeats. The strong inverse association of apo(a) protein
size with plasma Lp(a) level is due to the effect of apo(a) protein size on the rate of production, not
catabolism, of Lp(a). While apo(a) peptide translation appears to occur normally, longer intracel-
lular peptides are unstable and are targeted for pre-secretory degradation. While not shown here, in
vivo evidence also indicates that variation in plasma Lp(a) levels among individuals with the same
size isoform(s) is also due to differences in Lp(a) production, not catabolism
patients carrying variants in the LDL receptor that cause FH had higher Lp(a) levels
than unaffected subjects (Trinder et al. 2020). However, it was determined that the
population of FH patients studied was enriched in the rs10455872 SNP in LPA
which is associated with higher Lp(a) levels; when the presence of the SNP was
controlled for, it was found that Lp(a) levels were similar between patients with and
without FH. Lp(a) levels are within the normal range in patients with familial defec-
tive apoB, a disorder where there is an amino acid substitution in LDL receptor
binding domain of apoB (Innerarity et al. 1987). It is also of interest to note that
genome-wide association studies have identified variants at the LDLR locus as being
strongly associated with LDL cholesterol levels (Kathiresan et al. 2008); variants at
the LDLR locus have not been found to be associated with Lp(a) levels (Clarke et al.
2009). Thus, the genetic data do not strongly support a role for the LDL receptor in
directly mediating the catabolism of Lp(a).
There have also been experimental efforts to determine the role of the LDL
receptor in Lp(a) clearance. Lp(a) can bind to the LDL receptor in vitro although the
affinity of Lp(a) for the LDL receptor has been characterized as “weak” (Reblin
et al. 1997). Knight et al. (1991) studied the role of the LDL receptor in the in vivo
clearance of Lp(a) from plasma in hyperlipidemic patients with and without hetero-
zygous FH. They radiolabeled autologous Lp(a) and LDL isolated by density gradi-
ent ultracentrifugation and examined the clearance of each from plasma. They
found that there was no difference in the clearance rate of Lp(a) from plasma
82 J. S. Millar and D. J. Rader
between individuals with and without heterozygous FH despite there being signifi-
cant differences in the clearance of autologous LDL. They also conducted in vitro
studies that examined the ability of Lp(a) to compete with LDL binding to the LDL
receptor. They found that Lp(a) was unable to compete with LDL for binding to the
LDL receptor. They did find that after injection of radiolabeled Lp(a) that there was
appearance of approximately 25% of the Lp(a) tracer in the LDL fraction which
they interpreted as resulting from loss of apo(a) from the Lp(a) particle. They
hypothesized that the resulting LDL could be cleared by LDL receptors. These
authors also found no differences in the clearance of Lp(a) containing apo(a) iso-
forms of different sizes and that Lp(a) levels were correlated with the Lp(a) produc-
tion rate.
Rader et al. studied the catabolism of Lp(a) in five patients with homozygous FH
who had little to no LDL receptor function (Rader et al. 1995). Purified radioiodin-
ated Lp(a) and LDL were simultaneously injected into homozygous FH patients and
control subjects, and the catabolism was followed over time. While the catabolism
of LDL was markedly delayed as expected, the catabolism of Lp(a) was not slower
in homozygous FH patients than in control subjects. This study provided powerful
evidence that the absence of a functional LDL receptor does not result in delayed
catabolism of Lp(a) and suggested that the LDL receptor is not a physiologically
important route of Lp(a) catabolism in humans.
In mice with marked overexpression of the LDL receptor, there was an increased
uptake of Lp(a) levels resulting in decreased levels in plasma (Hofmann et al. 1990;
Romagnuolo et al. 2017). Cain et al. addressed the question using mice deficient in
the LDL receptor (Cain et al. 2005). When the catabolism of radiolabeled Lp(a) was
studied in mice deficient in the LDL receptor compared to wild-type mice, there
was no observed difference in Lp(a) turnover. This was consistent with the human
studies and provided additional evidence that the LDL receptor is not a major con-
tributor to Lp(a) clearance.
As another type of evidence, statins reduce LDL-C levels by causing the upregu-
lation of the LDLR in hepatocytes and increased clearance of LDL and its precur-
sors. However, as reviewed below, statins do not decrease Lp(a) levels and, if
anything, cause Lp(a) levels to increase slightly. This also argues against the LDL
receptor as being an important mediator of Lp(a) clearance. Overall, the data do not
support an important role for the LDL receptor in mediating clearance of Lp(a)
from blood.
The role of other receptors in Lp(a) clearance has been investigated and current
information suggests that Lp(a) can be cleared from plasma through multiple recep-
tors (Fig. 4.3). Available data suggest that apo(a) is the ligand responsible for
receptor-mediated binding of Lp(a). For example, while excess LDL was shown to
have minimal impact on Lp(a) clearance in mice, excess apo(a) significantly slowed
4 Lp(a) Metabolism 83
LDLR
LRP-1
Apo(a) ligand
LRP-8
VLDLR
SR-BI
Megalin/
Glycoprotein 330
TRL-2
Other
Receptors?
Fig. 4.3 Lp(a) clearance is not a major determinant of plasma Lp(a) levels and mostly occurs by
the liver. The mechanisms of hepatic Lp(a) clearance remain unknown and likely involve multiple
pathways. This figure lists many of the cell surface receptors that have been proposed as receptors
for Lp(a)
Lp(a) clearance from plasma (Cain et al. 2005). The scavenger receptor B-I, best
known for its role in regulating HDL cholesterol uptake, has been reported to bind
Lp(a) (Yang et al. 2013). The megalin/glycoprotein 330 receptor, a member of the
LDL receptor family, has been shown to bind and take up Lp(a) into cells in vitro
(Niemeier et al. 1999). The LDL receptor-related protein-1 has been shown to bind
Lp(a) weakly in vitro (Reblin et al. 1997) but has been reported to have no effect on
Lp(a) clearance in vivo in animal models (Romagnuolo et al. 2017). The plasmino-
gen receptor PlgRKT has been shown to mediate Lp(a) uptake by HepG2 cells
(Sharma et al. 2017). It is interesting to note that following uptake by PlgRKT, the
apo(a) component of Lp(a) was trafficked to recycling endosomes and subsequently
re-secreted into the cellular media. This would help explain the results of some
in vivo human studies that had results showing a slower clearance of apo(a) as com-
pared to apoB100 on Lp(a) which could be explained by apo(a) recycling (Jenner
et al. 2005; Diffenderfer et al. 2016). Other receptors that have been shown to have
no effect on Lp(a) clearance include the VLDL receptor, LDL receptor-related pro-
tein-8 (Romagnuolo et al. 2017), the asialoglycoprotein receptor (Cain et al. 2005),
and sortilin (Gemin et al. 2018). Variants in APOE have been identified as being
84 J. S. Millar and D. J. Rader
determinants of Lp(a) levels (Clarke et al. 2009; Mack et al. 2017; Li et al. 2015).
In addition, apoE in plasma has been shown to have a modest impact on Lp(a) clear-
ance (Li et al. 2015; Cain et al. 2005). It is of interest to note a case report of a
patient with apoE deficiency who had an Lp(a) level approximately three-fold
higher than the upper limit of normal (Mak et al. 2014) which might be expected if
apoE is involved in clearance of Lp(a) or its precursors.
Statins
PCSK9 Inhibitors
PCSK9 inhibitors block the effect of PCSK9 in mediating LDL receptor degrada-
tion, thus leading to increased LDL receptor protein and increased clearance of
LDL. They reduce LDL-C levels by about 60%. Treatment with PCSK9 inhibitors
has been shown to modestly lower Lp(a) levels by ~20% (Ajufo and Rader 2016).
This has been shown to be due to enhanced clearance of Lp(a) from plasma (Watts
et al. 2020; Reyes-Soffer et al. 2017). While increased LDL receptor numbers
could play a role, it is also possible that PCSK9 influences other factors that affect
Lp(a) clearance. Patients heterozygous for PCSK9 gain-of-function mutations have
been reported to have Lp(a) levels that were two-fold higher than control subjects
(Tada et al. 2016), while heterozygous carriers of PCSK9 loss-of-function muta-
tions have been reported to have Lp(a) levels that are 22% lower than those found
in control subjects (Mefford et al. 2019). PCSK9 expression has been shown to
slow the uptake of Lp(a) by cultured cells expressing the LDL receptor but had no
effect on Lp(a) uptake by cells deficient in the LDL receptor (Romagnuolo et al.
2017). The modest effect of PCSK9 inhibition on reducing Lp(a) is in direct con-
trast to the effect of statins on increasing Lp(a) and this mystery has yet to be
resolved.
4 Lp(a) Metabolism 85
Niacin
Niacin can modestly reduce Lp(a) levels. The degree of Lp(a) lowering by niacin
has been shown to be greater in subjects with smaller apo(a) isoform size that have
elevated Lp(a) levels (Artemeva et al. 2015). Ooi and colleagues examined the
mechanism by with niacin reduces Lp(a) have shown that niacin reduced apo(a) and
Lp(a) associated apoB100 production with no change in the FCR of these compo-
nents in subjects treated with niacin (1–2 g/day) with background rosuvastatin treat-
ment (Ooi et al. 2015). Croyal et al. also found that niacin (2 g/day) reduced the
production rate of apo(a) in hypertriglyceridemic subjects while the also reducing
the clearance rate (FCR) to a lesser degree (Croyal et al. 2015). The mechanism by
which niacin can influence Lp(a) metabolism is not clear, although variants in the
niacin receptor (hydroxyl-carboxylic receptor 2; HCAR2) have been shown to influ-
ence the Lp(a) response to niacin (Tuteja et al. 2017) suggesting that the mechanism
may lie downstream of HCAR2 receptor signaling. HCAR2 expression is relatively
high in white adipose tissue (Jadeja et al. 2019), and activation of HCAR2 on adipo-
cytes leads to inhibition of triglyceride lipolysis within adipose resulting in reduced
fatty acid delivery to liver. However, HCAR2 is also expressed to a lesser degree in
liver (Jadeja et al. 2019) and therefore it is possible that activation of HCAR2 on
hepatocytes by niacin had direct effects that lead to reduced LPA transcription.
Another drug class that has been shown to lower circulating Lp(a) levels are inhibitors
of cholesteryl ester transfer protein (CETP) which reduce Lp(a) levels from 24% to
40% (Thomas et al. 2017; Nicholls et al. 2016; Hovingh et al. 2015). This was shown
to be due to a decrease in the production of Lp(a) (Thomas et al. 2017), although the
mechanism behind this decrease is not entirely clear and requires more study.
86 J. S. Millar and D. J. Rader
A direct approach to lowering Lp(a) levels is to target the synthesis and production
of apo(a). The antisense oligonucleotide (ASO) pelacarsen targets the apo(a) mRNA
to promote degradation and thus reduces the synthesis of apo(a) protein. Treatment
of humans with pelacarsen resulted in a dose-dependent decrease in Lp(a) levels of
up to 80% (Tsimikas et al. 2020b). Pelacarsen is now being studied in a phase 3
cardiovascular outcome trial to assess the impact of this degree of Lp(a) reduction
on cardiovascular events.
Conclusion
Major advances have been made in our understanding of the factors that regulate
Lp(a) metabolism since its discovery 60 years ago. The synthesis of apo(a) is largely
under genetic control and ultimately determines the production rate and concentra-
tion of Lp(a) in plasma. A single receptor that regulates Lp(a) clearance has not
been identified to date; the LDL receptor does not appear to play a major physiolog-
ical role. Other receptors and apoE have been studied and may have modest effects
on Lp(a) clearance from plasma. Thus, it is possible that multiple receptors are
responsible for the catabolism of Lp(a). Future studies that examine Lp(a) metabo-
lism, particularly when conducted in response to treatment with novel drugs that
influence Lp(a) levels, should lead to further advances in our understanding of Lp(a)
biology and the factors that regulate its metabolism.
References
Bergmark C, Dewan A, Orsoni A, Merki E, Miller ER, Shin MJ, Binder CJ, Horkko S, Krauss
RM, Chapman MJ, et al. A novel function of lipoprotein [a] as a preferential carrier of oxidized
phospholipids in human plasma. J Lipid Res. 2008;49:2230–9.
Bersot TP, Innerarity TL, Pitas RE, Rall SC Jr, Weisgraber KH, Mahley RW. Fat feeding in humans
induces lipoproteins of density less than 1.006 that are enriched in apolipoprotein [a] and that
cause lipid accumulation in macrophages. J Clin Invest. 1986;77:622–30.
Bonen DK, Hausman AM, Hadjiagapiou C, Skarosi SF, Davidson NO. Expression of a recombi-
nant apolipoprotein(a) in HepG2 cells. Evidence for intracellular assembly of lipoprotein(a). J
Biol Chem. 1997;272:5659–67.
Cain WJ, Millar JS, Himebauch AS, Tietge UJ, Maugeais C, Usher D, Rader DJ. Lipoprotein [a]
is cleared from the plasma primarily by the liver in a process mediated by apolipoprotein [a]. J
Lipid Res. 2005;46:2681–91.
Clarke R, Peden JF, Hopewell JC, Kyriakou T, Goel A, Heath SC, Parish S, Barlera S, Franzosi
MG, Rust S, et al. Genetic variants associated with Lp(a) lipoprotein level and coronary dis-
ease. N Engl J Med. 2009;361:2518–28.
Croyal M, Ouguerram K, Passard M, Ferchaud-Roucher V, Chetiveaux M, Billon-Crossouard S,
de Gouville AC, Lambert G, Krempf M, Nobecourt E. Effects of extended-release nicotinic
acid on apolipoprotein (a) kinetics in hypertriglyceridemic patients. Arterioscler Thromb Vasc
Biol. 2015;35:2042–7.
Demant T, Seeberg K, Bedynek A, Seidel D. The metabolism of lipoprotein(a) and other apoli-
poprotein B-containing lipoproteins: a kinetic study in humans. Atherosclerosis. 2001;157:
325–39.
Diffenderfer MR, Lamon-Fava S, Marcovina SM, Barrett PH, Lel J, Dolnikowski GG, Berglund L,
Schaefer EJ. Distinct metabolism of apolipoproteins (a) and B-100 within plasma lipoprotein(a).
Metabolism. 2016;65:381–90.
Gemin M, Romagnuolo R, McIntyre AD, Scipione CA, Seidah NG, Boffa MB, Hegele RA,
Koschinsky ML. Sortilin promotes secretion of apolipoprotein(a) by hepatic cells. Circulation.
2018;134:A20583.
Hofmann SL, Eaton DL, Brown MS, McConathy WJ, Goldstein JL, Hammer RE. Overexpression
of human low density lipoprotein receptors leads to accelerated catabolism of Lp(a) lipoprotein
in transgenic mice. J Clin Invest. 1990;85:1542–7.
Hovingh GK, Kastelein JJ, van Deventer SJ, Round P, Ford J, Saleheen D, Rader DJ, Brewer
HB, Barter PJ. Cholesterol ester transfer protein inhibition by TA-8995 in patients with mild
dyslipidaemia (TULIP): a randomised, double-blind, placebo-controlled phase 2 trial. Lancet.
2015;386:452–60.
Innerarity TL, Weisgraber KH, Arnold KS, Mahley RW, Krauss RM, Vega GL, Grundy
SM. Familial defective apolipoprotein B-100: low density lipoproteins with abnormal receptor
binding. Proc Natl Acad Sci U S A. 1987;84:6919–23.
Jadeja RN, Jones MA, Fromal O, Powell FL, Khurana S, Singh N, Martin PM. Loss of GPR109A/
HCAR2 induces aging-associated hepatic steatosis. Aging (Albany NY). 2019;11:386–400.
Jenner JL, Seman LJ, Millar JS, Lamon-Fava S, Welty FK, Dolnikowski GG, Marcovina SM,
Lichtenstein AH, Barrett PH, deLuca C, et al. The metabolism of apolipoproteins (a) and B-100
within plasma lipoprotein (a) in human beings. Metabolism. 2005;54:361–9.
Kathiresan S, Melander O, Anevski D, Guiducci C, Burtt NP, Roos C, Hirschhorn JN, Berglund G,
Hedblad B, Groop L, et al. Polymorphisms associated with cholesterol and risk of cardiovascu-
lar events. N Engl J Med. 2008;358:1240–9.
Knight BL, Perombelon YF, Soutar AK, Wade DP, Seed M. Catabolism of lipoprotein(a) in famil-
ial hypercholesterolaemic subjects. Atherosclerosis. 1991;87:227–37.
Koschinsky ML, Tomlinson JE, Zioncheck TF, Schwartz K, Eaton DL, Lawn RM. Apolipo
protein(a): expression and characterization of a recombinant form of the protein in mammalian
cells. Biochemistry. 1991;30:5044–51.
Kostner GM, Gavish D, Leopold B, Bolzano K, Weintraub MS, Breslow JL. HMG CoA reductase
inhibitors lower LDL cholesterol without reducing Lp(a) levels. Circulation. 1989;80:1313–9.
88 J. S. Millar and D. J. Rader
Rader DJ, Cain W, Ikewaki K, Talley G, Zech LA, Usher D, Brewer HB Jr. The inverse association
of plasma lipoprotein(a) concentrations with apolipoprotein(a) isoform size is not due to differ-
ences in Lp(a) catabolism but to differences in production rate. J Clin Invest. 1994;93:2758–63.
Rader DJ, Mann WA, Cain W, Kraft HG, Usher D, Zech LA, Hoeg JM, Davignon J, Lupien P,
Grossman M, et al. The low density lipoprotein receptor is not required for normal catabolism
of Lp(a) in humans. J Clin Invest. 1995;95:1403–8.
Rainwater DL, Ludwig MJ, Haffner SM, VandeBerg JL. Lipid and lipoprotein factors associated
with variation in Lp(a) density. Arterioscler Thromb Vasc Biol. 1995;15:313–9.
Reblin T, Niemeier A, Meyer N, Willnow TE, Kronenberg F, Dieplinger H, Greten H, Beisiegel
U. Cellular uptake of lipoprotein[a] by mouse embryonic fibroblasts via the LDL receptor and
the LDL receptor-related protein. J Lipid Res. 1997;38:2103–10.
Reyes-Soffer G, Pavlyha M, Ngai C, Thomas T, Holleran S, Ramakrishnan R, Karmally W,
Nandakumar R, Fontanez N, Obunike J, et al. Effects of PCSK9 inhibition with alirocumab on
lipoprotein metabolism in healthy humans. Circulation. 2017;135:352–62.
Romagnuolo R, Scipione CA, Marcovina SM, Gemin M, Seidah NG, Boffa MB, Koschinsky
ML. Roles of the low density lipoprotein receptor and related receptors in inhibition of
lipoprotein(a) internalization by proprotein convertase subtilisin/kexin type 9. PLoS One.
2017;12:e0180869.
Santos RD, Raal FJ, Catapano AL, Witztum JL, Steinhagen-Thiessen E, Tsimikas S. Mipomersen,
an antisense oligonucleotide to apolipoprotein B-100, reduces lipoprotein(a) in various popula-
tions with hypercholesterolemia: results of 4 phase III trials. Arterioscler Thromb Vasc Biol.
2015;35:689–99.
Scanu AM, Lawn RM, Berg K. Lipoprotein(a) and atherosclerosis. Ann Intern Med.
1991;115:209–18.
Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of
lipoprotein(a). J Lipid Res. 2016;57:1339–59.
Sharma M, Redpath GM, Williams MJ, McCormick SP. Recycling of apolipoprotein(a) after
PlgRKT-mediated endocytosis of lipoprotein(a). Circ Res. 2017;120:1091–102.
Tada H, Kawashiri MA, Yoshida T, Teramoto R, Nohara A, Konno T, Inazu A, Mabuchi H,
Yamagishi M, Hayashi K. Lipoprotein(a) in familial hypercholesterolemia with proprotein
convertase subtilisin/kexin type 9 (PCSK9) gain-of-function mutations. Circ J. 2016;80:
512–8.
Tardif JC, Karwatowska-Prokopczuk E, Amour ES, Ballantyne CM, Shapiro MD, Moriarty PM,
Baum SJ, Hurh E, Bartlett VJ, Kingsbury J, et al. Apolipoprotein C-III reduction in subjects
with moderate hypertriglyceridaemia and at high cardiovascular risk. Eur Heart J. 2022;43:
1401–12.
Thomas T, Zhou H, Karmally W, Ramakrishnan R, Holleran S, Liu Y, Jumes P, Wagner JA,
Hubbard B, Previs SF, et al. CETP (cholesteryl ester transfer protein) inhibition with anacetra-
pib decreases production of lipoprotein(a) in mildly hypercholesterolemic subjects. Arterioscler
Thromb Vasc Biol. 2017;37:1770–5.
Trinder M, DeCastro ML, Azizi H, Cermakova L, Jackson LM, Frohlich J, Mancini GBJ, Francis
GA, Brunham LR. Ascertainment bias in the association between elevated lipoprotein(a) and
familial hypercholesterolemia. J Am Coll Cardiol. 2020;75:2682–93.
Tsimikas S, Witztum JL. The role of oxidized phospholipids in mediating lipoprotein(a) atheroge-
nicity. Curr Opin Lipidol. 2008;19:369–77.
Tsimikas S, Gordts P, Nora C, Yeang C, Witztum JL. Statin therapy increases lipoprotein(a) levels.
Eur Heart J. 2020a;41:2275–84.
Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, Tardif JC, Baum SJ, Steinhagen-
Thiessen E, Shapiro MD, Stroes ES, Moriarty PM, Nordestgaard BG, et al. Lipoprotein(a)
reduction in persons with cardiovascular disease. N Engl J Med. 2020b;382:244–55.
Tuteja S, Wang L, Dunbar RL, Chen J, DerOhannessian S, Marcovina SM, Elam M, Lader E,
Rader DJ. Genetic coding variants in the niacin receptor, hydroxyl-carboxylic acid receptor 2,
and response to niacin therapy. Pharmacogenet Genomics. 2017;27:285–93.
90 J. S. Millar and D. J. Rader
Utermann G, Menzel HJ, Kraft HG, Duba HC, Kemmler HG, Seitz C. Lp(a) glycoprotein phe-
notypes. Inheritance and relation to Lp(a)-lipoprotein concentrations in plasma. J Clin Invest.
1987;80:458–65.
Varvel S, McConnell JP, Tsimikas S. Prevalence of elevated Lp(a) mass levels and patient thresh-
olds in 532 359 patients in the United States. Arterioscler Thromb Vasc Biol. 2016;36:2239–45.
von Zychlinski A, Kleffmann T, Williams MJ, McCormick SP. Proteomics of Lipoprotein(a) identi-
fies a protein complement associated with response to wounding. J Proteome. 2011;74:2881–91.
Watts GF, Chan DC, Pang J, Ma L, Ying Q, Aggarwal S, Marcovina SM, Barrett PHR. PCSK9
Inhibition with alirocumab increases the catabolism of lipoprotein(a) particles in statin-treated
patients with elevated lipoprotein(a). Metabolism. 2020;107:154221.
White AL, Rainwater DL, Lanford RE. Intracellular maturation of apolipoprotein[a] and assembly
of lipoprotein[a] in primary baboon hepatocytes. J Lipid Res. 1993;34:509–17.
White AL, Hixson JE, Rainwater DL, Lanford RE. Molecular basis for “null” lipoprotein(a)
phenotypes and the influence of apolipoprotein(a) size on plasma lipoprotein(a) level in the
baboon. J Biol Chem. 1994;269:9060–6.
Yang XP, Amar MJ, Vaisman B, Bocharov AV, Vishnyakova TG, Freeman LA, Kurlander RJ,
Patterson AP, Becker LC, Remaley AT. Scavenger receptor-BI is a receptor for lipoprotein(a).
J Lipid Res. 2013;54:2450–7.
Yang X, Lee SR, Choi YS, Alexander VJ, Digenio A, Yang Q, Miller YI, Witztum JL, Tsimikas
S. Reduction in lipoprotein-associated apoC-III levels following volanesorsen therapy: phase 2
randomized trial results. J Lipid Res. 2016;57:706–13.
Yeang C, Wilkinson MJ, Tsimikas S. Lipoprotein(a) and oxidized phospholipids in calcific aortic
valve stenosis. Curr Opin Cardiol. 2016;31:440–50.
Chapter 5
Contemporary Aspects of Lp(a)
Metabolism and Therapies Based
on Tracer Kinetic Studies in Humans
Bullet Points
• Lipoprotein(a) [Lp(a)] is an inherited and causal risk factor for atherosclerotic
cardiovascular disease (ASCVD) and aortic valve stenosis.
• Use of stable isotope tracers and compartmental modelling has provided deeper
understanding of the physiology and pathophysiology of Lp(a) metabolism
in humans.
• Plasma Lp(a) concentration is predominantly determined by the rate of produc-
tion of Lp(a) particles, irrespective of apo(a) isoform size and background ther-
apy with statins.
• Niacin and cholesteryl ester transfer protein inhibitors lower plasma Lp(a) con-
centration by increasing the clearance or catabolism of apo(a).
• ApoB antisense oligonucleotides lower plasma Lp(a) concentration by decreas-
ing hepatic production.
• Proprotein convertase subtilisin kexin type 9 inhibitors can lower plasma Lp(a)
concentration by a dual mode of action involving both increased clearance and
decreased production of apo(a),
• Further studies should investigate nucleic acid-based inhibitors for apo(a),
angiopoietin-like 3 and apoC-III inhibitors on the metabolism of Lp(a) and other
lipoproteins.
D. C Chan · J. Pang
Medical School, University of Western Australia, Perth, WA, Australia
e-mail: [email protected]; [email protected]
G. F Watts (*)
Medical School, University of Western Australia, Perth, WA, Australia
Lipid Disorders Clinic, Department of Cardiology and Internal Medicine, Royal Perth
Hospital, Perth, WA, Australia
e-mail: [email protected]
Introduction
Lipoprotein(a) [Lp(a)] is one of the most important genetically determined risk fac-
tors for atherosclerotic cardiovascular disease (ASCVD) and aortic valve stenosis
(Nordestgaard and Langsted 2016; Saleheen et al. 2017; Tsimikas et al. 2018; Cegla
et al. 2009; Arsenault and Kamstrup 2022; Reyes-Soffer et al. 2022). Large clinical
trials have consistently shown that patients with elevated Lp(a), even when treated
with statins, are at an increased risk of ASCVD (Khera et al. 2014; Nicholls et al.
2010). The metabolic pathways governing the metabolism of Lp(a) have been
extensively studied in cellular and animal model systems (McCormick and
Schneider 2019; Boffa and Koschinsky 2022; Chemello et al. 2022a). However,
only scare information is available on the metabolism of this lipoprotein in humans.
Use of stable isotopically labelled tracers and compartmental modelling has greatly
enhanced our understanding of Lp(a) metabolism (Chan et al. 2004; Barrett et al.
2006). In the present chapter, we review use of these techniques and its contribution
to key knowledge of the physiology and pathophysiology of Lp(a) metabolism in
humans. We focus on subjects with elevated Lp(a) and the mode of action of lipid-
regulating agents.
GG), are strongly associated with an elevated Lp(a) concentration (Clarke et al.
2009). Genome-wide association studies (GWAS) have also identified many com-
mon genetic variants of small effect which can aggregately influence Lp(a) concen-
tration (Coassin and Kronenberg 2022). Accordingly, a polygenic risk score for
predicting Lp(a) concentration has recently been reported, explaining approxi-
mately 60–70% of the variance in Lp(a) levels in the EPIC-Norfolk and UK Biobank
cohorts (Wu et al. 2021). APOE gene is one of the most important genetic factors
modulating Lp(a) concentrations (Li et al. 2015; Zekavat et al. 2018; Mack et al.
2017; Chemello et al. 2022b); the ε2 allele is associated with reduced Lp(a) concen-
trations, whereas the ε4 allele is associated with increased Lp(a) concentrations
compared with the ε3 allele (Moriarty et al. 2017). Several physiological states,
such as kidney, thyroid and liver disease, and ancestry, also contribute to the vari-
ability in Lp(a) concentration (Enkhmaa and Berglund 2022).
Fig. 5.1 Compartmental model to describe Lp(a)-apo(a) and Lp(a)-apoB-100 tracer kinetics.
Plasma leucine kinetics are described by a four-compartment model, which is connected to intra-
hepatic delay compartments (compartments 5 and 6) that accounts for the synthesis and secretion
of Lp(a)-apo(a) and Lp(a)-apoB-100, with compartments 7 and 8 describing the plasma kinetics of
Lp(a)-apo(a) and Lp(a)-apoB-100, respectively
Metabolism of Lipoprotein(a)
Fig. 5.2 Lp(a)-apo(a) and Lp(a)-apoB tracer-tracee ratio (%) in 20 statin-treated subjects includ-
ing association of Lp(a)-apo(a) and Lp(a)-apoB-100 fractional catabolic rates (FCR)
Ramos-Cáceres et al. 2022). Earlier radiolabelled kinetic studies suggest that apo(a)
is unlikely to be adducted to a triglyceride-rich very low-density lipoprotein (VLDL)
as a precursor of Lp(a) in the LDL/HDL density range (Krempler et al. 1980). In
contrast, apo(a) can be associated with TRLs, such as chylomicrons and chylomi-
cron remnants, after oral ingestion of a fatty meal (Bersot et al. 1986). This is sup-
ported by experimental evidence that the apoB-100-apo(a) complex within Lp(a)
particles have a high affinity for TRL particles (Marcoux et al. 1997). A significant
proportion of Lp(a) particles can bind non-covalently to TRLs in the hypertriglyc-
eridemic state. Consistent with this, we and others have demonstrated a redistribu-
tion of a significant portion of apo(a) protein from Lp(a) to the TRL fraction,
particularly in the postprandial state (Cohn et al. 1991; Ying et al. 2022). In a recent
study of FH, we found that the impaired postprandial TRL-apo(a) response to a fat
load was partially corrected by fish oil supplementation (Ying et al. 2022). The
reduction in postprandial TRL-apo(a) with fish oil supplementation in response to
the fat load was significantly associated with the corresponding reduction in post-
prandial triglyceride response. Hence, interaction with TRLs may influence the
metabolism or catabolism of Lp(a). The underlying kinetic mechanism remains to
be investigated employing stable isotopes and compartmental modelling.
It is well established that the liver is the main site of Lp(a) clearance and, to a much
lesser extent, the kidney and the arterial wall (McCormick and Schneider 2019).
The mechanisms of Lp(a) clearance from the circulation and the catalytic pathways
96 D. C Chan et al.
involved remain uncertain, however. Several cellular receptors have been proposed
to mediate the clearance of Lp(a) from the liver. These include LDL receptor and
other members of the LDL-receptor family such as VLDL receptor, LDL receptor-
related protein 1 (LRP1), megalin/gp330, scavenger receptor class B type 1 (SR-BI)
and plasminogen receptor (McCormick and Schneider 2019).
The role of the LDL receptor in Lp(a) clearance remains controversial. Several
experimental studies have demonstrated that LDL receptor can facilitate Lp(a) bind-
ing and uptake (Havekes et al. 1981; Reblin et al. 1997; Romagnuolo et al. 2015),
and in mice overexpressing LDL receptor the clearance of Lp(a) particles is signifi-
cantly increased (Hofmann et al. 1990). Very few kinetic studies have specifically
investigated the metabolism of Lp(a) in patients with LDL receptor defects, such as
familial hypercholesterolemia (FH). Using exogenous radiolabelled tracers, Rader
et al. found that the clearance of Lp(a) did not differ significantly among homozy-
gous FH patients, heterozygous FH parents and non-FH control subjects (Rader
et al. 1995). Using endogenous stable isotope tracers, Croyal et al. reported that the
FCRs of Lp(a)-apo(a) were similar in patients with PCSK9 gain-of-function muta-
tions and control subjects (Croyal et al. 2020). Hence, defects in LDL receptor func-
tion do not appear to result in delayed clearance of Lp(a). In a study of healthy
normolipidemic men, there was no significant association between the FCRs of
apo(a) and LDL-apoB-100 (Chan et al. 2019). These kinetic findings suggest under
physiological conditions that the LDL receptors may not play a major role in Lp(a)
clearance. As discussed later, LDL receptor could play a role in Lp(a) clearance in
a supraphysiological condition in which the activity of LDL receptors is markedly
upregulated, such as in patients who are treated with a combination of statins and
PCSK9 inhibitors (Watts et al. 2018).
In a kinetic study of healthy normolipidemic men with a wide range of plasma Lp(a)
concentration, Lp(a) particle concentration was significantly and positively associ-
ated with apo(a) production rate (PR) and inversely with apo(a) FCR (Chan et al.
2019). In another study of statin-treated subjects, plasma concentration of apo(a)
was significantly and positively associated with apo(a) PR in patients with both
normal and elevated Lp(a) concentrations (Ma et al. 2019b). However, there was no
significant association between plasma apo(a) concentration and FCR in either of
the groups. Hence, these observations reinforce the notion that plasma concentra-
tions of Lp(a) are primarily determined by the rates of production and not clearance,
irrespective of background statin use. Accordingly, plasma concentration and PR of
apo(a) were significantly higher in statin-treated patients with elevated Lp(a)
5 Contemporary Aspects of Lp(a) Metabolism and Therapies Based on Tracer Kinetic… 97
b c
Fig. 5.3 Kinetic parameters of apo(a) in statin-treated subjects with (a) normal (<75 nmol/L), (b)
high (75–145 nmol/L) and (c) very high apo(a) concentrations (>145 nmol/L). Data presented as
mean ± SEM. Apo apolipoprotein, FCR fraction catabolic rate, PR production rate. *P < 0.001
compared with normal apo(a) group. †P < 0.001 compared with normal and moderate-high apo(a)
group using ANOVA
compared with those with normal Lp(a) (Fig. 5.3a, b). The FCR of apo(a) did not
differ significantly between the groups (Fig. 5.3c). This finding suggests that ele-
vated plasma Lp(a) concentration is a consequence of increased hepatic production
of Lp(a) particles in patients with elevated Lp(a). In a constant-feeding study of
healthy individuals, patients with high Lp(a) had increased apo(a) PR and reduced
FCR compared with those without elevated Lp(a) concentration (Jenner et al. 2005).
Plasma concentrations of Lp(a) were correlated significantly with both apo(a) PR
and negatively with apo(a) FCR. These findings implicate a role of Lp(a) catabolism
in determining Lp(a) plasma concentrations in the fed state.
isoforms of different sizes may have different binding affinities for the LDL recep-
tor or other receptors (März et al. 1993). Lp(a) particles with larger isoform size
have been shown to be more effectively removed via LDL receptor independent
pathways.
In a study of healthy normolipidemic subjects, subjects with smaller apo(a) iso-
form sizes (≤22 KIV repeats) had significantly higher apo(a) concentration and PR,
and lower apo(a) FCR than those with larger sizes (Chan et al. 2019). Plasma apo(a)
concentration was significantly associated with apo(a) PR, but not with FCR in
subjects with smaller apo(a) isoform size. In contrast, both apo(a) PR and FCR
were significantly associated with plasma apo(a) concentrations in subjects with
larger isoforms. Similar observations were observed in patients who were on statin
(Ma et al. 2019c). Taken together, these findings again suggest that the plasma
Lp(a) concentration is predominantly determined by the rate of production of Lp(a)
particles, irrespective of apo(a) isoform size and background statin use. Lp(a) par-
ticle catabolism may only play a modest role in determining Lp(a) concentration in
subjects with larger apo(a) isoform size. These observations also support the clini-
cal use of agents that target the hepatic production and secretion of Lp(a)
(Tsimikas 2017).
As discussed earlier, APOE genotype can influence the concentration of Lp(a)
(Moriarty et al. 2017; Croyal et al. 2020; Chemello et al. 2022a). However, the
effect of APOE genotype, particularly the presence of apoE2 and apoE4, on Lp(a)
concentrations is known to be affected by the size of apo(a) (Klausen et al. 1996;
Blanchard et al. 2021). Accordingly, the effect of apoE genotype on the metabolism
of Lp(a) in subjects with large and small apo(a) isoform merits further
investigation.
Statins
may tend to decrease plasma Lp(a) concentrations (Tsimikas et al. 2020). The
statin-induced increase in Lp(a) level is supported by experimental evidence in
HepG2 cells showing a higher LPA mRNA level in response to atorvastatin
(Tsimikas et al. 2020). In a study of healthy normolipidemic subjects, atorvastatin
(80 mg daily) did not significantly alter the FCR or PR of apo(a) (Watts et al. 2017).
This finding does not support a role of LDL receptor in the regulation of apo(a) FCR
under physiological condition. However, it remains unclear whether statin has a
potential impact on Lp(a) metabolism in subjects with high Lp(a) concentration.
There is also evidence showing that statins increase Lp(a) levels exclusively in
patients with a small size apo(a) defined as ≤22 KIV repeats (Yahya et al. 2019).
The precise mechanisms of action of this effect on Lp(a) metabolism remain to be
investigated.
Niacin
Niacin is one of few agents that can significantly lower plasma Lp(a) concentra-
tions. Experimental data suggest that niacin decreases the expression of LPA
mRNA (Chennamsetty et al. 2012). This is consistent with a kinetic study showing
that niacin lowered Lp(a) concentration by decreasing the production of apo(a) in
non-diabetic, obese and hypertriglyceridemic men (Croyal et al. 2015). The lower-
ing of the PR of apo(a) by niacin was confirmed in another postprandial kinetic
study in statin-treated patients with type 2 diabetes (Ooi et al. 2015). In this study,
extended-release niacin (1–2 g/day) significantly decreased plasma Lp(a) concen-
tration and the production rates of apo(a), with greater treatment effect in indi-
viduals with elevated Lp(a) concentration. This is consistent with another study
showing that extended-release niacin was more effective in lowering Lp(a) level in
subjects with small apo(a) isoform than those with large isoform (Artemeva
et al. 2015).
PCSK9 Inhibitors
in Lp(a) could also partly mediate the cardiovascular benefit of PCSK9 mAbs
(Bittner et al. 2020; Schwartz et al. 2021).
In a kinetic study of healthy normolipidemic men, evolocumab monotherapy
significantly decreased plasma Lp(a) concentration chiefly by reducing the PR of
apo(a) with no effect on the corresponding FCR (Watts et al. 2018). This effect is
consistent with a tracer study conducted in non-human primates in which ali-
rocumab decreased the PR of apo(a) (Croyal et al. 2018). The mechanistic effect of
evolocumab may involve reduced hepatic production of Lp(a) by decreasing the
assembly of Lp(a) particles through the reduction of apo(a) binding with LDL on
the surface of hepatocytes (Lambert et al. 2017). This speculation is supported by
in vitro studies showing that PCSK9 induces Lp(a) intracellular assembly and secre-
tion, whereas PCSK9 mAbs reduce the extracellular release of Lp(a) (Villard
et al. 2016).
However, as combination therapy with high-dose atorvastatin, evolocumab
reduced the plasma concentration of Lp(a) chiefly by a significant increase in the
FCR of apo(a) (Watts et al. 2018). The PR of Lp(a) was not significantly altered
with the combination. Similar results were also found in another kinetic study in
healthy individuals receiving alirocumab treatment (Reyes-Soffer et al. 2017).
However, the increase in apo(a) FCR in the latter study was not statistically signifi-
cant, probably owing to greater variability in study subject characteristics (e.g.
mixed race and gender). The mechanistic effect of evolocumab in combination with
atorvastatin may involve supraphysiological upregulation of the activity of LDL
receptors and decreased competition of Lp(a) with very low concentrations of LDLs
for clearance by these receptors. This mechanism suggests that the LDL receptor
likely plays a significant role in mediating Lp(a) clearance only when its expression
is markedly upregulated and when LDL plasma levels are substantially lowered,
allowing decreased competition between LDL and Lp(a) for receptor-mediated
uptake in the liver.
The mechanism of action of PCSK9 inhibition has recently been studied in
statin-treated patients with high Lp(a). Using stable isotopes, PCSK9 inhibition
with alirocumab-lowered plasma Lp(a) concentration by increasing apo(a) FCR in
patients with elevated Lp(a) receiving maximally tolerated statin therapy (Watts
et al. 2020). However, in patients with very high-Lp(a) concentration, alirocumab
significantly lowered plasma Lp(a) concentration by a dual mode of action involv-
ing both increased clearance and decreased production of apo(a) (Ying et al. 2022).
Taken together, the mechanistic action of PCSK9 mAbs on the PR and FCR of
apo(a) appears to be dependent on background statin use and Lp(a) concentration at
baseline.
Unlike evolocumab or alirocumab, small interfering RNA on PCSK9 mRNA
transcript (e.g. Inclisiran) is a new approach to targeting PCSK9 intracellularly
(German and Shapiro 2020; Smith and White 2022). This novel agent was shown to
inhibit hepatic synthesis of the PCSK9 protein, and lower apoB-100-containing
lipoproteins, including Lp(a) (Ray et al. 2020; Raal et al. 2020). This implies that
the effect of PCSK9 inhibition on Lp(a) is irrespective of mode of inhibition of
102 D. C Chan et al.
CETP Inhibitors
supporting a role for the LDL receptors or related receptors in the clearance of Lp(a)
particles.
Other Therapies
ASCVD. This atherogenic disorder has received little attention due to a significant
knowledge gap in understanding Lp(a) pathophysiology. Stable isotope tracer meth-
ods provide unique information of the dynamics of Lp(a) particles in the circulation.
The interferences from these studies are important for understanding the metabo-
lism of Lp(a) and for developing new therapies. Knowledge of the mode of action
of therapeutic interventions is also important for informing shared-decision making
and improving adherences to therapies. Future research is still needed to understand
whole body metabolism of Lp(a), including the stability of the covalent bonding
between apo(a) and apoB-100, the potential recycling of apo(a) in the circulation,
the possible formation of Lp(a) complexes with TRLs, and the relative roles of
hepatic and renal receptors in the clearance of Lp(a) particles. The precise modes of
action of CETP inhibitors, apoB ASO and THR agonists on the metabolism of
Lp(a), particularly in patients with high Lp(a), also merit further clarification.
While several therapeutic interventions can lower plasma Lp(a) concentrations
(Korneva et al. 2021), it is uncertain that it would mitigate the adverse effects of
elevated Lp(a) on ASCVD. Nevertheless, some of the cardiovascular benefit of
PCSK9 mAbs in clinical outcome trials are known to be mediated by the lowering
of Lp(a) independently of the concurrent reduction in LDL cholesterol. More
aggressive treatment strategies involve use of multiple lipid-regulating agents to
treat elevated Lp(a). This approach harnesses the complementary mechanisms of
action of the different agents. Possible combinations include PCSK9 inhibitor with
niacin, CETP inhibitor or THR agonist. Inhibiting hepatic apo(a) synthesis with
nucleic acid therapeutics has emerged as a potent approach to reduce plasma Lp(a)
levels up to 90% which is not affected by LPA gene variants and isoform size
(Karwatowska-Prokopczuk et al. 2021). The effect of this novel and specific agent
on the metabolism of Lp(a) and other apoB-containing lipoproteins warrants inves-
tigation. Further studies are required to characterize the mode of action of newer
lipid-regulating agents on the metabolism TRLs and Lp(a). These include inhibitors
of angiopoietin-like protein 3 (ANGPTL3) and apoC-III (antibodies and/or nucleic
acid-based ASO therapies) (Ward et al. 2022).
Conflicts of Interest GFW has received honoraria for lectures and advisory boards
or research grants from Amgen, Arrowhead, AstraZeneca, Esperion, Kowa, Novartis,
Regeneron and Sanofi. DCC has nothing to declare.
Acknowledgement JP was supported by the National Health and Medical Research Council
(HMRC) Investigator Grant.
5 Contemporary Aspects of Lp(a) Metabolism and Therapies Based on Tracer Kinetic… 105
References
Chemello K, Chan DC, Lambert G, Watts GF. Recent advances in demystifying the metabolism of
lipoprotein(a). Atherosclerosis. 2022b;349:82–91.
Chennamsetty I, Kostner KM, Claudel T, Vinod M, Frank S, Weiss TS, Trauner M, Kostner
GM. Nicotinic acid inhibits hepatic APOA gene expression: studies in humans and in trans-
genic mice. J Lipid Res. 2012;53:2405–12.
Clarke R, Peden JF, Hopewell JC, Kyriakou T, Goel A, Heath SC, Parish S, Barlera S, Franzosi
MG, Rust S, Bennett D. Genetic variants associated with Lp (a) lipoprotein level and coronary
disease. N Engl J Med. 2009;361:2518–28.
Coassin S, Kronenberg F. Lipoprotein(a) beyond the kringle IV repeat polymorphism: the com-
plexity of genetic variation in the LPA gene. Atherosclerosis. 2022;349:17–35.
Cohn JS, Lam CW, Sullivan DR, Hensley WJ. Plasma lipoprotein distribution of apolipoprotein(a)
in the fed and fasted states. Atherosclerosis. 1991;90:59–66.
Croyal M, Ouguerram K, Passard M, Ferchaud-Roucher V, Chétiveaux M, Billon-Crossouard S,
de Gouville AC, Lambert G, Krempf M, Nobécourt E. Effects of extended-release nicotinic
acid on apolipoprotein (a) kinetics in hypertriglyceridemic patients. Arterioscler Thromb Vasc
Biol. 2015;35:2042–7.
Croyal M, Tran TT, Blanchard RH, Le Bail JC, Villard EF, Poirier B, Aguesse A, Billon-Crossouard
S, Ramin-Mangata S, Blanchard V, Nativel B, Chemello K, Khantalin I, Thedrez A, Janiak
P, Krempf M, Boixel C, Lambert G, Guillot E. PCSK9 inhibition with alirocumab reduces
lipoprotein(a) levels in nonhuman primates by lowering apolipoprotein(a) production rate. Clin
Sci (Lond). 2018;132:1075–83.
Croyal M, Blanchard V, Ouguerram K, Chétiveaux M, Cabioch L, Moyon T, Billon-Crossouard
S, Aguesse A, Bernardeau K, Le May C, Flet L, Lambert G, Hadjadj S, Cariou B, Krempf
M, Nobécourt-Dupuy E. VLDL (very-low-density lipoprotein)-apo E (apolipoprotein E)
may influence Lp(a) (lipoprotein [a]) synthesis or assembly. Arterioscler Thromb Vasc Biol.
2020;40:819–29.
Cuchel M, Bloedon LT, Szapary PO, Kolansky DM, Wolfe ML, Sarkis A, Millar JS, Ikewaki K,
Siegelman ES, Gregg RE, Rader DJ. Inhibition of microsomal triglyceride transfer protein in
familial hypercholesterolemia. N Engl J Med. 2007;356:148–56.
Cuchel M, Rader DJ. Microsomal transfer protein inhibition in humans. Curr Opin Lipidol.
2013;24:246–50.
de Boer LM, Oorthuys AOJ, Wiegman A, Langendam MW, Kroon J, Spijker R, Zwinderman AH,
Hutten BA. Statin therapy and lipoprotein(a) levels: a systematic review and meta-analysis. Eur
J Prev Cardiol. 2022;29:779–92.
Deedwania P, Murphy SA, Scheen A, Badariene J, Pineda AL, Honarpour N, Keech AC, Sever
PS, Pedersen TR, Sabatine MS, Giugliano RP. Efficacy and safety of PCSK9 inhibition with
evolocumab in reducing cardiovascular events in patients with metabolic syndrome receiv-
ing statin therapy: secondary analysis from the FOURIER randomized clinical trial. JAMA
Cardiol. 2021;6:139–47.
Duprez DA, Handelsman Y, Koren M. Cardiovascular outcomes and proprotein convertase sub-
tilisin/kexin type 9 inhibitors: current data and future prospects. Vasc Health Risk Manag.
2020;16:403–18.
Enkhmaa B, Berglund L. Non-genetic influences on lipoprotein(a) concentrations. Atherosclerosis.
2022;349:53–62.
Erion MD, Cable EE, Ito BR, Jiang H, Fujitaki JM, Finn PD, Zhang BH, Hou J, Boyer SH, van
Poelje PD, Linemeyer DL. Targeting thyroid hormone receptor-beta agonists to the liver
reduces cholesterol and triglycerides and improves the therapeutic index. Proc Natl Acad Sci
U S A. 2007;104:15490–4595.
Ferri N, Grego MF, Corsini A, Ruscica M. Proprotein convertase subtilisin/kexin type 9: an update
on the cardiovascular outcome studies. Eur Heart J Suppl. 2020;22:E64–7.
German CA, Shapiro MD. Small interfering RNA therapeutic inclisiran: a new approach to target-
ing PCSK9. BioDrugs. 2020;34:1–9.
Ginsberg HN. Efficacy and mechanisms of action of statins in the treatment of diabetic dyslipid-
emia. J Clin Endocrinol Metab. 2006;91:383–92.
5 Contemporary Aspects of Lp(a) Metabolism and Therapies Based on Tracer Kinetic… 107
Lincoff AM, Nicholls SJ, Riesmeyer JS, Barter PJ, Brewer HB, Fox KAA, Gibson CM, Granger C,
Menon V, Montalescot G, Rader D, Tall AR, McErlean E, Wolski K, Ruotolo G, Vangerow B,
Weerakkody G, Goodman SG, Conde D, McGuire DK, Nicolau JC, Leiva-Pons JL, Pesant Y,
Li W, Kandath D, Kouz S, Tahirkheli N, Mason D, Nissen SE. Evacetrapib and cardiovascular
outcomes in high-risk vascular disease. N Engl J Med. 2017;376:1933–42.
Lobentanz E-M, Krasznai K, Gruber A, Brunner C, Müller H-J, Sattler J, Kraft H-G, Utermann G,
Dieplinger H. Intracellular metabolism of human apolipoprotein(a) in stably transfected Hep
G2 cells. Biochemistry. 1998;37:5417–25.
Ma L, Chan DC, Ooi EMM, Barrett PHR, Watts GF. Fractional turnover of apolipoprotein(a)
and apolipoprotein B-100 within plasma lipoprotein(a) particles in statin-treated patients with
elevated and normal Lp(a) concentration. Metabolism. 2019a;96:8–11.
Ma L, Chan DC, Ooi EMM, Marcovina SM, Barrett PHR, Watts GF. Apolipoprotein(a) kinetics
in statin-treated patients with elevated plasma lipoprotein(a) concentration. J Clin Endocrinol
Metab. 2019b;104:6247–55.
Ma L, Waldmann E, Ooi EM, Chan DC, Barrett HP, Watts GF, Parhofer KG. Lipoprotein (a)
and low-density lipoprotein apolipoprotein B metabolism following apheresis in patients with
elevated lipoprotein (a) and coronary artery disease. Eur J Clin Investig. 2019c;49:e13053.
Mack S, Coassin S, Rueedi R, Yousri NA, Seppälä I, Gieger C, Schönherr S, Forer L, Erhart G,
Marques-Vidal P, Ried JS, Waeber G, Bergmann S, Dähnhardt D, Stöckl A, Raitakari OT,
Kähönen M, Peters A, Meitinger T, Strauch K, Kedenko L, Paulweber B, Lehtimäki T, Hunt
SC, Vollenweider P, Lamina C, Kronenberg F. A genome-wide association meta-analysis
on lipoprotein (a) concentrations adjusted for apolipoprotein (a) isoforms. J Lipid Res.
2017;58:1834–44.
Marcoux C, Lussier-Cacan S, Davignon J, Cohn JS. Association of Lp(a) rather than integrally-
bound apo(a) with triglyceride-rich lipoproteins of human subjects. Biochim Biophys Acta.
1997;1346:261–74.
Marcovina SM, Koschinsky ML. Lipoprotein(a) concentration and apolipoprotein(a) size—a syn-
ergistic role in advanced atherosclerosis? Circulation. 1999;100:1151–3.
Marcovina SM, Hobbs HH, Albers JJ. Relation between number of apolipoprotein(a) kringle 4
repeats and mobility of isoforms in agarose gel: basis for a standardized isoform nomenclature.
Clin Chem. 1996;42:436–9.
März W, Beckmann A, Scharnagl H, Siekmeier R, Mondorf U, Held I, Schneider W, Preissner KT,
Curtiss LK, Gross W, Huttinger M. Heterogeneous lipoprotein (a) size isoforms differ by their
interaction with the low density lipoprotein receptor and the low density lipoprotein receptor-
related protein/alpha 2-macroglobulin receptor. FEBS Lett. 1993;325:271–5.
McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors.
Pathology. 2019;51:155–64.
Moriarty PM, Varvel SA, Gordts PL, McConnell JP, Tsimikas S. Lipoprotein(a) mass levels
increase significantly according to APOE genotype: an analysis of 431 239 patients. Arterioscler
Thromb Vasc Biol. 2017;37:580–8.
Nandakumar R, Matveyenko A, Thomas T, Pavlyha M, Ngai C, Holleran S, Ramakrishnan R,
Ginsberg HN, Karmally W, Marcovina SM, Reyes-Soffer G. Effects of mipomersen, an apo-
lipoprotein B100 antisense, on lipoprotein (a) metabolism in healthy subjects. J Lipid Res.
2018;59:2397–402.
Nassir F, Bonen DK, Davidson NO. Apolipoprotein(a) synthesis and secretion from hepatoma
cells is coupled to triglyceride synthesis and secretion. J Biol Chem. 1998;273:17793–800.
Nicholls SJ, Tang WW, Scoffone H, Brennan DM, Hartiala J, Allayee H, Hazen SL. Lipoprotein(a)
levels and long-term cardiovascular risk in the contemporary era of statin therapy. J Lipid Res.
2010;51:3055–61.
Nordestgaard BG, Langsted A. Lipoprotein (a) as a cause of cardiovascular disease: insights from
epidemiology, genetics, and biology. J Lipid Res. 2016;57:1953–75.
Ooi EM, Watts GF, Chan DC, Pang J, Tenneti VS, Hamilton SJ, McCormick SP, Marcovina
SM, Barrett PH. Effects of extended-release niacin on the postprandial metabolism of Lp(a)
5 Contemporary Aspects of Lp(a) Metabolism and Therapies Based on Tracer Kinetic… 109
A, Price JF, Hughes AD, Gaunt TR, Franceschini N, Mook-Kanamori DO, Zwierzyna M, Sofat
R, Hingorani AD, Finan C. Cholesteryl ester transfer protein (CETP) as a drug target for car-
diovascular disease. Nat Commun. 2021;12:5640.
Schwartz GG, Ballantyne CM. Existing and emerging strategies to lower lipoprotein(a).
Atherosclerosis. 2022;349:110–22.
Schwartz GG, Olsson AG, Abt M, Ballantyne CM, Barter PJ, Brumm J, Chaitman BR, Holme IM,
Kallend D, Leiter LA, Leitersdorf E, McMurray JJ, Mundl H, Nicholls SJ, Shah PK, Tardif JC,
Wright RS, dal OI. Effects of dalcetrapib in patients with a recent acute coronary syndrome. N
Engl J Med. 2012;367:2089–99.
Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Edelberg JM, Goodman SG,
Hanotin C, Harrington RA, Jukema JW, Lecorps G, Mahaffey KW, Moryusef A, Pordy R,
Quintero K, Roe MT, Sasiela WJ, Tamby JF, Tricoci P, White HD, Zeiher AM. Alirocumab and
cardiovascular outcomes after acute coronary syndrome. N Engl J Med. 2018;379:2097–107.
Schwartz GG, Szarek M, Bittner VA, Diaz R, Goodman SG, Jukema JW, Landmesser U,
López-Jaramillo P, Manvelian G, Pordy R, Scemama M, Sinnaeve PR, White HD, Gabriel
SP. Lipoprotein(a) and benefit of PCSK9 inhibition in patients with nominally controlled LDL
cholesterol. J Am Coll Cardiol. 2021;78:421–33.
Smith KW, White CM. Inclisiran: a novel small interfering RNA drug for low-density lipoprotein
reduction. J Clin Pharmacol. 2022; https://doi.org/10.1002/jcph.2045. Epub ahead of print.
Takagi H, Umemoto T. Atorvastatin decreases lipoprotein (a): a meta-analysis of randomized tri-
als. Int J Cardiol. 2012;154:183–6.
Tall AR. Plasma cholesteryl ester transfer protein. J Lipid Res. 1993;34:1255–74.
Thomas T, Zhou H, Karmally W, Ramakrishnan R, Holleran S, Liu Y, Jumes P, Wagner JA,
Hubbard B, Previs SF, Roddy T, Johnson-Levonas AO, Gutstein DE, Marcovina SM, Rader
DJ, Ginsberg HN, Millar JS, Reyes-Soffer G. CETP (Cholesteryl ester transfer protein) inhibi-
tion with anacetrapib decreases production of lipoprotein(a) in mildly hypercholesterolemic
subjects. Arterioscler Thromb Vasc Biol. 2017;37:1770–5.
Tsimikas S. Lipoprotein(a): diagnosis, prognosis, controversies, and emerging therapies. J Am
Coll Cardiol. 2017;69:692–711.
Tsimikas S, Fazio S, Ferdinand KC, Ginsberg HN, Koschinsky ML, Marcovina SM, Moriarty PM,
Rader DJ, Remaley AT, Reyes-Soffer G, Santos RD, Thanassoulis G, Witztum JL, Danthi S,
Olive M, Liu L. NHLBI working group recommendations to reduce lipoprotein(a)-mediated
risk of cardiovascular disease and aortic stenosis. J Am Coll Cardiol. 2018;71:177–92.
Tsimikas S, Gordts PLSM, Nora C, Yeang C, Witztum JL. Statin therapy increases lipoprotein (a)
levels. Eur Heart J. 2020;41:2275–84.
Villard EF, Thedrez A, Blankenstein J, Croyal M, Tran TT, Poirier B, Le Bail JC, Illiano S,
Nobécourt E, Krempf M, Blom DJ, Marais AD, Janiak P, Muslin AJ, Guillot E, Lambert
G. PCSK9 modulates the secretion but not the cellular uptake of lipoprotein(a) ex vivo: an
effect blunted by alirocumab. JACC Basic Transl Sci. 2016;1:419–27.
Wade DP, Knight BL, Harders-Spengel K, Soutar AK. Detection and quantitation of
apolipoprotein(a) mRNA in human liver and its relationship with plasma lipoprotein(a) con-
centration. Atherosclerosis. 1991;91:63–72.
Wang X, Li J, Ju J, Fan Y, Xu H. Effect of different types and dosages of statins on plasma
lipoprotein(a) levels: a network meta-analysis. Pharmacol Res. 2021;163:105275.
Ward NC, Chan DC, Watts GF. A tale of two new targets for hypertriglyceridaemia: which choice
of therapy? BioDrugs. 2022;36:121–35.
Watts GF, Chan DC, Dent R, Somaratne R, Wasserman SM, Scott R, Burrows S, Barrett
PHR. Factorial effects of evolocumab and atorvastatin on lipoprotein metabolism. Circulation.
2017;135:338–51.
Watts GF, Chan DC, Somaratne R, Wasserman SM, Scott R, Marcovina SM, Barrett PHR. Controlled
study of the effect of proprotein convertase subtilisin-kexin type 9 inhibition with evolocumab
on lipoprotein(a) particle kinetics. Eur Heart J. 2018;39:2577–85.
5 Contemporary Aspects of Lp(a) Metabolism and Therapies Based on Tracer Kinetic… 111
Watts GF, Chan DC, Pang J, Ma L, Ying Q, Aggarwal S, Marcovina SM, Barrett PHR. PCSK9
inhibition with alirocumab increases the catabolism of lipoprotein(a) particles in statin-treated
patients with elevated lipoprotein(a). Metabolism. 2020;107:154221.
White AL, Hixson JE, Rainwater DL, Lanford RE. Molecular basis for “null” lipoprotein(a)
phenotypes and the influence of apolipoprotein(a) size on plasma lipoprotein(a) level in the
baboon. J Biol Chem. 1994;269:9060–6.
Wu H, Luan J, Forgetta V, Engert JC, Thanassoulis G, Mooser V, Wareham NJ, Langenberg C,
Richards JB. Utility of genetically predicted lp(a) (lipoprotein [a]) and apoB levels for cardio-
vascular risk assessment. Circ Genom Precis Med. 2021;14:e003312.
Yahya R, Berk K, Verhoeven A, Bos S, van der Zee L, Touw J, Erhart G, Kronenberg F, Timman
R, Sijbrands E, Roeters van Lennep J, Mulder M. Statin treatment increases lipoprotein(a)
levels in subjects with low molecular weight apolipoprotein(a) phenotype. Atherosclerosis.
2019;289:201–5.
Ying Q, Chan DC, Watts GF. New insights into the regulation of lipoprotein metabolism by PCSK9:
lessons from stable isotope tracer studies in human subjects. Front Physiol. 2021;12:603910.
Ying Q, Croyal M, Chan DC, Blanchard V, Pang J, Krempf M, Watts GF. Effect of omega-3 fatty
acid supplementation on the postprandial metabolism of apolipoprotein(a) in familial hyper-
cholesterolemia. J Atheroscler Thromb. 2022; https://doi.org/10.5551/jat.63587. Epub ahead
of print.
Youssef A, Clark JR, Marcovina SM, Boffa MB, Koschinsky ML. Apo(a) and apoB interact non-
covalently within hepatocytes: implications for regulation of lp(a) levels by modulation of
apoB secretion. Arterioscler Thromb Vasc Biol. 2022;42:289–304.
Zekavat SM, Ruotsalainen S, Handsaker RE, Alver M, Bloom J, Poterba T, Seed C, Ernst J,
Chaffin M, Engreitz J, Peloso GM, Manichaikul A, Yang C, Ryan KA, Fu M, Johnson WC,
Tsai M, Budoff M, Vasan RS, Cupples LA, Rotter JI, Rich SS, Post W, Mitchell BD, Correa
A, Metspalu A, Wilson JG, Salomaa V, Kellis M, Daly MJ, Neale BM, McCarroll S, Surakka
I, Esko T, Ganna A, Ripatti S, Kathiresan S, Natarajan P. Deep coverage whole genome
sequences and plasma lipoprotein(a) in individuals of European and African ancestries. Nat
Commun. 2018;9:2606.
Chapter 6
Role of Proprotein Convertase Subtilisin
Kexin Type 9 in Lipoprotein(a)
Metabolism
PCSK9 is a serine protease mainly expressed in the liver (Seidah et al. 2014). It is
synthesized as a precursor that undergoes autocatalytic intramolecular processing to
form a mature enzyme. In 2003, Abifadel and colleagues identified missense muta-
tions in the gene encoding PCSK9 causative of familial hypercholesterolemia (FH)
(Abifadel et al. 2003). These mutations were later shown to be gain-of-function muta-
tions. In 2005, a causative association was established between loss-of-function muta-
tions in PCSK9 and low plasma concentrations of LDL-C, which was accompanied by
an astonishing reduction in global coronary heart disease risk (Cohen et al. 2006).
Strikingly, studies of individual homozygotes for PCSK9 loss-of-function mutations
demonstrate that a complete or near-complete absence of PCSK9 resulting in very low
levels of LDL-C is perfectly compatible with normal human health (Zhao et al. 2006).
Evidence for a direct role for PCSK9 in LDL-C metabolism came initially from
a series of studies showing that overexpression of PCSK9 promotes the accumula-
tion of LDL-C in the plasma of control mice but not in that of LDLR-deficient ani-
mals (Maxwell and Breslow 2004; Lagace et al. 2006). Mechanistically, the LDLR
promotes the cellular uptake of LDL by endocytosis. In the absence of PCSK9, the
acidic environment of the endosome promotes the dissociation of the receptor from
the LDL particle, and the LDLR is recycled to the cell surface, while LDL is routed
to the lysosome for degradation (Nassoury et al. 2007; Qian et al. 2007). PCSK9,
which is secreted from hepatocytes, binds to the LDLR at the surface of cells.
Following endocytosis, in the presence of PCSK9, the LDLR fails to change
a b
c d
Fig. 6.1 Lipoprotein(a) plasma levels are reduced in the absence or upon pharmacological inhibi-
tion of PCSK9—Lp(a) plasma concentrations are primarily determined genetically at the LPA
locus that chiefly governs the level of apo(a) synthesis and the subsequent rate of Lp(a) particle
assembly and secretion by hepatocytes (Panel a). PCSK9 targets the LDLR for lysosomal degrada-
tion, reducing its abundance at the surface of hepatocytes, and thus lowering the cellular uptake of
LDL (Panel b). In the absence of PCSK9, the intracellular assembly of Lp(a) may be reduced
(Panel c), whereas the abundance of LDLR at the cell surface is maximal, allowing optimal LDL
uptake by endocytosis without significantly altering Lp(a) clearance (Panel d)
6 Role of Proprotein Convertase Subtilisin Kexin Type 9 in Lipoprotein(a) Metabolism 115
Given the mode of action of PCSK9 as a circulating inhibitor of the LDLR, as well
as the healthy profile of individuals with reduced or absent PCSK9 function, PCSK9
rapidly gained status of a very clean drug target to lower LDL-C in humans. Several
drug development strategies have been tested to pharmacologically inhibit PCSK9,
the most advanced in terms of clinical development being two fully human mono-
clonal antibodies (mAbs). Large phase III programs with these mAbs have been
reported: the FOURIER program with evolocumab (Sabatine et al. 2015) and the
ODYSSEY program with alirocumab (Robinson et al. 2015) (Table 6.1). In a
Table 6.1 Lipoprotein (a) reductions by PCSK9 inhibition in Phase III trials
CV outcome
Patients’ number Pharmacological Lp(a) % % reduction
and agent and median (hazard
Trial characteristics follow-up duration reduction ratios) References
FOURIER 25,096 subjects Evolocumab, −26.9 Lp(a) upper O’Donoghue
with established 48 weeks median: et al. (2019)
CVD −23%
(HR, 0.77
[0.67–0.88])
Lp(a) lower
median −7%
(HR 0.93
[0.80–1.08])
4465 subjects Evolocumab, −25.5 – Sabatine et al.
from 5 phase 2 12 weeks (2015)
and 7 phase 3
trials
1359 subjects Evolocumab, −27.0 – Stein et al.
without CVD 12 weeks (2014)
with/without
background LLT
ODYSSEY 18,924 subjects Alirocumab, −23.6 −15% Bittner et al.
with CVD 146 weeks (HR 0.85 (2020)
[0.78–0.93])
3499 subjects Alirocumab, −25.6 (vs. −12% Ray et al.
(placebo- 84.6 weeks placebo) (HR 0.88 (2019)
controlled) −21.4 (vs. [0.78–0.98])
1484 ezetimibe)
(ezetimibe-
controlled)
ORION 482 subjects with Inclisiran, −17.2 – Raal et al.
heterozygous FH 77 weeks (2020)
1561 subjects Inclisiran, −25.6 – Ray et al.
with established 77 weeks −18.6 (2020)
CVD
1617 subjects
with CVD
equivalent
116 A. Gallo et al.
nutshell, these trials have unequivocally shown that PCSK9 inhibition robustly and
safely lowers LDL-C levels regardless of background lipid-lowering therapy and
reduces cardiovascular disease (CVD). In addition to mAbs that sequester PCSK9 in
circulation and that have been approved by regulating bodies and are now prescribed
to patients in many countries, other approaches to PCSK9 inhibition are currently in
late-stage clinical development. The small interfering RNA (siRNA) inclisiran that
targets PCSK9 hepatic production was approved by the US Food and Drug
Administration in December 2021, following reports of positive results of the
ORION phase III clinical trial with this drug (Ray et al. 2020).
Statins are the most prescribed lipid-lowering drugs. They enhance LDLR gene
and protein expression and thereby markedly reduce LDL-C. As mentioned above,
PCSK9 inhibitors lower the intracellular degradation of the LDLR, thus increasing
the abundance of LDLR at the cell surface and thus reducing LDL-C levels. Whereas
statins are neutral or can even elevate the plasma concentrations of Lp(a), PCSK9
inhibitors not only reduce LDL-C (by 50–60% on average) but also concomitantly
lower Lp(a) plasma levels by 20–30% (Gaudet et al. 2014; Raal et al. 2016; Lambert
et al. 2017). This intriguing observation has led to a flurry of research aimed at
investigating the role of PCSK9 in Lp(a) metabolism.
Several investigators have tried to elucidate the molecular, cellular, and meta-
bolic pathways governing the production of Lp(a), the contribution of Lp(a) to
lipid transport in the plasma, and the catabolic fate of Lp(a). The metabolism of
this enigmatic lipoprotein nevertheless remains incompletely understood. Unlike
LDL, Lp(a) is not the direct product of very low-density lipoprotein (VLDL)
metabolism (Krempler et al. 1979). Lp(a) assembly appears to be a two-step pro-
cess: (1) apo(a) and apoB associate noncovalently through the interactions between
weak lysine binding sites located in apo(a) KIV7 and KIV8 domains and lysine
residues located on apoB100; (2) a disulfide bond is formed between the only
“free” cysteine of apo(a) located in its KIV9 domain and a cysteine located in the
C-terminal domain of apoB100 (Gabel and Koschinsky 1998; Youssef et al. 2022).
Most in vitro and in vivo kinetic studies suggest that the noncovalent association
between apoB100 and apo(a) takes place within hepatocytes, whereas their cova-
lent attachment which is enzyme-catalyzed occurs extracellularly (Youssef et al.
2022). As referred to above, the plasma concentrations of Lp(a) have a strong heri-
table component related to genetic variations in the number of KIV2 repeats, with
epidemiological studies consistently demonstrating an inverse association between
the size of apo(a) and plasma Lp(a) concentrations (Kronenberg and Utermann
2013). Apo(a) production rate and apo(a) isoform size, but not apo(a) fractional
catabolic rate (FCR), were shown to be significant predictors of plasma Lp(a) con-
centrations (Watts et al. 2018). In addition, patients with elevated Lp(a) concentra-
tions have smaller apo(a) isoform sizes and higher apo(a) production rates than
patients with normal Lp(a) concentration, the FCR of Lp(a)-apo(a) not differing
significantly between these groups of patients. These observations clearly support
that plasma concentrations of Lp(a) are primarily determined by the rates of pro-
duction and not clearance.
The mechanisms of Lp(a) clearance from the blood and the catalytic pathways
involved remain highly uncertain. It is well established that the liver is the major site
of Lp(a) clearance followed to a much lower extent by the kidney (Kronenberg
2014). Multiple pathways for Lp(a) clearance have been proposed (McCormick and
Schneider 2019). For instance, the scavenger receptor BI (SR-BI) has been shown
to promote the selective uptake of Lp(a) cholesterol esters in cells and in SR-BI
transgenic mice (Yang et al. 2013). Given the strong homology between apo(a) and
plasminogen, the role of plasminogen receptors in mediating Lp(a) clearance has
been evaluated (Sharma et al. 2017). One of them, the plasminogen receptor pre-
senting a C-terminal lysine (PLGRKT), was shown to mediate the cellular uptake of
Lp(a) by human hepatoma cells and primary human fibroblasts. This study also
showed that the LDL component of Lp(a) undergoes lysosomal degradation,
whereas apo(a) traffics through recycling endosomes and is re-secreted. Several
members of the LDLR family of receptors have also been proposed to mediate
118 A. Gallo et al.
whole Lp(a) particle cellular uptake. Thus, the VLDL receptor binds apo(a) and
allows the internalization and subsequent degradation of Lp(a) in macrophages
(Argraves et al. 1997). The LDLR-related protein 1 (LPR1) and megalin/gp330
(known as LRP2) also play a role in Lp(a) binding (Reblin et al. 1997; Niemeier
et al. 1999) cellular uptake and degradation in vitro. LRP8 (formerly known as the
apoB,E receptor) is also able to bind Lp(a) at the plasma membrane (Steyrer and
Kostner 1990), but it remains to be seen whether this promotes Lp(a) particle cel-
lular uptake and degradation. The cellular uptake of Lp(a) was, however, recently
shown to be unaffected in HepG2 hepatoma cells overexpressing either the VLDLR,
LRP1, or LRP8 (Romagnuolo et al. 2017). Given the important structural similari-
ties between LDL and Lp(a), and the Lp(a) lowering effects of PCSK9 inhibitors,
the LDLR has received the most attention as a candidate receptor for Lp(a) over the
past decades.
The initial reports showed that Lp(a) can bind to the LDLR with a lower affinity
than LDL (Snyder et al. 1992). It has also been proposed that Lp(a) could associate
with LDL and undergo LDLR-mediated clearance by a hitchhiking process (Hofer
et al. 1997). In HepG2 and primary human fibroblasts, PCSK9 was shown to reduce
the binding and the cellular uptake of Lp(a) via the LDLR (Raal et al. 2016;
Romagnuolo et al. 2015). These results were confirmed in HuH7 hepatoma cells
and in primary mouse hepatocytes (Romagnuolo et al. 2017). In contrast, other
studies found no significant role for the LDLR in mediating Lp(a) cellular uptake in
primary human hepatocytes, but also in fibroblasts and HepG2 cells (Sharma et al.
2017; Villard et al. 2016). Neither did they find any significant difference in Lp(a)
cellular uptake in primary lymphocytes isolated from normolipemic individuals and
patients with homozygous FH who totally lack LDLR function (Chemello et al.
2020). Noteworthy, LDLR expression in human primary lymphocytes positively
and significantly correlates with individuals’ LDL-C, but not with Lp(a) plasma
concentrations (Thedrez et al. 2018).
Studies conducted in mice, rabbits, or nonhuman primates have also yielded
opposite conclusions regarding the role of the LDLR and the effects of PCSK9
inhibitors on Lp(a) catabolism. Thus, compared with wild-type animals, mice
overexpressing the LDLR display accelerated Lp(a) plasma clearance (Hofmann
et al. 1990), but LDLR knockout mice have similar Lp(a) clearance than wild-type
animals (Cain et al. 2005). Furthermore, the catabolism of Lp(a) in rabbits is
slower than that of LDL, suggesting that Lp(a) uptake is not fully dependent on the
LDLR and may be mediated by other mechanisms (Liu et al. 1993). In addition,
alirocumab did not alter the catabolic rate of Lp(a) but was found to enhance Lp(a)
production in nonhuman primates (Croyal et al. 2018). Likewise, alirocumab had
no effect on the hepatic capture of Lp(a) in liver-humanized mice (Chemello
et al. 2020).
Studies of FH and non-FH siblings with identical apo(a) isoforms have clearly
demonstrated that Lp(a) is approximately twice higher in FH patients than in nonaf-
fected family members (Lingenhel et al. 1998). Homozygous FH subjects with two
nonfunctional LDLR alleles also display twofold higher Lp(a) levels than their
6 Role of Proprotein Convertase Subtilisin Kexin Type 9 in Lipoprotein(a) Metabolism 119
Conclusion
The fact that unlike statins PCSK9 inhibitors reduce Lp(a) has clearly raised interest
in deciphering the molecular mechanisms by which this may occur. Despite much
effort, there is no consensus at present indicating that the lowering of Lp(a) induced
by PCSK9 inhibitors directly results from the reduction in LDLR expression and
function, as is the case for LDL. Figure 6.1 summarizes the potential pathways by
which PCSK9 (and hence PCSK9 inhibitors) modulates Lp(a) plasma concentra-
tions. For instance, the LDLR may play some role in mediating Lp(a) clearance
when its expression is starkly upregulated (e.g., by concomitant use of statins and
PCSK9 inhibitors) and when LDL plasma levels are substantially reduced, allowing
decreased competition between LDL and Lp(a) for receptor-mediated uptake. In
addition to an effect of PCSK9 on Lp(a) plasma clearance, the latest in vitro evi-
dence points toward a direct role for PCSK9 in enhancing Lp(a) production, assem-
bly, and subsequent secretion from liver cells (Youssef et al. 2022; Villard et al.
2016) (Fig. 6.1). Further exciting research is now needed to extensively explore this
fascinating observation in vivo.
References
Chemello K, Beeské S, Trang Tran TT, Blanchard V, Villard EF, Poirier B, Le Bail J-C, Dargazanli
G, Ho-Van-Guimbal S, Boulay D, Bergis O, Pruniaux M-P, Croyal M, Janiak P, Guillot E,
Lambert G. Lipoprotein(a) cellular uptake ex vivo and hepatic capture in vivo is insensitive to
PCSK9 inhibition with alirocumab. JACC Basic Transl Sci. 2020;5:549–57.
Cohen JC, Boerwinkle E, Mosley TH, Hobbs HH. Sequence variations in PCSK9, low LDL, and
protection against coronary heart disease. N Engl J Med. 2006;354:1264–72.
Croyal M, Tran T-T-T, Blanchard RH, Le Bail J-C, Villard EF, Poirier B, Aguesse A, Billon-
Crossouard S, Ramin-Mangata S, Blanchard V, Nativel B, Chemello K, Khantalin I, Thedrez
A, Janiak P, Krempf M, Boixel C, Lambert G, Guillot E. PCSK9 inhibition with alirocumab
reduces lipoprotein(a) levels in nonhuman primates by lowering apolipoprotein(a) production
rate. Clin Sci (Lond). 2018;132:1075–83.
Edmiston JB, Brooks N, Tavori H, Minnier J, Duell B, Purnell JQ, Kaufman T, Wojcik C, Voros
S, Fazio S, Shapiro MD. Discordant response of low-density lipoprotein cholesterol and
lipoprotein(a) levels to monoclonal antibodies targeting proprotein convertase subtilisin/kexin
type 9. J Clin Lipidol. 2017;11:667–73.
Gabel BR, Koschinsky ML. Sequences within apolipoprotein(a) kringle IV types 6-8 bind directly
to low-density lipoprotein and mediate noncovalent association of apolipoprotein(a) with apo-
lipoprotein B-100. Biochemistry. 1998;37:7892–8.
Gaudet D, Kereiakes DJ, McKenney JM, Roth EM, Hanotin C, Gipe D, Du Y, Ferrand A-C,
Ginsberg HN, Stein EA. Effect of alirocumab, a monoclonal proprotein convertase subtilisin/
kexin 9 antibody, on lipoprotein(a) concentrations (a pooled analysis of 150 mg every two
weeks dosing from phase 2 trials). Am J Cardiol. 2014;114:711–5.
Gaudet D, Watts GF, Robinson JG, Minini P, Sasiela WJ, Edelberg J, Louie MJ, Raal FJ. Effect of
alirocumab on lipoprotein(a) over ≥1.5 years (from the Phase 3 ODYSSEY Program). Am J
Cardiol. 2017;119:40–6.
Hofer G, Steyrer E, Kostner GM, Hermetter A. LDL-mediated interaction of Lp[a] with HepG2
cells: a novel fluorescence microscopy approach. J Lipid Res. 1997;38:2411–21.
Hofmann SL, Eaton DL, Brown MS, McConathy WJ, Goldstein JL, Hammer RE. Overexpression
of human low density lipoprotein receptors leads to accelerated catabolism of Lp(a) lipoprotein
in transgenic mice. J Clin Invest. 1990;85:1542–7.
Koschinsky ML, Beisiegel U, Henne-Bruns D, Eaton DL, Lawn RM. Apolipoprotein(a) size
heterogeneity is related to variable number of repeat sequences in its mRNA. Biochemistry.
1990;29:640–4.
Kraft HG, Menzel HJ, Hoppichler F, Vogel W, Utermann G. Changes of genetic apolipoprotein
phenotypes caused by liver transplantation. Implications for apolipoprotein synthesis. J Clin
Invest. 1989;83:137–42.
Kraft HG, Lingenhel A, Raal FJ, Hohenegger M, Utermann G. Lipoprotein(a) in homozygous
familial hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2000;20:522–8.
Krempler F, Kostner G, Bolzano K, Sandhofer F. Lipoprotein (a) is not a metabolic product of
other lipoproteins containing apolipoprotein B. Biochim Biophys Acta. 1979;575:63–70.
Kronenberg F. Lipoprotein(a) in various conditions: to keep a sense of proportions. Atherosclerosis.
2014;234:249–51.
Kronenberg F, Utermann G. Lipoprotein(a): resurrected by genetics. J Intern Med. 2013;273:6–30.
Lagace TA, Curtis DE, Garuti R, McNutt MC, Park SW, Prather HB, Anderson NN, Ho YK,
Hammer RE, Horton JD. Secreted PCSK9 decreases the number of LDL receptors in hepato-
cytes and in livers of parabiotic mice. J Clin Invest. 2006;116:2995–3005.
Lambert G, Sjouke B, Choque B, Kastelein JJP, Hovingh GK. The PCSK9 decade. J Lipid Res.
2012;53:2515–24.
Lambert G, Thedrez A, Croyal M, Ramin-Mangata S, Couret D, Diotel N, Nobécourt-Dupuy E,
Krempf M, LeBail JC, Poirier B, Blankenstein J, Villard EF, Guillot E. The complexity of
lipoprotein (a) lowering by PCSK9 monoclonal antibodies. Clin Sci (Lond). 2017;131:261–8.
Langsted A, Kamstrup PR, Benn M, Tybjærg-Hansen A, Nordestgaard BG. High lipoprotein(a) as
a possible cause of clinical familial hypercholesterolaemia: a prospective cohort study. Lancet
Diabetes Endocrinol. 2016;4:577–87.
122 A. Gallo et al.
Li J, Tumanut C, Gavigan J-A, Huang W-J, Hampton EN, Tumanut R, Suen KF, Trauger JW,
Spraggon G, Lesley SA, Liau G, Yowe D, Harris JL. Secreted PCSK9 promotes LDL receptor
degradation independently of proteolytic activity. Biochem J. 2007;406:203–7.
Lingenhel A, Kraft HG, Kotze M, Peeters AV, Kronenberg F, Kruse R, Utermann G. Concentrations
of the atherogenic Lp(a) are elevated in FH. Eur J Hum Genet. 1998;6:50–60.
Liu R, Saku K, Kostner GM, Hirata K, Zhang B, Shiomi M, Arakawa K. In vivo kinetics of
lipoprotein(a) in homozygous Watanabe heritable hyperlipidaemic rabbits. Eur J Clin Investig.
1993;23:561–5.
Mahmood T, Minnier J, Ito MK, Li QH, Koren A, Kam IW, Fazio S, Shapiro MD. Discordant
responses of plasma low-density lipoprotein cholesterol and lipoprotein(a) to alirocumab: a
pooled analysis from 10 ODYSSEY Phase 3 studies. Eur J Prev Cardiol. 2021;28:816–22.
Maxwell KN, Breslow JL. Adenoviral-mediated expression of Pcsk9 in mice results in a low-
density lipoprotein receptor knockout phenotype. Proc Natl Acad Sci U S A. 2004;101:7100–5.
McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors.
Pathology. 2019;51:155–64.
McNutt MC, Lagace TA, Horton JD. Catalytic activity is not required for secreted PCSK9 to
reduce low density lipoprotein receptors in HepG2 cells. J Biol Chem. 2007;282:20799–803.
Nassoury N, Blasiole DA, Tebon Oler A, Benjannet S, Hamelin J, Poupon V, McPherson PS, Attie
AD, Prat A, Seidah NG. The cellular trafficking of the secretory proprotein convertase PCSK9
and its dependence on the LDLR. Traffic. 2007;8:718–32.
Niemeier A, Willnow T, Dieplinger H, Jacobsen C, Meyer N, Hilpert J, Beisiegel U. Identification
of megalin/gp330 as a receptor for lipoprotein(a) in vitro. Arterioscler Thromb Vasc Biol.
1999;19:552–61.
O’Donoghue ML, Fazio S, Giugliano RP, Stroes ESG, Kanevsky E, Gouni-Berthold I, Im K, Lira
Pineda A, Wasserman SM, Češka R, Ezhov MV, Jukema JW, Jensen HK, Tokgözoğlu SL,
Mach F, Huber K, Sever PS, Keech AC, Pedersen TR, Sabatine MS. Lipoprotein(a), PCSK9
inhibition, and cardiovascular risk. Circulation. 2019;139:1483–92.
Qian Y-W, Schmidt RJ, Zhang Y, Chu S, Lin A, Wang H, Wang X, Beyer TP, Bensch WR, Li W,
Ehsani ME, Lu D, Konrad RJ, Eacho PI, Moller DE, Karathanasis SK, Cao G. Secreted PCSK9
downregulates low density lipoprotein receptor through receptor-mediated endocytosis. J Lipid
Res. 2007;48:1488–98.
Raal FJ, Giugliano RP, Sabatine MS, Koren MJ, Blom D, Seidah NG, Honarpour N, Lira A, Xue
A, Chiruvolu P, Jackson S, Di M, Peach M, Somaratne R, Wasserman SM, Scott R, Stein
EA. PCSK9 inhibition-mediated reduction in Lp(a) with evolocumab: an analysis of 10 clinical
trials and the LDL receptor’s role. J Lipid Res. 2016;57:1086–96.
Raal FJ, Kallend D, Ray KK, Turner T, Koenig W, Wright RS, Wijngaard PLJ, Curcio D, Jaros MJ,
Leiter LA, Kastelein JJP, ORION-9 Investigators. Inclisiran for the treatment of heterozygous
familial hypercholesterolemia. N Engl J Med. 2020;382:1520–30.
Rader DJ, Mann WA, Cain W, Kraft HG, Usher D, Zech LA, Hoeg JM, Davignon J, Lupien P,
Grossman M. The low density lipoprotein receptor is not required for normal catabolism of
Lp(a) in humans. J Clin Invest. 1995;95:1403–8.
Ray KK, Vallejo-Vaz AJ, Ginsberg HN, Davidson MH, Louie MJ, Bujas-Bobanovic M, Minini
P, Eckel RH, Cannon CP. Lipoprotein(a) reductions from PCSK9 inhibition and major
adverse cardiovascular events: pooled analysis of alirocumab phase 3 trials. Atherosclerosis.
2019;288:194–202.
Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, Bisch JA, Richardson T, Jaros M,
Wijngaard PLJ, Kastelein JJP, ORION-10 and ORION-11 Investigators. Two phase 3 trials of
inclisiran in patients with elevated LDL cholesterol. N Engl J Med. 2020;382:1507–19.
Reblin T, Niemeier A, Meyer N, Willnow TE, Kronenberg F, Dieplinger H, Greten H, Beisiegel
U. Cellular uptake of lipoprotein[a] by mouse embryonic fibroblasts via the LDL receptor and
the LDL receptor-related protein. J Lipid Res. 1997;38:2103–10.
Reyes-Soffer G, Pavlyha M, Ngai C, Thomas T, Holleran S, Ramakrishnan R, Karmally W,
Nandakumar R, Fontanez N, Obunike J, Marcovina SM, Lichtenstein AH, Matthan NR, Matta
6 Role of Proprotein Convertase Subtilisin Kexin Type 9 in Lipoprotein(a) Metabolism 123
J, Maroccia M, Becue F, Poitiers F, Swanson B, Cowan L, Sasiela WJ, Surks HK, Ginsberg
HN. Effects of PCSK9 inhibition with alirocumab on lipoprotein metabolism in healthy
humans. Circulation. 2017;135:352–62.
Robinson JG, Farnier M, Krempf M, Bergeron J, Luc G, Averna M, Stroes ES, Langslet G, Raal
FJ, El Shahawy M, Koren MJ, Lepor NE, Lorenzato C, Pordy R, Chaudhari U, Kastelein JJP,
ODYSSEY LONG TERM Investigators. Efficacy and safety of alirocumab in reducing lipids
and cardiovascular events. N Engl J Med. 2015;372:1489–99.
Romagnuolo R, Scipione CA, Boffa MB, Marcovina SM, Seidah NG, Koschinsky
ML. Lipoprotein(a) catabolism is regulated by proprotein convertase subtilisin/kexin type 9
through the low density lipoprotein receptor. J Biol Chem. 2015;290:11649–62.
Romagnuolo R, Scipione CA, Marcovina SM, Gemin M, Seidah NG, Boffa MB, Koschinsky
ML. Roles of the low density lipoprotein receptor and related receptors in inhibition of
lipoprotein(a) internalization by proprotein convertase subtilisin/kexin type 9. PLoS One.
2017;12:e0180869.
Sabatine MS, Giugliano RP, Wiviott SD, Raal FJ, Blom DJ, Robinson J, Ballantyne CM,
Somaratne R, Legg J, Wasserman SM, Scott R, Koren MJ, Stein EA, Open-Label Study of
Long-Term Evaluation against LDL Cholesterol (OSLER) Investigators. Efficacy and safety
of evolocumab in reducing lipids and cardiovascular events. N Engl J Med. 2015;372:1500–9.
Seidah NG, Awan Z, Chrétien M, Mbikay M. PCSK9: a key modulator of cardiovascular health.
Circ Res. 2014;114:1022–36.
Shapiro MD, Minnier J, Tavori H, Kassahun H, Flower A, Somaratne R, Fazio S. Relationship
between low-density lipoprotein cholesterol and lipoprotein(a) lowering in response to PCSK9
inhibition with evolocumab. J Am Heart Assoc. 2019;8:e010932.
Sharma M, Redpath GM, Williams MJA, McCormick SPA. Recycling of apolipoprotein(a) after
PlgRKT-mediated endocytosis of lipoprotein(a). Circ Res. 2017;120:1091–102.
Snyder ML, Polacek D, Scanu AM, Fless GM. Comparative binding and degradation of
lipoprotein(a) and low density lipoprotein by human monocyte-derived macrophages. J Biol
Chem. 1992;267:339–46.
Stein EA, Giugliano RP, Koren MJ, Raal FJ, Roth EM, Weiss R, Sullivan D, Wasserman SM,
Somaratne R, Kim JB, Yang J, Liu T, Albizem M, Scott R, Sabatine MS, Investigators
PROFICIO. Efficacy and safety of evolocumab (AMG 145), a fully human monoclonal anti-
body to PCSK9, in hyperlipidaemic patients on various background lipid therapies: pooled
analysis of 1359 patients in four phase 2 trials. Eur Heart J. 2014;35:2249–59.
Steyrer E, Kostner GM. Interaction of lipoprotein Lp[a] with the B/E-receptor: a study using iso-
lated bovine adrenal cortex and human fibroblast receptors. J Lipid Res. 1990;31:1247–53.
Tada H, Kawashiri M-A, Yoshida T, Teramoto R, Nohara A, Konno T, Inazu A, Mabuchi H,
Yamagishi M, Hayashi K. Lipoprotein(a) in familial hypercholesterolemia with proprotein
convertase subtilisin/kexin type 9 (PCSK9) gain-of-function mutations. Circ J. 2016;80:
512–8.
Thedrez A, Blom DJ, Ramin-Mangata S, Blanchard V, Croyal M, Chemello K, Nativel B, Pichelin
M, Cariou B, Bourane S, Tang L, Farnier M, Raal FJ, Lambert G. Homozygous FH patients
with identical mutations variably express the LDL receptor: implications for the efficacy of
evolocumab. Arterioscler Thromb Vasc Biol. 2018;38:592–8.
Trinder M, DeCastro ML, Azizi H, Cermakova L, Jackson LM, Frohlich J, Mancini GBJ, Francis
GA, Brunham LR. Ascertainment bias in the association between elevated lipoprotein(a) and
familial hypercholesterolemia. J Am Coll Cardiol. 2020;75:2682–93.
van der Hoek YY, Lingenhel A, Kraft HG, Defesche JC, Kastelein JJ, Utermann G. Sib-pair analy-
sis detects elevated Lp(a) levels and large variation of Lp(a) concentration in subjects with
familial defective ApoB. J Clin Invest. 1997;99:2269–73.
Villard EF, Thedrez A, Blankenstein J, Croyal M, Tran T-T-T, Poirier B, Le Bail J-C, Illiano S,
Nobécourt E, Krempf M, Blom DJ, Marais AD, Janiak P, Muslin AJ, Guillot E, Lambert
G. PCSK9 modulates the secretion but not the cellular uptake of lipoprotein(a) ex vivo: an
effect blunted by alirocumab. JACC Basic Transl Sci. 2016;1:419–27.
124 A. Gallo et al.
Watts GF, Chan DC, Somaratne R, Wasserman SM, Scott R, Marcovina SM, Barrett PHR. Controlled
study of the effect of proprotein convertase subtilisin-kexin type 9 inhibition with evolocumab
on lipoprotein(a) particle kinetics. Eur Heart J. 2018;39:2577–85.
Watts GF, Chan DC, Pang J, Ma L, Ying Q, Aggarwal S, Marcovina SM, Barrett PHR. PCSK9
Inhibition with alirocumab increases the catabolism of lipoprotein(a) particles in statin-treated
patients with elevated lipoprotein(a). Metabolism. 2020;107:154221.
White AL, Guerra B, Wang J, Lanford RE. Presecretory degradation of apolipoprotein [a] is medi-
ated by the proteasome pathway. J Lipid Res. 1999;40:275–86.
Yang X-P, Amar MJ, Vaisman B, Bocharov AV, Vishnyakova TG, Freeman LA, Kurlander RJ,
Patterson AP, Becker LC, Remaley AT. Scavenger receptor-BI is a receptor for lipoprotein(a).
J Lipid Res. 2013;54:2450–7.
Youssef A, Clark JR, Marcovina SM, Boffa MB, Koschinsky ML. ApoA and ApoB interact non-
covalently within hepatocytes: implications for regulation of Lp(a) (lipoprotein[a]) levels by
modulation of ApoB secretion. Arterioscler Thromb Vasc Biol. 2022;42:289–304.
Zhao Z, Tuakli-Wosornu Y, Lagace TA, Kinch L, Grishin NV, Horton JD, Cohen JC, Hobbs
HH. Molecular characterization of loss-of-function mutations in PCSK9 and identification of a
compound heterozygote. Am J Hum Genet. 2006;79:514–23.
Chapter 7
The Role of Cell Surface Receptors
in Lp(a) Catabolism
Introduction
Lamia Ismail and Déanna Shea contributed equally with all other contributors.
S-S
P LBP LPB
3
KIV
KV
LPA ORM2
KIV 10
4
KIV
KIV9
5
PLA2G7 PLA2G7
KIV
KIV 8
KIV7
6
KIV
PON1 SAA4
SERPINA1
Fig. 7.1 Lp(a) structure. Lp(a) consists of an LDL molecule attached to apo(a) via a disulphide
link to apoB. Oxidised phospholipids (OxPL) are found bound to the apo(a) KV domain and con-
tained in the phospholipid (PL) monolayer on the LDL surface. A number of lipid metabolism
proteins (green) are associated with the LDL particle, as are many wound-healing proteins (blue),
some of which may be bound to apo(a). The proteins are designated by their gene names.
(Reproduced from McCormick and Schneider 2019 with permission)
secreted. This was first shown by studies in primary hepatocytes with different-
sized isoforms (White et al. 1997; Brunner et al. 1996) and is supported by recent
studies of an apo(a) mutant that displayed defective glycosylation and folding
(Morgan et al. 2020). These observations underpin the well-known inverse relation-
ship between Lp(a) levels and isoform size (Kraft et al. 1996).
The location of Lp(a) assembly is not certain but there is significant evidence for
an extracellular assembly with LDL (on the surface of hepatocytes or in circulation)
after apo(a) secretion (Chiesa et al. 1992; White and Lanford 1994). The process of
assembly involves an initial noncovalent binding between apolipoprotein B (apoB)
lysine residues exposed on the LDL surface and lysine-binding sites in apo(a)
(Gabel and Koschinsky 1998; Becker et al. 2004). This is followed by the formation
of a disulphide bond between specific cysteine residues in apo(a) and apoB
(Koschinsky et al. 1993; McCormick et al. 1995).
Lp(a) Catabolism
The liver provides the principal route of Lp(a) clearance from the circulation (Cain
et al. 2005). However, the presence of apo(a) fragments in the urine of patients with
kidney disease (Kostner et al. 1996; Albers et al. 2007) and their reduction after
transplantation (Black and Wilcken 1992) also suggest a role for the kidney. In mac-
rophages, it has been documented that Lp(a) binds to and triggers the CD36/TLR2
apoptotic pathway (Seimon et al. 2010).
Unlike LDL, which has a clearly defined uptake pathway via the low-density
lipoprotein receptor (LDLR) (Brown and Goldstein 1986) that can be specifically
targeted by drugs, i.e. statins and proprotein convertase subtilisin/kexin type 9
(PCSK9) inhibitors, the clearance of Lp(a) is rather more complicated. Many recep-
tors including lipoprotein, plasminogen, lectin and scavenger receptors have been
documented as being involved in Lp(a) catabolism (Hoover-Plow and Huang 2013;
McCormick and Schneider 2019). Lp(a)’s association with multiple receptors is
likely because it has a more complex composition than LDL with more potential
ligands (Fig. 7.1); apo(a), apoB and OxPL elements of Lp(a) have been shown to be
ligands for the various Lp(a) receptors with apolipoprotein E (apoE), orosomucoid
and alpha-2-macroglobulin also reported as possible ligands (McCormick and
Schneider 2019).
Liver Receptors
There are multiple receptors that have been shown to interact with Lp(a). Figure 7.2
represents the receptors that have been reported as being involved in Lp(a) uptake
that are expressed in the liver and kidney as well as receptors expressed in macro-
phages which promote cell signalling events from Lp(a). Figure 7.3 shows the gene
128 L. Ismail et al.
Fig. 7.2 Cell surface receptors for Lp(a). Receptors expressed in the liver and kidney for which
there is evidence of a role in binding to and promoting Lp(a) uptake are shown. These include the
lipoprotein receptors, LDLR, VLDLR, LRP-1 and megalin; plasminogen receptors, annexin A2,
S100A10 and PlgRKT; lectin receptors, galectin-1 and ASGR1; and the scavenger receptor,
SR-B1. Also shown are receptors on macrophages which mediate cell signalling events via Lp(a).
These include the scavenger receptor; CD36; and toll-like receptors, TLR2 and TLR6
expression profiles of the Lp(a) receptors in both liver and kidney. Most attention
has been paid to the LDLR with early cell culture studies showing Lp(a) to bind to
fibroblasts via the LDLR (Havekes et al. 1981; Floren et al. 1981) and subsequent
hepatocytes studies showing the same (Romagnuolo et al. 2015, 2017). These find-
ings were well supported by a study on fibroblasts from familial hypercholesterol-
aemia (FH) patients with defective LDL receptors which showed a much-reduced
binding to Lp(a) (Krempler et al. 1983). In the same study, kinetic experiments also
showed a delayed clearance of Lp(a) in FH subjects (Krempler et al. 1983). However,
there are many studies countering these findings including a knockout of the LDLR
in mice which showed no difference in Lp(a) clearance compared to wild-type mice
(Cain et al. 2005) and kinetics studies indicating no difference in Lp(a) catabolism
between FH and non-FH subjects (Rader et al. 1995; Knight 1994). Most impor-
tantly, there is the conundrum that statins do not lower Lp(a) and may increase it
(Yeang et al. 2016) and that PCSK9 inhibitors, which also upregulate LDLR, have
no effect on Lp(a) catabolism (Chemello et al. 2020).
One other LDLR family member that is expressed in the liver (Fig. 7.3) and has
been shown to interact with Lp(a) is low-density lipoprotein receptor-related protein
7 The Role of Cell Surface Receptors in Lp(a) Catabolism 129
Liver Kidney
150 150
144
60 60
50 50
40 40
RPKM
RPKM
30 30
20 20
10 10
0 0
LR 1
AS S1
1
B1
P1
AS 1
1
B1
PL 10
VL R
R
P2
S1 A2
PL A10
S1 A2
P2
LR R
KT
KT
VL R
P
GR
GR
S
DL
L
DL
L
A
AR
AL
LR
AL
AR
X
LR
X
LD
GR
GR
LD
00
00
AN
AN
LG
LG
SC
SC
Plasminogen Lipoprotein Lectin Scavenger Plasminogen Lipoprotein Lectin Scavenger
Fig. 7.3 Relative gene expression of Lp(a) receptors. The gene expression data for each of the
receptors in the liver and kidney are shown based on reads per kilobase of transcript per million
reads mapped (RPKM). Gene expression data were taken from NCBI (https://www.ncbi.nlm.
nih.gov/)
enhanced Lp(a) uptake significantly in both cell lines (Sharma et al. 2017).
Furthermore, a much-diminished internalisation of Lp(a) was observed in PlgRKT
knockdown HepG2 cells and in PlgRKT−/− fibroblast cells (Sharma et al. 2017).
Whether PlgRKT is a bona fide receptor for Lp(a) or whether it enhances the sur-
face binding of apo(a) to allow for uptake by other receptors, or via macropinocyto-
sis, is not yet known.
Another receptor highly expressed by the liver (Fig. 7.3) for which evidence is
mounting for a role in Lp(a) catabolism is scavenger receptor B1 (SR-B1). Well
known for its role in high-density lipoprotein (HDL) metabolism by facilitating
selective uptake of cholesterol esters (Acton et al. 1996), it has also been shown to
mediate whole particle uptake of very-low-density lipoprotein (VLDL), LDL and
HDL (Wang et al. 1998; Zanoni et al. 2018). Evidence for SR-B1’s involvement in
Lp(a) catabolism has come from transgenic mouse models in which overexpression
of SR-B1 significantly increases Lp(a) uptake, and contrariwise, SR-B1 knockout
mice show a reduced Lp(a) clearance (Yang et al. 2013). Another study showed that
SR-B1 facilitated the uptake of OxPL from Lp(a) in liver cells (Sharma et al. 2015).
Interestingly, individuals harbouring a mutation that reduces the ability of SR-B1 to
facilitate lipid uptake display both elevated HDL and Lp(a) levels (Yang et al. 2016)
suggesting clinical relevance.
One further receptor which is highly expressed in the liver (Fig. 7.3) shown to
mediate Lp(a) uptake is the asialoglycoprotein receptor (ASGPR), a lectin receptor
that mediates endocytosis of desialylated glycoproteins (Igdoura 2017). Mice lack-
ing the ASGPR showed a much-reduced clearance and degradation of Lp(a) by the
liver compared to wild-type mice (Hrzenjak et al. 2003). As apo(a) has a significant
content of desialylated O-linked sugars, it is a likely ligand for ASGPR. Indeed,
removal of sialic acids from Lp(a) greatly enhanced the clearance rate providing
support for an apo(a)/ASGPR interaction (Hrzenjak et al. 2003). Lastly, a lectin
receptor, galectin-1, highly expressed in the liver (Fig. 7.3), has been shown to bind
to Lp(a) (Chellan et al. 2007).
Kidney Receptors
The kidney expresses many of the same receptors as the liver (Fig. 7.2) including
the plasminogen receptors and LDLR. Two other members of the LDLR family
expressed in the kidney which have been shown to bind Lp(a) are the VLDL recep-
tor and the megalin receptor (otherwise known as LRP2). The VLDLR has been
shown to promote endocytosis and degradation of Lp(a) in fibroblasts with apo(a)
mediating the binding (Argraves et al. 1997). Furthermore, mice lacking the VLDLR
showed delayed clearance of Lp(a) (Argraves et al. 1997). The megalin receptor is
highly expressed in the kidney and plays a role in nutrient uptake via many different
ligands (Christensen and Birn 2001). It has been shown to bind to Lp(a) in a yolk
sac cell line via its apoB component (Niemeier et al. 1999).
7 The Role of Cell Surface Receptors in Lp(a) Catabolism 131
Macrophage Receptors
It is well documented that Lp(a) promotes inflammation via many different signal-
ling pathways through its OxPL content (Tsimikas and Hall 2012; Van der Valk
et al. 2016). Macrophages express an array of receptors which bind OxPL and stim-
ulate inflammatory signalling pathways and immune responses (Taylor et al. 2005).
These include the TLRs, which often work as co-receptors in conjunction with the
CD36 scavenger receptor to sense ligands. The TLR2/TLR6 heterodimer along with
CD36 (Fig. 7.2) has been shown to interact with OxPLs on apo(a) to promote
inflammation and apoptosis of macrophages (Seimon et al. 2010). Another study
indicated that both TLR2 and CD36 were necessary for the ability of Lp(a) to pro-
mote IL-8 production from macrophages (Scipione et al. 2015). These studies indi-
cate that the TLRs mediate the signalling promoting properties of Lp(a).
From this array of possible Lp(a) receptors, it is difficult to speculate which
receptors might contribute the most to Lp(a) clearance in the physiological setting
of the human body. If one considers Lp(a) clearance from a tissue aspect, then evi-
dence suggests that the focus should be on receptors that are highly expressed in the
liver. If one considers Lp(a) clearance from a ligand aspect, then the focus might be
best placed on receptors that specifically interact with apo(a) since it is a ligand
unique to Lp(a).
Summary
The Lp(a) molecule has a complex structure with several of its components known
to be ligands for various receptors. Cell culture studies have shown Lp(a) to bind to
a diverse range of cell surface receptors on multiple cell types. This situation makes
it difficult to pinpoint any one receptor pathway as being important in Lp(a) catabo-
lism and currently precludes targeting Lp(a) catabolism as a route for Lp(a) lower-
ing. Future studies will require a careful teasing out of the roles of the different
Lp(a) receptors in the liver and kidney with an emphasis on in vivo studies to gauge
clinical relevance.DisclosureNothing to disclose for all authors.
References
Becker L, Cook PM, Wright TG, Koschinsky ML. Quantitative evaluation of the contribution of
weak lysine-binding sites present within apolipoprotein(a) kringle IV types 6-8 to lipoprotein(a)
assembly. J Biol Chem. 2004;279(4):2679–88.
Beisiegel U, Weber W, Ihrke G, Herz J, Stanley KK. The LDL-receptor-related protein, LRP, is an
apolipoprotein E-binding protein. Nature. 1989;341(6238):162–4.
Berg K. A new serum type system in man—the Lp system. Acta Pathol Microbiol Scand.
1963;59(3):369–82.
Bharadwaj A, Kempster E, Waisman DM. The Annexin A2/S100A10 complex: the mutualistic
symbiosis of two distinct proteins. Biomol. 2021;11(12):1849.
Black IW, Wilcken DE. Decreases in apolipoprotein(a) after renal transplantation: implications for
lipoprotein(a) metabolism. Clin Chem. 1992;38(3):353–7.
Brown MS, Goldstein JL. A receptor-mediated pathway for cholesterol homeostasis. Science.
1986;232(4746):34–47.
Brunner C, Lobentanz EM, Petho-Schramm A, Ernst A, Kang C, Dieplinger H, et al. The num-
ber of identical kringle IV repeats in apolipoprotein(a) affects its processing and secretion by
HepG2 cells. J Biol Chem. 1996;271(50):32403–10.
Cain WJ, Millar JS, Himebauch AS, Tietge UJ, Maugeais C, Usher D, et al. Lipoprotein [a] is
cleared from the plasma primarily by the liver in a process mediated by apolipoprotein [a]. J
Lipid Res. 2005;46(12):2681–91.
Chellan B, Narayani J, Appukuttan PS. Galectin-1, an endogenous lectin produced by arte-
rial cells, binds lipoprotein(a) [Lp(a)] in situ: relevance to atherogenesis. Exp Mol Pathol.
2007;83(3):399–404.
Chemello K, Beeské S, Trang Tran TT, Blanchard V, Villard EF, Poirier B, et al. Lipoprotein(a)
cellular uptake ex vivo and hepatic capture in vivo is insensitive to PCSK9 inhibition with
alirocumab. JACC Basic Transl Sci. 2020;5(6):549–57.
Chiesa G, Hobbs HH, Koschinsky ML, Lawn RM, Maika SD, Hammer RE. Reconstitution of
lipoprotein(a) by infusion of human low density lipoprotein into transgenic mice expressing
human apolipoprotein(a). J Biol Chem. 1992;267(34):24369–74.
Christensen EI, Birn H. Megalin and cubilin: synergistic endocytic receptors in renal proximal
tubule. Am J Physiol Renal Physiol. 2001;280(4):F562–73.
Coassin S, Erhart G, Weissensteiner H, Guimarães E, de Araújo M, Lamina C, Schönherr S, et al.
A novel but frequent variant in LPA KIV-2 is associated with a pronounced Lp(a) and cardio-
vascular risk reduction. Eur Heart J. 2017;38(23):1823–31.
Floren CH, Albers JJ, Bierman EL. Uptake of Lp(a) lipoprotein by cultured fibroblasts. Biochem
Biophys Res Commun. 1981;102(2):636–9.
Gabel BR, Koschinsky ML. Sequences within apolipoprotein(a) kringle IV types 6-8 bind directly
to low-density lipoprotein and mediate noncovalent association of apolipoprotein(a) with apo-
lipoprotein B-100. Biochemistry. 1998;37(21):7892–8.
Hajjar KA, Krishnan S. Annexin II: a mediator of the plasmin/plasminogen activator system.
Trends Cardiovasc Med. 1999;9(5):128–38.
Havekes L, Vermeer BJ, Brugman T, Emeis J. Binding of Lp(a) to the low density lipoprotein
receptor of human fibroblasts. FEBS Lett. 1981;132(2):169–73.
Hoover-Plow J, Huang M. Lipoprotein(a) metabolism: potential sites for therapeutic targets.
Metabolism. 2013;62(4):479–91.
Hrzenjak A, Frank S, Wo X, Zhou Y, Van Berkel T, Kostner GM. Galactose-specific asialogly-
coprotein receptor is involved in lipoprotein(a) catabolism. Biochem J. 2003;376(3):765–71.
Igdoura SA. Asialoglycoprotein receptors as important mediators of plasma lipids and atheroscle-
rosis. Curr Opin Lipidol. 2017;28(2):209–12.
Knight BL. Lp(a) catabolism in hypercholesterolaemic individuals. Chem Phys Lipids.
1994;67-68:233–9.
Koschinsky ML, Cote GP, Gabel B, van der Hoek YY. Identification of the cysteine residue in
apolipoprotein(a) that mediates extracellular coupling with apolipoprotein B-100. J Biol Chem.
1993;268(26):19819–25.
7 The Role of Cell Surface Receptors in Lp(a) Catabolism 133
Kostner KM, Maurer G, Huber K, Stefenelli T, Dieplinger H, Steyrer E, et al. Urinary excre-
tion of apo(a) fragments. Role in apo(a) catabolism. Arterioscler Thromb Vasc Biol.
1996;16(8):905–11.
Kostner KM, Marz W, Kostner GM. When should we measure lipoprotein (a)? Eur Heart
J. 2013;34(42):3268–76.
Kraft HG, Lingenhel A, Pang RW, Delport R, Trommsdorff M, Vermaak H, et al. Frequency distri-
butions of apolipoprotein(a) kringle IV repeat alleles and their effects on lipoprotein(a) levels
in Caucasian, Asian, and African populations: the distribution of null alleles is non-random.
Eur J Hum Genet. 1996;4(2):74–87.
Krempler F, Kostner GM, Roscher A, Haslauer F, Bolzano K, Sandhofer F. Studies on the role
of specific cell surface receptors in the removal of lipoprotein (a) in man. J Clin Invest.
1983;71(5):1431–41.
Kruth HS, Jones NL, Huang W, Zhao B, Ishii I, Chang J, et al. Macropinocytosis is the endocytic
pathway that mediates macrophage foam cell formation with native low density lipoprotein. J
Biol Chem. 2005;280(3):2352–60.
Leibundgut G, Scipione C, Yin H, Schneider M, Boffa MB, Green S, et al. Determinants of
binding of oxidized phospholipids on apolipoprotein (a) and lipoprotein (a). J Lipid Res.
2013;54(10):2815–30.
Marz W, Beckmann A, Scharnagl H, Siekmeier R, Mondorf U, Held I, et al. Heterogeneous lipo-
protein (a) size isoforms differ by their interaction with the low density lipoprotein receptor
and the low density lipoprotein receptor-related protein/alpha 2-macroglobulin receptor. FEBS
Lett. 1993;325(3):271–5.
McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors.
Pathology. 2019;51(2):155–64.
McCormick SP, Ng JK, Taylor S, Flynn LM, Hammer RE, Young SG. Mutagenesis of the human
apolipoprotein B gene in a yeast artificial chromosome reveals the site of attachment for
apolipoprotein(a). Proc Natl Acad Sci U S A. 1995;92(22):10147–51.
McLean JW, Tomlinson JE, Kuang WJ, Eaton DL, Chen EY, Fless GM, et al. cDNA sequence of
human apolipoprotein(a) is homologous to plasminogen. Nature. 1987;330(6144):132–7.
Morgan BM, Brown AN, Deo N, Harrop TWR, Taiaroa G, Mace PD, et al. Nonsynonymous SNPs
in LPA homologous to plasminogen deficiency mutants represent novel null apo(a) alleles. J
Lipid Res. 2020;61(3):432–44.
Niemeier A, Willnow T, Dieplinger H, Jacobsen C, Meyer N, Hilpert J, et al. Identification of
megalin/gp330 as a receptor for lipoprotein(a) in vitro. Arterioscler Thromb Vasc Biol.
1999;19(3):552–61.
Nordestgaard BG, Chapman MJ, Ray K, Boren J, Andreotti F, Watts GF, et al. Lipoprotein(a) as a
cardiovascular risk factor: current status. Eur Heart J. 2010;31(23):2844–53.
Rader DJ, Mann WA, Cain W, Kraft HG, Usher D, Zech LA, et al. The low density lipoprotein
receptor is not required for normal catabolism of Lp(a) in humans. J Clin Invest. 1995;95(3):
1403–8.
Reblin T, Niemeier A, Meyer N, Willnow TE, Kronenberg F, Dieplinger H, et al. Cellular uptake
of lipoprotein[a] by mouse embryonic fibroblasts via the LDL receptor and the LDL receptor-
related protein. J Lipid Res. 1997;38(10):2103–10.
Reyes-Soffer G, Ginsberg HN, Berglund L, Duell PB, Heffron SP, Kamstrup PR, et al.
Lipoprotein(a): a genetically determined, causal, and prevalent risk factor for atherosclerotic
cardiovascular disease: a scientific statement from the American Heart Association. Arterioscler
Thromb Vasc Biol. 2022;42(1):e48–60.
Romagnuolo R, Scipione CA, Boffa MB, Marcovina SM, Seidah NG, Koschinsky
ML. Lipoprotein(a) catabolism is regulated by proprotein convertase subtilisin/kexin type 9
through the low density lipoprotein receptor. J Biol Chem. 2015;290(18):11649–62.
Romagnuolo R, Scipione CA, Marcovina SM, Gemin M, Seidah NG, Boffa MB, et al. Roles of
the low density lipoprotein receptor and related receptors in inhibition of lipoprotein(a) inter-
nalization by proprotein convertase subtilisin/kexin type 9. PLoS One. 2017;12(7):e0180869.
134 L. Ismail et al.
Schachtl-Riess JF, Kheirkhah A, Grüneis R, Maio SD, Schoenherr S, Streiter G, et al. Frequent
LPA KIV-2 variants lower lipoprotein(a) concentrations and protect against coronary artery
disease. J Am Coll Cardiol. 2021;78(5):437–49.
Scipione CA, Sayegh SE, Romagnuolo R, Tsimikas S, Marcovina SM, Boffa MB, et al. Mechanistic
insights into Lp(a)-induced IL-8 expression: a role for oxidized phospholipid modification of
apo(a). J Lipid Res. 2015;56(12):2273–85.
Seimon TA, Nadolski MJ, Liao X, Magallon J, Nguyen M, Feric NT, et al. Atherogenic lipids and
lipoproteins trigger CD36-TLR2-dependent apoptosis in macrophages undergoing endoplas-
mic reticulum stress. Cell Metab. 2010;12(5):467–82.
Sharma M, Von Zychlinski-Kleffmann A, Porteous CM, Jones GT, Williams MJ, McCormick
SP. Lipoprotein (a) upregulates ABCA1 in liver cells via scavenger receptor-B1 through its
oxidized phospholipids. J Lipid Res. 2015;56(7):1318–28.
Sharma M, Redpath GM, Williams MJ, McCormick SP. Recycling of apolipoprotein(a) after
PlgRKT-mediated endocytosis of lipoprotein(a). Circ Res. 2017;120(7):1091–102.
Siddiqui H, Yevstigneyev N, Madani G, McCormick S. Approaches to visualising endocytosis of
LDL-related lipoproteins. Biomol. 2022;12(2):158.
Tam SP, Zhang X, Koschinsky ML. Interaction of a recombinant form of apolipoprotein[a]
with human fibroblasts and with the human hepatoma cell line HepG2. J Lipid Res.
1996;37(3):518–33.
Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD, Gordon S. Macrophage receptors
and immune recognition. Annu Rev Immunol. 2005;23:901–44.
Tomlinson JE, McLean JW, Lawn RM. Rhesus monkey apolipoprotein(a). Sequence, evolution,
and sites of synthesis. J Biol Chem. 1989;264(10):5957–65.
Tsimikas S, Hall JL. Lipoprotein(a) as a potential causal genetic risk factor of cardiovascular dis-
ease: a rationale for increased efforts to understand its pathophysiology and develop targeted
therapies. J Am Coll Cardiol. 2012;60(8):716–21.
Tsimikas S, Moriarty PM, Stroes ES. Emerging RNA therapeutics to lower blood levels of Lp(a):
JACC focus seminar 2/4. J Am Coll Cardiol. 2021;77(12):1576–89.
Van der Valk FM, Bekkering S, Kroon J, Yeang C, Van den Bossche J, van Buul JD, et al. Oxidized
phospholipids on lipoprotein(a) elicit arterial wall inflammation and an inflammatory mono-
cyte response in humans. Circulation. 2016;134(8):611–24.
Von Zychlinski A, Kleffmann T, Williams MJ, McCormick SP. Proteomics of lipoprotein(a)
identifies a protein complement associated with response to wounding. J Proteome.
2011;74(12):2881–91.
Wang N, Arai T, Ji Y, Rinninger F, Tall AR. Liver-specific overexpression of scavenger receptor BI
decreases levels of very low density lipoprotein apoB, low density lipoprotein apoB, and high
density lipoprotein in transgenic mice. J Biol Chem. 1998;273(49):32920–6.
White AL, Lanford RE. Cell surface assembly of lipoprotein(a) in primary cultures of baboon
hepatocytes. J Biol Chem. 1994;269(46):28716–23.
White AL, Guerra B, Lanford RE. Influence of allelic variation on apolipoprotein(a) folding in the
endoplasmic reticulum. J Biol Chem. 1997;272(8):5048–55.
Yang XP, Amar MJ, Vaisman B, Bocharov AV, Vishnyakova TG, Freeman LA, et al. Scavenger
receptor-BI is a receptor for lipoprotein(a). J Lipid Res. 2013;54(9):2450–7.
Yang X, Sethi A, Yanek LR, Knapper C, Nordestgaard BG, Tybjaerg-Hansen A, et al. SCARB1
gene variants are associated with the phenotype of combined high high-density lipoprotein
cholesterol and high lipoprotein(a). Circ Cardiovasc Genet. 2016;9(5):408–18.
Yeang C, Hung MY, Byun YS, Clopton P, Yang X, Witztum JL, et al. Effect of therapeutic inter-
ventions on oxidized phospholipids on apolipoprotein B100 and lipoprotein(a). J Clin Lipidol.
2016;10(3):594–603.
Zanoni P, Velagapudi S, Yalcinkaya M, Rohrer L, von Eckardstein A. Endocytosis of lipoproteins.
Atherosclerosis. 2018;275:273–95.
Chapter 8
Physiological Roles and Functions of
Lipoprotein(a)
It has been proposed that Lp(a) has a role in the transport of proinflammatory oxi-
dized phospholipids (OxPL). This hypothesis was driven by observations made by
Bergmark et al. who initially developed a method to measure OxPL bound to
apoB-100 by using a monoclonal antibody. They demonstrated that plasma OxPL/
apoB levels were quantitatively predictive of the presence and extent of angiograph-
ically determined CAD, identifying the presence and progression of carotid and
femoral atherosclerosis, and predicting CVD events over a 10-year interval. The
OxPL/apoB ratio was independent of all known risk factors, except for Lp[a], and,
remarkably, in all clinical studies performed at that time, there was an unusually
strong correlation of OxPL/apoB with Lp(a) (Bergmark et al. 2008).
These clinical observations suggested that OxPL may be bound to Lp[a] and may
thereby contribute to atherosclerotic plaque. In vitro studies demonstrated that not
only did OxPL bind to Lp(a), but it was the preferential carrier of OxPL in serum
(Bergmark et al. 2008). Notably, when compared to low-density lipoprotein (LDL),
Lp(a) bound three times as much OxPL. This led to the proposition that Lp(a)’s
physiologic roles may be to preferentially bind and transport OxPLs that are derived
from apoptosis and cell death, as occurs during inflammation and oxidative stress,
or when OxPLs are mobilized from tissues during iatrogenic plaque rupture during
Z. N. Safiullah
Osler Medical House Staff, The Johns Hopkins Hospital, Baltimore, MD, USA
e-mail: [email protected]
T. Leucker · S. R. Jones · P. P. Toth (*)
Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University
School of Medicine, Baltimore, MD, USA
e-mail: [email protected]; [email protected]; [email protected]
O
O O
O
O PC
O PC
Arachidonoyl-PC
HO
O O
EI-PC
H O PC O O
OV-PC
O PC
O O H
HO O PC O OH
G-PC IsoLGE2-PC
OH O O O
H
O PC O PC
O
HOOA-PC HO OH
O O
8-isoPGE2-PC
H O PC
O
OH O (CH2)15CH3
HOHA-PC O O
+
O O O P O
PAF N
HO OH
O PC
O
KOdiA-PC
O O O
O (CH2)15CH3
O PC HO O O
+
HAzPC O P O
N
OH
OH
12-HETE-PC HOO O
O
O PC
O PC
9-HPETE-PC
OH
13-HODE-PC
Fig. 8.2 Comparison of LDL and Lp(a) particles. (Left) Low-density lipoprotein (LDL) particle;
(right) lipoprotein(a) [Lp(a)] particle. Apoprotein (apo) B is the scaffolding for lipidation of both
lipoprotein species. Lp(a) is an LDL particle that is modified by the covalent addition of apo(a) to
apoB. Apo(a) is comprised of a series of kringles (protein loops; kringle IV [1–10] followed by
kringle V) and a protease terminus. The number of repeats in kringle IV type 2 are highly variable
person to person, genetically determined, and correlate with serum levels of Lp(a) as well as the
magnitude of risk for cardiovascular disease exerted by this lipoprotein. LDL-C low-density lipo-
protein cholesterol. (Figure and legend reproduced with permission from Toth PP. Familial
Hypercholesterolemia and Lipoprotein(a): Unraveling the Knot That Binds Them. Journal of the
American College of Cardiology 2020;75:2694–2697)
Promoter of Inflammation
a b c
d e f
Fig. 8.3 Increased arterial wall inflammation in subjects with elevated lipoprotein(a) [lp(a)]. (a)
Cross-sectional 18F-fluorodeoxyglucose uptake (18F-FDG) positron emission tomographic/com-
puted tomographic (PET/CT) images demonstrating an increased 18F-FDG uptake (yellow) in the
left carotid artery (top, white arrow) and aorta (bottom) in a subject with normal Lp(a) (left) and a
subject with elevated Lp(a) (right) quantified as the maximum target-to-background ratio (TBR) in
the (b) carotid arteries and (c) ascending aorta in subjects with elevated Lp(a) (n = 30) and normal
Lp(a) (n = 30). (d) Cross-sectional single-photon emission CT (SPECT)/CT images demonstrating
increased autologous technetium-99m (99mTc)-labeled peripheral blood mononuclear cell
(PBMC) accumulation (blue; at 6 h after infusion), depicted as the arterial wall–to–blood pool
ratio (ABR) at the level of (e) the carotid arteries and (f) ascending aorta in subjects with elevated
Lp(a) (n = 15) and normal Lp(a) (n = 15). **P < 0.01. (Figure and legend reproduced with permis-
sion from Valk FMvd, Bekkering S, Kroon J et al. Oxidized Phospholipids on Lipoprotein(a) Elicit
Arterial Wall Inflammation and an Inflammatory Monocyte Response in Humans. Circulation
2016;134:611–624)
deposition, as well as plaque formation and instability. The detrimental link between
inflammation and atherosclerotic CVD risk is further supported by those studies
showing that attenuation of inflammation is paralleled by improvement of surrogate
indicators of arterial function (e.g., endothelial function, aortic stiffness, ratio of
endothelial microparticles to endothelial progenitors) and cardiovascular prognosis
(Pirro et al. 2017).
Lp(a) is a well-known acute phase reactant whose production is stimulated by
inflammation. Notably, inflammatory response elements are present on the Lp(a)
gene. In primary monkey hepatocyte cultures, interleukin-6 (IL-6), IL-4, IL-13,
transforming growth factor-β (TGF-β), and tumor necrosis factor-α (TNF-α) can
8 Physiological Roles and Functions of Lipoprotein(a) 139
modulate Lp(a) gene expression. While IL-6 significantly increases plasma Lp(a)
levels, other cytokines inhibit Lp(a) synthesis by 50% or more at 10 ng/mL, with the
most powerful effect exerted by TGF-β and TNF-α (Ramharack et al. 1998).
Although there is sufficient evidence that inflammation may increase plasma Lp(a)
levels, data have emerged suggesting that a bidirectional regulatory loop involves
Lp(a) and inflammation; thus, Lp(a) may be proinflammatory in most cases, while
exerting anti-inflammatory effects in other conditions (Pirro et al. 2017). This pro-
inflammatory relationship leads to endothelial dysfunction, fragmentation, detach-
ment, and loss of repair activity, and activates inflammatory signaling cascades.
There is a close link between lipoproteins and inflammation in the arterial wall.
Oxidized LDLs (OxLDLs) trigger both directly and indirectly a proinflammatory
cascade leading to atherosclerosis development, progression, and complications
(Orsó and Schmitz 2017). On entry and trapping by interstitial matrix molecules
within the arterial intima, triglyceride-rich lipoprotein degradation by lipoprotein
lipase liberates free fatty acids and diacylglycerols, both of which are able to pre-
cipitate local inflammation (Orsó and Schmitz 2017). Oxidatively modified lipopro-
teins can be recognized by toll-like receptors, a type of pattern-recognition receptor
that responds against invading microbes, activating early innate recognition, and
host inflammatory responses. Also, lipoprotein exposure to reactive oxygen species
(superoxide anion, hydroxyl and peroxynitrite radicals) generates diverse oxidized
phospholipids (OxPLs) which can contribute to the initiation and the amplification
of the inflammatory response. Specifically, OxPLs present on OxLDLs can elicit
strong proinflammatory cytokine and chemokine responses from murine macro-
phages and human monocytes; they can also alter intracellular redox status and
directly activate proinflammatory genes, leading to arterial wall inflammation (Pirro
et al. 2017; van der Valk et al. 2016).
Lp(a) additionally has a role in initiating and stimulating inflammation, while
inhibiting anti-inflammatory pathways (Huang et al. 2014). Lp(a) binds monocyte
chemoattractant protein-1 (MCP-1), a chemokine that promotes the binding, roll-
ing, and transmigration of monocytes into the arterial intima (Deshmane et al.
2009). OxPLs have been shown to be major determinants for MCP-1 binding in the
vascular endothelium (Wiesner et al. 2013). Lp(a) inhibits the activation of trans-
forming growth factor-beta (TGF-β), a multifunctional and pleiotropic immune
regulatory cytokine that participates in peripheral immune tolerance and a negative
regulator of inflammation (Kojima et al. 1991). Lp(a) stimulates mRNA and cell
surface expression of intercellular adhesion molecule-1 (ICAM-1) in cultured
human umbilical vein endothelial cells (HUVECs) (Takami et al. 1998). Lp(a)-
induced expression of ICAM-1 in HUVECs appears to be mediated by decreasing
active TGF-β availability. Furthermore, Lp(a) promotes monocyte adhesion and
trans-endothelial migration by stimulation of MCP-1 and chemokine I-309, which
is the principal monocyte chemotactic cytokine produced by T helper cells (Haque
et al. 2000). Lp(a) promotes the differentiation of proinflammatory M1-type macro-
phages, leading to activation of T-helper-1 lymphocytes and natural killer cells.
Thus, macrophage expression of interleukin-1 (IL-1), IL8, and TNF-α is stimulated
by Lp(a) (Klezovitch et al. 2001). Secretion of these proinflammatory cytokines can
140 Z. N. Safiullah et al.
Impact on Malignancies
Following from its role as a proinflammatory agent, Lp(a) has a complicated role in
cancer biology. Observational studies have elucidated varying levels of Lp(a) in dif-
ferent malignancies. Elevations in serum Lp(a) are associated with lung cancer and
metastatic breast cancer (Orsó and Schmitz 2017; Lippi et al. 2007). However,
Lp(a) levels are found to be mostly decreased in hepatocellular cancer with some
variability (Orsó and Schmitz 2017; Gao et al. 2018). Lp(a) serum levels seem to be
independent of ovarian cancer, as well as acute lymphoblastic lymphoma.
Observationally, it is unclear if there is a singular relationship between Lp(a) con-
centration and cancer.
Lp(a) staining studies have identified Lp(a) in the tumor vasculature, rather than
in the primary parenchyma of the tumor (Lippi et al. 2007; Correc et al. 1990).
8 Physiological Roles and Functions of Lipoprotein(a) 141
improved mortality was seen in mice expressing LK68 and bearing human colorec-
tal cancer, lung cancer, or hepatocellular carcinoma (Lippi et al. 2007).
A large prospective cohort study of over 10,000 patients showed that low serum
Lp(a) levels correlated with increased mortality from cancer, specifically hepatocel-
lular carcinoma (HCC) (Lippi et al. 2007; Katzke et al. 2017). In HCC, increased
Lp(a) has been linked with decreased recurrence after resection. Further, it has util-
ity as a prognostic marker in α-fetoprotein <400 ng/mL level and tumor size <5 cm
in subgroups from a small prospective study. Similarly, a retrospective study that
compared serum Lp(a) concentration with tumor size in papillary thyroid cancer
found a significant negative correlation and concluded that Lp(a) may have a protec-
tive effect. Finally, a German cohort study found that increased Lp(a) levels were
associated with a total reduction of cancer mortality (Lippi et al. 2007; Katzke
et al. 2017).
Alternatively, a retrospective analysis done to investigate the association between
Lp(a) levels and clinical pathologic features of prostate cancer showed that higher
Lp(a) levels correlated with a high-risk prostate cancer phenotype (Wang and Zhang
2019). These results were similar to a separate German analysis (Katzke et al. 2017).
The authors theorized that increased Lp(a) levels may be compensatory to chronic
inflammation secondary to aggressive prostate cancer. Furthermore, increased Lp(a)
may promote increased cancer cell adhesion, invasion, and metastasis through its
role as a competitive inhibitor of plasmin-induced fibrinolysis (Wang and Zhang
2019). The clinical significance of Lp(a) in malignancy requires further investiga-
tion. Presently, Lp(a) may be protective in HCC and papillary thyroid cancer (Ma
et al. 2021). Conversely, it may be deleterious in prostate cancer. More research is
needed to understand the paradoxical underpinnings of these phenotypes.
Lp(a)’s angiostatic properties may make it a novel target for cancer therapeutics.
There are preclinical studies that have shown the utility of angiostatin gene therapy
in decreasing angiogenesis. Small molecule delivery and gene therapy do not have
cytotoxic side effects seen in conventional chemotherapy and immunotherapy.
Further research is needed to investigate whether the derived angiostatic mecha-
nisms of Lp(a)’s kringle domain can be developed into cancer therapies (Lippi
et al. 2007).
Lp(a) in Thrombosis
activator (Patthy 1985). The apo(a) subunit consists of ten different types of kringle
domains, differing in amino acid sequence, which are most homologous to plas-
minogen kringle IV (KIV), and a single plasminogen kringle V (KV)-like domain
and a protease-like domain (McLean et al. 1987). Of the ten KIV types in apo(a),
nine are present in single copy in all apo(a) isoforms (van der Hoek et al. 1993),
while KIV type 2 (KIV2) is encoded in a variable number of tandemly repeated
copies by the apo(a) gene (LPA), generating a series of different-sized LPA alleles
and, hence, apo(a) isoforms in the human population. Known alleles encode as few
as 1 and as many as 34 KIV2 repeats, giving rise to apo(a) isoforms containing
between 10 and 43 KIV-like domains, and polypeptide molecular masses between
200 and 800 kDa (Boffa and Koschinsky 2016).
Mechanistically, kringles are thought to function in ligand interactions with
lysine-containing substrates. Several of the kringles in plasminogen contain lysine-
binding sites (LBSs), defined structurally by a hydrophobic trough, lined by two or
three key aromatic side chains, which binds the aliphatic backbone of the lysine side
chain and that is flanked on either end by a cationic and anionic center (Hoover et al.
1993; McCance et al. 1994). Regarding LBS in plasminogen, the LBS in kringle I
has the highest affinity for lysine analogs, followed by KIV and KV (Castellino and
McCance 1997). The LBSs in plasminogen have been shown to be important for
both lysine-dependent interactions with substrates, such as fibrin and cell-surface
receptors, as well as for intramolecular interactions that maintain the closed native
conformation of plasminogen (Boffa and Koschinsky 2016; Cockell et al. 1998;
Violand et al. 1978).
The KIV types within apo(a) have varying affinities for lysine binding, with
KIV10 having a stronger affinity than KIV5–KIV8. The differential in lysine-
binding affinity is due to conservative amino acid substitutions (Ye et al. 2001;
Rahman et al. 2002). KIV10 is the only kringle domain that interacts with lysine-
containing substrates because the LBS in KIV5–KIV8 are masked when bound to
apoB-100 in Lp(a). In fact, KIV7–KIV8 have been explicitly shown to participate
in noncovalent interactions with specific lysine residues on apoB-100 that precede
covalent Lp(a) formation (Boffa and Koschinsky 2016).
In addition to the kringle domain, apo(a) has a protease domain. The protease-
like domain in apo(a) is catalytically inactive, despite having an intact Ser-His-Asp
catalytic triad (Gabel and Koschinsky 1995). An Arg to Ser substitution at the loca-
tion analogous to the site on plasminogen that is cleaved by plasminogen activators
ensures that an activating cleavage of apo(a) cannot occur (Hajjar et al. 1989). In
addition, several other amino acid substitutions relative to plasminogen, as well as
a key nine amino acid deletion in apo(a), have been proposed to render the protease-
like domain in apo(a) inactive (Boffa and Koschinsky 2016; Gabel and
Koschinsky 1995).
Lp(a) lacks protease activity while retaining the ability to bind to lysine-
containing substrates; hence, it is possible that Lp(a) may interfere with the func-
tions of plasminogen through molecular mimicry. This has been characterized by
several in vitro studies that have shown that Lp(a) interferes with plasminogen
activity (Boffa and Koschinsky 2016; Hajjar et al. 1989; Miles et al. 1989;
Romagnuolo et al. 2014). The lysine-binding function of plasminogen is crucial to
144 Z. N. Safiullah et al.
its fibrinolytic role. Activation of plasminogen by tPA occurs slowly in the absence
of a fibrin surface. In the presence of fibrin, a ternary complex is formed that results
in efficient production of plasmin (Hoylaerts et al. 1982). Plasminogen binding to
fibrin converts the protein from a closed to an open conformation that makes it a
better substrate for tPA (Urano et al. 1988). Further, partial degradation of fibrin by
plasmin results in the formation of carboxyl terminal lysine residues that mediate
positive feedback in the fibrinolytic cascade by promoting: (1) plasminogen binding
(Suenson and Petersen 1986), (2) plasmin-mediated conversion of native Glu1-
plasminogen to Lys77-plasminogen, which lacks the tail domain and is a better
substrate for tPA (Suenson and Thorsen 1988), and (3) binding to plasmin and thus
protecting it from consumption by antiplasmin (Boffa and Koschinsky 2016; Wiman
and Collen 1978).
The first studies that explored the functional implications of the homology
between apo(a) and plasminogen demonstrated the ability of Lp(a) and apo(a) to
inhibit binding of plasminogen to cell surface receptors on monocytes and endothe-
lial cells (Hajjar et al. 1989; Miles et al. 1989). Apo(a) was presumed to be a com-
petitive inhibitor of pericellular plasminogen activation. This was confirmed by
later experiments (Romagnuolo et al. 2014). It is also thought that this mechanism
may contribute to atherogenesis secondary to persistence of mural thrombi or extra-
cellular matrix degradation of the vascular wall (Boffa and Koschinsky 2016).
Regarding the effect of apo(a) on tPA, in vitro studies have demonstrated that
apo(a) and Lp(a) are capable of inhibiting tPA-mediated clot lysis and inhibiting
tPA-mediated plasminogen activation (Boffa and Koschinsky 2016). Furthermore,
apo(a) can inhibit the positive feedback step of plasmin-mediated Glu- to Lys-
plasminogen conversion in the context of fibrin. However, when it comes to under-
standing the mechanistic underpinnings of this observation, the data have been
mixed. Some data support that apo(a) directly competes with plasminogen for bind-
ing to fibrin. Whereas an alternate mechanism has been postulated that apo(a) forms
a quaternary complex with plasminogen, tPA, and fibrin that has a much lower turn-
over number than the ternary complex lacking apo(a) (Boffa and Koschinsky 2016).
Thus far we have explored the antifibrinolytic mechanisms of Lp(a). Additionally,
Lp(a) has well-characterized prothrombotic mechanisms including the promotion
of platelet aggregation and tissue factor pathway inhibitor (TFPI) binding (Boffa
and Koschinsky 2016). Lp(a) has a dual role in platelet aggregation (Boffa and
Koschinsky 2016). Experiments have shown that Lp(a) enhances platelet aggrega-
tion and granule release mediated by the thrombin receptor activation peptide
SFLLRN. It has also been shown that Lp(a) potentiates arachidonic acid-induced
platelet aggregation. Mechanistically, this is mediated by binding of apo(a) to lysine
residues on platelet receptors. Alternatively, there is evidence that Lp(a) or apo(a)
decreases platelet activation induced by collagen, ADP, or platelet-activating factor.
This duality of function may exist as a counterbalance to Lp(a)’s antifibrinolytic
activity (Boffa and Koschinsky 2016).
Lp(a) also directly binds tissue factor pathway inhibitor (TFPI). TFPI inhibits the
extrinsic coagulation cascade by binding to Factor Xa and then the tissue factor/
Factor VIIa complex. Apo(a) binds to TFPI through lysine residues in the carboxyl-
terminal portion of TFPI and decreases its fluid phase and cell surface activity,
8 Physiological Roles and Functions of Lipoprotein(a) 145
thereby inhibiting coagulation. Furthermore, the binding of Lp(a) to TFPI may con-
tribute to its atherogenic potential. Immunostaining studies on coronary atherec-
tomy samples have showed TFPI and apo(a) in smooth muscle cell-rich areas of the
intima (Boffa and Koschinsky 2016; Caplice et al. 2001).
Lp(a) also interacts with the fibrin clot. The fibrin clot is the final product of
primary hemostasis. The clot structure is important in determining both the stability
and fibrinolytic capacity of the fibrin clot, which has implications for abnormal
thrombolysis. Unfortunately, as compared with the role of Lp(a) in antifibrolytic
and prothrombotic, its mechanism of interaction with fibrin is not as well understood.
Observational studies have shown that elevated Lp(a) levels have been associated
with an altered fibrin clot structure that is accompanied by reduced fibrin clot per-
meability and impaired fibrinolysis (Boffa and Koschinsky 2016). This is thought to
be one of the etiologies of how Lp(a) contributes to CAD. Furthermore, from a
genetics perspective, the LPA gene contains a SNP (rs3798220) that results in an Ile
to Met substitution at amino acid 4399 within the protease-like domain of apo(a).
The allele encoding Met has been associated with elevated plasma Lp(a) levels,
small apo(a) isoform sizes, and increased risk for congenital heart disease (Luke
et al. 2007). Caucasians heterozygous for the Ile4399Met variant exhibit elevated
Lp(a) levels, increased clot density, and increased clot lysis times, while non-
Caucasian carriers showed increased clot permeability and shorter lysis times, with
no significant increase in Lp(a) levels. Interestingly, in the Women’s Heart Study,
individuals heterozygous for the Ile4399Met variant exhibited elevated Lp(a) levels
and an increased risk for CAD and benefited more from aspirin therapy than wild-
type subjects (Boffa and Koschinsky 2016). This could suggest a prothrombotic role
for the Ile4399Met polymorphism.
Most studies that examined the association between Lp(a) and venous thrombo-
embolic events (VTE) are cross-sectional. A meta-analysis of ten studies revealed
that among 13,541 subjects, those with a history of deep vein thrombosis were more
likely to have elevated Lp(a) (Dentali et al. 2017). Elevated Lp(a) was associated
with the presence of VTE at an odds ratio of 1.56 (95% CI 1.36–1.79). There was
also a stronger association between Lp(a) and VTE in patients with other predispos-
ing risk factors. A study of 467 patients with first VTE followed up for 1 year by
Marcucci et al. found a fivefold increased risk of recurrent VTE for Lp(a) >30 mg/
dL (OR 5.1, 95% CI 3.1–8.4), a level of risk similar to that seen in hyperhomocys-
teinemia and even higher than that for factor V Leiden or the factor II 20210GA
polymorphism (Caplice et al. 2001; Dentali et al. 2017; Crowther 2004; Nave and
von Eckardstein 2019).
Lp(a) in Diabetes
The mechanism by which Lp(a) influences the development of type 2 diabetes is not
well understood. Retrospective cohort studies have shown a negative correlation
between Lp(a) and insulin resistance (Mora et al. 2010). In a sample of 607 dyslip-
idemic patients, Lp(a) correlated inversely with serum triglycerides (TG) levels, TG/
146 Z. N. Safiullah et al.
HDL-C ratios, insulin, HOMA-IR, C-peptide, body mass index, and waist circum-
ference (Vaverková et al. 2017). Another study has also illustrated that there is a
sharp decrease in Lp(a) levels in the transition from prediabetes to type 2 diabetes
(Kaya et al. 2017). Genetic data from Chinese and Danish populations have shown
an increased risk of type 2 diabetes in individuals with genetically determined low
lipoprotein(a) plasma concentration due to large lipoprotein(a) isoform size related
to the number of kringle IV type 2 repeats. Alternatively, a Mendelian randomization
analysis showed that genetic variants associated with fasting insulin levels bore no
relation to Lp(a) concentration (Mora et al. 2010; Kamstrup and Nordestgaard 2013).
Like retrospective data, large prospective studies have also shown that there is a
negative association between Lp(a) and the risk of developing type 2 diabetes (Mora
et al. 2010; Schwartz et al. 2021). The negative association between Lp(a) and type
2 diabetes has also been observed to be dependent on the concentration of Lp(a).
Lower Lp(a) levels were associated with an increased risk of type 2 diabetes.
Similarly, it has been shown that Lp(a) levels are increased in prediabetes compared
to normoglycemic controls (Paige et al. 2017). Regarding sex differences, the
inverse relationship between Lp(a) and increased glucose levels is observed in men
prior to prediabetes. Whereas in women, the inverse relationship between Lp(a) and
glucose levels is observed only starting in prediabetes (Paige et al. 2017). The sig-
nificance of this observation is not yet clear. There was also no association between
isoform size and risk of diabetes (Kamstrup and Nordestgaard 2013). One hypoth-
esized mechanism that explains this observation is that insulin suppresses apo(a) in
hepatocytes. The biologic role of Lp(a) in insulin resistance and hyperglycemia
requires more interrogation.
In those with cardiovascular disease, low levels of Lp(a) have also been associ-
ated with a greater prevalence of type II diabetes mellitus in prospective, retrospec-
tive, and genetic studies. In an analysis of 13,480 patients in the ODESSY
OUTCOMES trial, similar findings were observed with negative correlation
between Lp(a) and the prevalence of type 2 diabetes. Furthermore, in the same
analysis, reduction of Lp(a) levels by the PCSK9 inhibitor alirocumab in those with
high baseline Lp(a) level increased the estimated risk of incident type 2 diabetes
compared with placebo hazard ratio 1.07 (95% CI 1.03–1.12; P < 0.0002) (Schwartz
et al. 2021). However, there was an interaction between treatment with alirocumab
and baseline Lp(a) on the risk of incident type 2 diabetes. The concentration of
Lp(a) at which alirocumab had a neutral effect on incident type 2 diabetes was
around 50 mg/dL. It was shown that PCSK9 inhibitor-induced reductions of Lp(a)
levels lead to an increased risk of type 2 diabetes (Schwartz et al. 2021).
Although the mechanism by which low levels of Lp(a) contribute to an increased
risk of type 2 diabetes is unknown, the risk may be modifiable as evidenced by
Lp(a) levels in the above study that were neutral. Further, it seems that those with
higher baseline Lp(a) who are treated with PCSK9 inhibitor therapy have an
increased risk to develop type 2 diabetes than those with lower baseline Lp(a) levels
(Schwartz et al. 2021). This observation, if confirmed in additional studies, is impor-
tant to consider when prescribing this therapy to mitigate cardiovascular risk.
8 Physiological Roles and Functions of Lipoprotein(a) 147
In summary, retrospective and prospective data have elucidated that Lp(a) levels
are inversely related to an increased incidence of type 2 diabetes. The mechanism
by which Lp(a) participates in diabetogenic pathophysiology is not yet understood.
However, based on the studies discussed previously, the risk is modifiable through
PCSK9 inhibitor therapy. Additional investigation is needed to understand whether
Lp(a)-directed therapies currently in development impact risk for the development
of diabetes.
Lp(a) is implicated in wound healing. Histologic studies have identified Lp(a) in all
four stages of wound healing from infiltration of inflammatory cells to formation of
granulation tissue. In the first stage of healing, there is an infiltration of inflamma-
tory cells followed by formation of a fibrin clot mixed with red blood cells covers
the exposed wound surface. In the second stage, the immature cell mass is replaced
by granulation tissue, which is produced by fibroblasts, endothelial cells, and vas-
cular sprouts from adjacent viable tissues, induced by growth factors released dur-
ing the first stage. In this stage, granulation tissue is often covered with loose fibrous
connective tissue with various thickness, which forms the fibrous cap. Angiogenesis
also takes place in this stage. The epithelial sheets are spread to cover the granula-
tion tissue in the third stage. In the last stage, collagen fibers replace the granulation
tissue, resulting in reduction of wound size. Finally, the healing process is com-
pleted by replacement of granulation tissue with new epithelium or by organization
(Yano et al. 1997).
The apo(a) and apoB100 subunits of Lp(a) are more strongly identified in the
fibrous cap, endothelial cells, and plasminogen and fibrinogen-rich surfaces than in
the re-epithelized tissue surface. The mechanism by which Lp(a) influences wound
healing has not been described. Following from discussions elsewhere of Lp(a)’s
role in angiogenesis and antifibrinolysis, it is possible that Lp(a) promotes the pro-
liferation of endothelium with accompanying vasculogenesis; and is also involved
in maintenance of the fibrin cap and preventing excessive fibrinolysis (Yano
et al. 1997).
Lp(a) elevation is associated with several autoimmune diseases (Missala et al. 2012;
Toms et al. 2011). The mechanisms by which Lp(a) contributes to autoimmune
disease are through acute phase reactions, autoantibodies, and fibrinolysis (Missala
et al. 2012). The interplay of these mechanisms leads to increased inflammation
which contributes to clinical autoimmune phenotypes.
148 Z. N. Safiullah et al.
Calcific aortic valve stenosis (CAVS) is the most common valve disease in the
elderly population, affecting >1 million patients in the USA, and is associated with
significant morbidity and mortality (Guddeti et al. 2020). Elevated Lp(a) is linked
to increased risk for calcific aortic valve stenosis (CAVS). Observational studies
8 Physiological Roles and Functions of Lipoprotein(a) 149
from the early 1990s showed that increased Lp(a) levels were associated with aortic
valve calcification and stenosis. This relationship is linear, with higher Lp(a) levels
correlating with higher risk. Multivariate analyses have shown that increased Lp(a)
is an independent predictor of developing CAVS. Additionally, both prospective
and retrospective genetic studies have shown that the LPA locus carries a greater
risk of CAVS and may be causative. Interestingly, some studies have shown aortic
stenosis after the sixth decade does not correlate with Lp(a) levels (Guddeti
et al. 2020).
The relationship between Lp(a) and CAVS is driven in part by oxidized phospho-
lipids. Oxidized phospholipid apoB-100 was linked to faster progression of aortic
stenosis in those with elevated Lp(a). This observation leads to the postulation that
Lp(a) leads to aortic valve stenosis by phospholipid oxidation. Mechanistically,
OxPL are proinflammatory, can lead to endothelial dysfunction, and promote osteo-
genic differentiation which leads to calcification. Interestingly, the relationship
between aortic stenosis progression and OxPL content may be linear, based on a
subgroup analysis of the ASTRONOMER (Effects of Rosuvastatin on Aortic
Stenosis Progression) clinical trial (Vavuranakis et al. 2020).
Increased Lp(a) levels have also been associated with valve calcification in
patients with bicuspid aortic valves. Fewer KIV-2 repeats have also been linked to
more severe calcification. In the context of calcification of a bicuspid aortic valve,
Lp(a) could be a useful marker to identify those at risk to develop valve calcification
and stenosis (Guddeti et al. 2020).
CAVS is often present in the setting of CAD. Interestingly, the relationship
between Lp(a) and CAVS is independent of CAD. However, studying patients with
this comorbidity has yielded novel insights into the mechanism of how Lp(a) pro-
motes CAVS. Autotaxin (ATX), a lysophospholipase D enzyme, transforms lyso-
phosphatidylcholine into lysophosphatidic acid (LysoPA). ATX is transported in the
aortic valve via the bloodstream by Lp(a) and is also secreted by valve interstitial
cells. ATX-LysoPA has been shown to promote inflammation and leads to calcifica-
tion of the aortic valve, thus promoting CAVS. Autotaxin also indirectly promotes
the nuclear translocation of the transcription factor NF-κB, which leads to height-
ened inflammation (Nsaibia et al. 2016).
In addition to deposition of oxidized phospholipids and inflammation mediated
by autotaxin, Lp(a) has other pleiotropic mechanisms that lead to CAVS. Lp(a) is
thought to participate in cholesterol deposition on the aortic valve cusps causing
thickening. Lp(a) is also implicated in macrophage apoptosis and might contribute
to early valve lesion progression. Further, following from Lp(a)’s role in thrombo-
sis, it can cause fibrin deposition on the leaflets which can cause stenosis (Guddeti
et al. 2020; Vavuranakis et al. 2020; Nsaibia et al. 2016).
In summary, Lp(a) contributes to the pathogenesis and progression of calcific
aortic stenosis. Lp(a) has several pleiotropic mechanisms that contribute to CAVS,
including, cholesterol deposition, delivery of OxPL, fibrin deposition, and inflam-
mation mediated by autotaxin. More research is needed to determine if Lp(a) lower-
ing therapy can mitigate the development of calcific aortic stenosis.
150 Z. N. Safiullah et al.
Elevated Lp(a) leads to an increased risk of coronary artery disease (CAD) and
cardiovascular events (Tsimikas et al. 2020). Lp(a) contributes to atherosclerosis
independent of LDL-C by many of the mechanisms discussed previously. These
mechanisms include delivery of oxidized phospholipids and promotion of inflam-
mation and thrombosis (Rehberger Likozar et al. 2020). Additionally, Lp(a) is asso-
ciated with IL-8, a proinflammatory, prothrombotic, and proatherogenic cytokine,
which attracts leukocytes, triggers tissue factor production, and promotes adhesion
of monocytes to early atherosclerotic plaques (Lippi et al. 2021).
Observational data of the role of Lp(a) in CAD have shown that Lp(a) levels are
higher in the setting of stable angina compared to unstable angina.
Immunohistochemically, 90% of the Lp(a) area in coronary atheromas co-localizes
with plaque macrophages, and 30% of which correlates with plaque a-actin, which
might be related to the role of Lp(a) in plaque enlargement (Dangas et al. 1998).
Similarly, in acute myocardial infarction (MI), Lp(a) levels increase significantly
within the first 24 h and normalize within about 30 days (Rehberger Likozar
et al. 2020).
The compete mechanism of how Lp(a) contributes to atherogenesis is not yet
fully understood. In addition to the mechanisms discussed previously, it is pro-
posed that Lp(a) is deposited on the vascular wall and is readily taken up by mac-
rophage scavenger receptors. The macrophages soon become foam cells and the
canonical pathway of atherogenesis follows. Lp(a) also induces endothelial dys-
function which is proatherogenic (van der Valk et al. 2016; Rehberger Likozar
et al. 2020). As detailed previously, Lp(a) leads to coronary thrombi formation by
antifibrinolysis (i.e., competitive inhibition of tPA). In addition, Lp(a) promotes
coagulation and platelet aggregation and boosts inflammation (Boffa and
Koschinsky 2016).
Based on the increased CVD mortality conferred by increased Lp(a) levels, cur-
rent European Society of Cardiology guidelines recommend screening at least once.
The 2018 American College of Cardiology/American Heart Association guidelines
on blood cholesterol defined Lp(a) 50 mg/dL, or 125 nmol/L, as a risk-enhancing
factor; according to their guidelines, this is a relative indication for its measurement
with a family history of premature CVD. Those with elevated Lp(a) >180 mg/dL
carry a risk of atherosclerotic CVD equivalent to patients heterozygous for familial
hypercholesteremia (Rehberger Likozar et al. 2020).
Multiple studies have shown that the association of genetically predicted Lp(a)
levels with the risk of CVD is independent of changes in LDL cholesterol levels.
This is thought due to genetic variants that mimic the LDL lowering effects of
statins, PCSK9 inhibitors, and ezetimibe to the risk of CVD (Burgess et al. 2018).
This observation is more significant in younger patients. In those less than 45 years
old, in whom elevated Lp(a) levels (>120 nmol/L, 80th percentile) are associated
with a threefold increased risk of MI. The clinical benefit of lowering Lp(a) levels
is proportional to the absolute reduction in Lp(a) levels. An absolute reduction in
Lp(a) levels of approximately 100 mg/dL should result in a clinically relevant
8 Physiological Roles and Functions of Lipoprotein(a) 151
reduction in the risk of CVD. Such a decrease in Lp(a) represents the same magni-
tude of CVD risk reduction achieved by lowering LDL cholesterol levels by
38.67 mg/dL (Tsimikas et al. 2020; Rehberger Likozar et al. 2020).
Specifically, in acute coronary syndromes (ACS), Lp(a) elevation is observed for
up to 4 months after an event. Concomitantly, OxPL levels are also elevated, which
could mean that Lp(a) participates in OxPL delivery during acute plaque rupture.
Interestingly, Lp(a) and OxPL transient elevations have also been observed after
percutaneous coronary intervention for stable CAD (Tsimikas et al. 2020). Lp(a)
levels are also inversely related to the age of first presentation with ACS. This means
that younger patients presenting with ACS or observed to have higher Lp(a) levels
than older patients with a similar ACS presentation. This reflects the importance of
other, traditional atherosclerotic risk factors in older individuals, in contrast to a
more important role of Lp(a) in younger individuals (Vavuranakis et al. 2020).
Evolution of Lp(a)
Further genetic analysis has elucidated the boundaries of intron and exon
sequences are remarkably similar between these genes and only differ between 1%
and 5% (Lawn et al. 1997; Ichinose 1992). Also, the intron and exon junction posi-
tions are almost identical. These findings suggest that apo(a) and these other genes
may have developed during recent primate evolution from a common ancestral
component of the kringle-related serine protease, most likely plasminogen, via
duplication and exon shuffling (Lippi and Guidi 2000).
The apo(a) gene is most homologous with the proenzyme plasminogen. They
both share the Kringle V domain. However, the kringle IV domain of plasminogen
is in the apo(a) gene as multiple variable tandem repeats (McLean et al. 1987).
Furthermore, a point mutation in the domain homologous to the protease domain of
plasminogen deprives apo(a) of enzymatic activity (Lippi and Guidi 2000).
It is evident that Lp(a) has been conserved through evolution. This may be due to
its positive influence on wound healing, thrombosis in the face of injury and, when
at high concentrations, may be protective against diabetes. Its structural similarities
to other local genes inform on its diverse functions. Lp(a) has a complex mechanism
of action and although may seem deleterious in some contexts; it could also confer
an evolutionary advantage that is not yet fully understood.
COVID-19 infection has led to increased mortality in those with significant pulmo-
nary disease, ischemic heart disease, diabetes, and human immunodeficiency virus
infection (Enkhmaa and Berglund 2022). COVID-19 infection leads to a hyperin-
flammatory state that is linked with an increased risk of venous thromboembolism
and cardiovascular complications (Enkhmaa and Berglund 2022).
It was hypothesized that Lp(a) has a synergistic effect with COVID-19 infection.
This follows from its proinflammatory and prothrombotic roles discussed else-
where. Briefly, because Lp(A) is an acute phase reactant and contributes to inflam-
mation, in part, by carrying oxidized phospholipids. Regarding thrombosis, one
major mechanism is the inhibition of endogenous fibrinolysis (Boffa and Koschinsky
2016). Furthermore, the stimulation of IL-6 from COVID-19 infection was thought
to promote the acute phase expression of Lp(a). When the relationship between
Lp(a) levels and COVID-19 infection was interrogated, there was no significant dif-
ference in the serum Lp(a) levels between those infected with COVID-19 and con-
trols (Enkhmaa and Berglund 2022).
However, Lp(a) levels increase over the course of hospitalization and increase in
concentrations that are associated with the severity of COVID-19 infection and
stage of acute kidney injury in these patients. In the COVID-19 population in a
small observational study, Lp(a) was not associated with IL-6, a well-known inflam-
matory marker. Taken together, these results suggest that a hyper Lp(a) state,
8 Physiological Roles and Functions of Lipoprotein(a) 153
Conclusions
References
Cockell CS, Marshall JM, Dawson KM, Cederholm-Williams SA, Ponting CP. Evidence that the
conformation of unliganded human plasminogen is maintained via an intramolecular inter-
action between the lysine-binding site of kringle 5 and the N-terminal peptide. Biochem
J. 1998;333(Pt 1):99–105.
Correc P, Kostner GM, Burtin P. A comparative study of the localization of plasminogen and
apolipoprotein(a) in human carcinomas. Thromb Res. 1990;58:213–20.
Cox LA, Jett C, Hixson JE. Molecular basis of an apolipoprotein[a] null allele: a splice site muta-
tion is associated with deletion of a single exon. J Lipid Res. 1998;39:1319–26.
Crowther M. Lipoprotein(a): another risk factor for venous thrombosis? Am J Med.
2004;115(8):667–8.
Dangas G, Mehran R, Harpel PC, et al. Lipoprotein(a) and inflammation in human coronary ather-
oma: association with the severity of clinical presentation. J Am Coll Cardiol. 1998;32:2035–42.
Dentali F, Gessi V, Marcucci R, Gianni M, Grandi AM, Franchini M. Lipoprotein(a) as a risk factor
for venous thromboembolism: a systematic review and meta-analysis of the literature. Semin
Thromb Hemost. 2017;43:614–20.
Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1):
an overview. J Interf Cytokine Res. 2009;29:313–26.
Dominguez M, Rojas G, Loyau S, Bazurco M, Sorell L, Anglés-Cano E. Kringles of the plasmino-
gen–prothrombin gene family share conformational epitopes with recombinant apolipoprotein
(a): specificity of the fibrin-binding site. Biochim Biophys Acta. 2001;1548:72–80.
Enkhmaa B, Berglund L. Lp(a) and SARS-CoV-2: a conspiracy of two mysteries. J Intern Med.
2022;291:8–10.
Forsgren M, Råden B, Israelsson M, Larsson K, Hedén L. Molecular cloning and characterization
of a full-length cDNA clone for human plasminogen. FEBS Lett. 1987;213:254–60.
Frank SL, Klisak I, Sparkes RS, et al. The apolipoprotein(a) gene resides on human chromo-
some 6q26-27, in close proximity to the homologous gene for plasminogen. Hum Genet.
1988;79:352–6.
Gabel BR, Koschinsky ML. Analysis of the proteolytic activity of a recombinant form of
apolipoprotein(a). Biochemistry (Easton). 1995;34:15777–84.
Gao XH, Zhang SS, Chen H, Wang K, Xie W, Wang FB. Lipoprotein (a): a promising prognos-
tic biomarker in patients with hepatocellular carcinoma after curative resection. Onco Targets
Ther. 2018;11:5917–24.
Guddeti RR, Patil S, Ahmed A, et al. Lipoprotein(a) and calcific aortic valve stenosis: a systematic
review. Prog Cardiovasc Dis. 2020;63:496–502.
Hajjar KA, Gavishi D, Breslow JL, Nachman RL. Lipoprotein(a) modulation of endothelial cell
surface fibrinolysis and its potential role in atherosclerosis. Nature (London). 1989;339:303–5.
Haque NS, Zhang X, French DL, et al. CC chemokine I-309 is the principal monocyte che-
moattractant induced by apolipoprotein(a) in human vascular endothelial cells. Circulation.
2000;102:786–92.
Hoover GJ, Menhart N, Martin A, Warder S, Castellino FJ. Amino acids of the recombinant krin-
gle 1 domain of human plasminogen that stabilize its interaction with omega-amino acids.
Biochemistry (Easton). 1993;32:10936–43.
Hoylaerts M, Rijken DC, Lijnen HR, Collen D. Kinetics of the activation of plasminogen by
human tissue plasminogen activator. role of fibrin. J Biol Chem. 1982;257:2912–9.
Huang M, Gong Y, Grondolsky J, Hoover-Plow J. Lp(a)/apo(a) modulate MMP-9 activation and
neutrophil cytokines in Vivo in inflammation to regulate leukocyte recruitment. Am J Pathol.
2014;184:1503–17.
Ichinose A. Multiple members of the plasminogen-apolipoprotein(a) gene family associated with
thrombosis. Biochemistry (Easton). 1992;31:3113–8.
Kamstrup PR, Nordestgaard BG. Lipoprotein(a) concentrations, isoform size, and risk of type 2
diabetes: a mendelian randomisation study. Lancet Diabetes Endocrinol. 2013;1:220–7.
Katzke VA, Sookthai D, Johnson T, Kühn T, Kaaks R. Blood lipids and lipoproteins in relation to
incidence and mortality risks for CVD and cancer in the prospective EPIC-Heidelberg cohort.
BMC Med. 2017;15:218.
156 Z. N. Safiullah et al.
Paige E, Masconi KL, Tsimikas S, et al. Lipoprotein(a) and incident type-2 diabetes: results
from the prospective Bruneck study and a meta-analysis of published literature. Cardiovasc
Diabetol. 2017;16:38.
Patthy L. Evolution of the proteases of blood coagulation and fibrinolysis by assembly from mod-
ules. Cell. 1985;41:657–63.
Pirro M, Bianconi V, Paciullo F, Mannarino MR, Bagaglia F, Sahebkar A. Lipoprotein(a) and
inflammation: a dangerous duet leading to endothelial loss of integrity. Pharmacol Res.
2017;119:178–87.
Rahman MN, Becker L, Petrounevitch V, Hill BC, Jia Z, Koschinsky ML. Comparative analyses of
the lysine-binding site properties of apolipoprotein(a) kringle IV types 7 and 10. Biochemistry
(Easton). 2002;41:1149–55.
Ramanujan S, Koenig GC, Padera TP, Stoll BR, Jain RK. Local imbalance of proangiogenic and
antiangiogenic factors: a potential mechanism of focal necrosis and dormancy in tumors.
Cancer Res. 2000;60:1442–8.
Ramharack R, Barkalow D, Spahr MA. Dominant negative effect of TGF-beta1 and TNF-alpha
on basal and IL-6-induced lipoprotein(a) and apolipoprotein(a) mRNA expression in primary
monkey hepatocyte cultures. Arterioscler Thromb Vasc Biol. 1998;18:984–90.
Rehberger Likozar A, Zavrtanik M, Šebeštjen M. Lipoprotein(a) in atherosclerosis: from patho-
physiology to clinical relevance and treatment options. Ann Med (Helsinki). 2020;52:162–77.
Romagnuolo R, Marcovina SM, Boffa MB, Koschinsky ML. Inhibition of plasminogen activation
by apo(a): role of carboxyl-terminal lysines and identification of inhibitory domains in apo(a)
[S]. J Lipid Res. 2014;55:625–34.
Romero FI, Atsumi T, Tinahones FJ, et al. Autoantibodies against malondialdehyde-modified
lipoprotein(a) in antiphospholipid syndrome. Arthritis Rheum. 1999;42:2606–11.
Schmitz G, Orsó E. Lipoprotein(a) hyperlipidemia as cardiovascular risk factor: pathophysiologi-
cal aspects. Clin Res Cardiol Suppl. 2015;10:21–5.
Schwartz GG, Szarek M, Bittner VA, et al. PMC8132323; relation of lipoprotein(a) levels to incident
type 2 diabetes and modification by alirocumab treatment. Diabetes Care. 2021;44:1219–27.
Staton CA, Lewis CE. Angiogenesis inhibitors found within the haemostasis pathway. J Cell Mol
Med. 2005;9:286–302.
Suenson E, Petersen LC. Fibrin and plasminogen structures essential to stimulation of plasmin
formation by tissue-type plasminogen activator. Biochim Biophys Acta. 1986;870:510–9.
Suenson E, Thorsen S. The course and prerequisites of lys-plasminogen formation during fibrino-
lysis. Biochemistry (Easton). 1988;27:2435–43.
Takami S, Yamashita S, Kihara S, et al. Lipoprotein(a) enhances the expression of intercel-
lular adhesion molecule-1 in cultured human umbilical vein endothelial cells. Circulation.
1998;97:721–8.
Toms TE, Panoulas VF, Kitas GD. Dyslipidaemia in rheumatological autoimmune diseases. Open
Cardiovasc Med J. 2011;5:64–75.
Tsimikas S, Witztum JL. The role of oxidized phospholipids in mediating lipoprotein(a) atheroge-
nicity. Curr Opin Lipidol. 2008;19:369–77.
Tsimikas S, Brilakis ES, Miller ER, et al. Oxidized phospholipids, lp(a) lipoprotein, and coronary
artery disease. N Engl J Med. 2005;353:46–57.
Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, et al. Lipoprotein(a) reduction in per-
sons with cardiovascular disease. N Engl J Med. 2020;382:244–55.
Urano T, Sator de Serrano V, Gaffney PJ, Castellino FJ. Effectors of the activation of human
[Glu1]plasminogen by human tissue plasminogen activator. Biochemistry (Easton).
1988;27:6522–8.
van der Hoek YY, Wittekoek ME, Beisiegel U, JJP K, Koschinsky M. The apolipoprotein(a) krin-
gle IV repeats which differ from the major repeat kringle are present in variably sized isoforms.
Hum Mol Genet. 1993;2:361–6.
van der Valk FM, Bekkering S, Kroon J, et al. Oxidized phospholipids on lipoprotein(a) elicit
arterial wall inflammation and an inflammatory monocyte response in humans. Circulation.
2016;134:611–24.
158 Z. N. Safiullah et al.
Introduction
Anastasiya Matveyenko and Marianna Pavlyha contributed equally with all other contributors.
no evidence that Lp(a) levels in plasma are related to lipoprotein lipase activity, and
it is unlikely that it is derived from catabolism of other lipoproteins. Similarly, Lp(a)
clearance may be regulated by various pathways (Reyes-Soffer and Ramakrishnan
2017; Chemello et al. 2022), depending on the particle composition. Several studies
show that Lp(a) clearance can be dependent on LDL receptors; however, studies
with PCSK9 inhibitors varied in their results (Reyes-Soffer and Ramakrishnan
2017; Chemello et al. 2022). The latter may be related to the use of statins and dif-
ferences in ethnicities of the cohorts evaluated (Reyes-Soffer and Ramakrishnan
2017; Chemello et al. 2022). Large studies have shown an inverse relationship
between Lp(a) plasma concentrations and isoform sizes, based on the KIV-2 repeats
(Sandholzer et al. 1992; Kraft et al. 1992; Stefanutti et al. 2020). This relationship
can account for 30–70% of the plasma levels. Notably, Black and Asian Indian eth-
nicities have higher levels of Lp(a) when compared to Caucasians, pointing again to
genetics as being one of the determining factors (Kronenberg and Utermann 2013;
Enkhmaa and Berglund 2016; Reyes-Soffer et al. 2021; Patel et al. 2021).
Additionally, small differences in Lp(a) levels have been found between men and
women (Markus et al. 2021; Forbang et al. 2016; Simony et al. 2022).
Inflammation
Triglycerides
apo(a) Thrombosis
Coagulation
Cholesterol
Associated
Proteins*
Genetic Variation
KV3
Protease
domain Thrombosis and plaque
KV KV10 KV9 KV8 KV7 KV6 KV5 KV4
Fig. 9.1 Lipoprotein(a): biology, pathophysiology, and disease development. (Panel a) The struc-
ture and function of circulating lipoprotein particles have been nicely described. Proteins (*) on the
lipoprotein(a) particle have led to further understanding of its link to disease development
(McCormick and Schneider 2018). (Panel b) Lp(a) has been linked to atherosclerosis and the roles
of genetics and the additional proteins bound to the particle are still to be fully described
(Kronenberg 2022). (Panel c) As an apoB100-containing particle, Lp(a) can lead to plaque forma-
tion, yet, further research is needed to understand its link to specific disease presentation. Genome-
wide association, epidemiological, and Mendelian randomization studies support its role as causal
in development of ASCVD
162 A. Matveyenko et al.
macrophages with subsequent transformation into foam cells sheds light onto its
integral role in atherogenesis. The role of Lp(a) in coronary artery disease (CAD)
has been studied previously (Rasouli et al. 2006), with a recent study reaffirming
that high Lp(a) levels are associated with increased progression of coronary low-
attenuation plaque (necrotic core) between baseline and 12 months of follow-up in
patients with advanced stable CAD (Kaiser et al. 2022). The role of Lp(a) in these
specific atherogenic processes has been studied by various authors (Marchini et al.
2021), but its exact pathophysiology, not related to conventional apoB injury, has
not been completely described.
Myocardial Infarction
Multiple studies have demonstrated the association between Lp(a) and myocardial
infarctions (MIs), although these were mostly conducted in Caucasian populations
(Afanasieva et al. 2022). Lp(a) levels greater than 50 mg/dL were linked to greater
risk of developing an MI (Kamstrup et al. 2008). A study looking at a Danish popu-
lation showed that there was a step-wise effect with Lp(a) levels and risk of MI
(Emerging Risk Factors Collaboration et al. 2009; Erqou et al. 2009; Langsted et al.
2015). Particularly those patients with Lp(a) levels in the 95th percentile had as high
as three to fourfold risk for an experiencing at event. This risk was noted to be
higher in men compared to women.
Aortic Stenosis
There is a strong association between Lp(a) and Aortic Stenosis (AS) with main
driver in the mechanism of the disease being oxidized phospholipids. There is also
an association between higher Lp(a) measurements with a more rapid progression
of stenosis and greater need for aortic valve replacement compared to the group
with lower Lp(a). Exposure of valvular interstitial cells to Lp(a) increases the
expression of the osteoblastic transcription factors, runt-related transcription factor
2 (RUNX2) and bone morphogenetic protein 2 (BMP2), suggesting that Lp(a) plays
a role in osteogenic differentiation of valvular interstitial cells with oxidizing phos-
pholipids playing an integral role in this mechanism (Zhiduleva et al. 2018).
Additionally, Lp(a) plays a major role in calcific aortic valve stenosis (CAVS)
Calcific aortic valve stenosis (CAVS). The latter highlighted by human proteomic
studies that found lifelong exposure to elevated Lp(a) contributes to the develop-
ment and progression of CAVS Calcific aortic valve stenosis (CAVS) through mul-
tiple pathways (Bourgeois et al. 2021).
Stroke
There has been conflicting data regarding the association between levels of Lp(a)
and stroke (Colantonio et al. 2022; Pan et al. 2022). It is well established that Black
populations are at a higher risk for CAD stroke, and mortality compared to other
ethnicities and tend to have higher levels of Lp(a). The REasons for Geographic
and Racial Differences in Stroke (REGARDS) study published data on Lp(a) and
164 A. Matveyenko et al.
stroke in a race and sex stratified cohort. Their findings, after adjusting for other
risk factors, showed a correlation between high Lp(a) levels and ischemic stroke
with higher hazard ration in Black populations. Women had higher on average
Lp(a) levels, but no statistically significant association with stroke. The driving
mechanisms are thought to be similar as in CAD and PAD, involving increased
cholesterol deposition in plaque, inflammation, and prothrombotic effects (Arora
et al. 2019).
While there are numerous publications linking Lp(a) and CAD data pertaining to
PAD are not as robust (Norgren et al. 2007). Review of current publications does,
however, suggest that elevated levels of Lp(a) are associated with increased inci-
dence, progression, and post-treatment recurrence of PAD (Tmoyan et al. 2018).
The pathophysiology behind this relationship is driven by the ability of Lp(a) par-
ticles to migrate more easily into the subendothelial space when compared to LDL
particles (Kraaijenhof et al. 2021). This is facilitated by endothelial activation via
oxidized phospholipids and upregulation of chemokines and adhesion molecules.
Additionally, Lp(a) has been shown to compete for binding of plasminogen and
plasmin, generating a prothrombotic state (Boffa 2022). The latter is especially
important for those patients with limb threat due to tibial disease (below the knee)
(Tsimikas 2017). High Lp(a) levels have been shown to be associated with increased
incidence of claudication, symptom progression, re-stenosis after intervention, hos-
pitalization due to PAD, and limb amputation (Price et al. 2001). Patients with ele-
vated Lp(a) also have higher risk of combined PAD outcomes after adjusting for
other traditional risk factors (Kosmas et al. 2019). A recent prospective, observa-
tional study in symptomatic lower extremity arterial disease comparing patients
with high and low Lp(a) was performed. It showed that compared with low-Lp(a)
group, patients with high-Lp(a) had a higher proportion of heart failure, CLTI, and
multivessel lesions as well as higher LDL cholesterol. A 5-year incidence of all-
cause mortality was significantly higher in the high Lp(a) cohort than in those with
low Lp(a) (48.1% vs. 27.3%). Additionally, the cumulative 5-year incidence of
major adverse limb occurrence was also significantly higher in patients with high
Lp(a) levels (67.9% vs. 27.2%) (Tomoi et al. 2022).
Lp(a) is a known independent risk factor for increased mortality. A study published
in 2019, examining a Danish population, reported high risk of both cardiovascular
and all-cause mortality with no difference in noncardiovascular-related mortality
(Langsted et al. 2019). Reported median survival was the lowest in those patients
9 The Role of Lp(a) in Atherosclerosis: An Overview 165
who had the highest measured Lp(a) (>93 mg/dL). The known causal factors driving
high mortality in patients with Lp(a) are mainly coronary heart disease, myocardial
infarction, atherosclerotic stenosis, and aortic valve stenosis (Nordestgaard and
Langsted 2016). Another cross-sectional study done in United Kingdom looking at
a large cohort of patients determined via Mendelian randomization that genetically
elevated Lp(a) levels were associated with parental life span. High Lp(a) levels were
also shown to be associated with increased all-cause mortality (Patel et al. 2021;
Arsenault et al. 2020).
Multiple genetic studies and significant observations of a link between Lp(a) and
cardiovascular disease risk have been published (Mehta et al. 2020; Arsenault and
Kamstrup 2022). These focus on examining associations between Lp(a) and cardio-
vascular risk, CHD, lifespan, using genetic make-up, including family history, to
better understand genetic role of Lp(a) in ASCVD. The role of genetic variants
outside and within the KIV2 region of the LPA gene have been recently described.
There have been numerous variants proposed for CAD (Clarke et al. 2009). In stud-
ies by Clarke et al., a number of chromosomal regions were associated with risk of
CAD, and the LPA locus region 6q26–27 has the strongest relationship between
high Lp(a) levels and risk of CAD (Clarke et al. 2009). Various single nucleotide
polymorphisms (SNPs), rs10455872 and rs3798220 within the LPA site, have been
described and are highly associated with high Lp(a) levels. These variants are more
common in those of European ancestry. In the work of Kamstrup and Nordestgaard,
the genotypes mentioned above and high Lp(a) levels were associated with an
increased risk of heart failure, consistent with causal association (Kamstrup and
Nordestgaard 2016). More recently, the same authors have highlighted the effects of
Lp(a) on morbidity and mortality (Simony et al. 2022). Beyond the effect of specific
single nucleotide polymorphisms (SNPs), possible SNP-SNP interactions and SNPs
in the KIV-2 repeat region have to be taken into account, which might not be picked
up by conventional sequencing methods (Coassin and Kronenberg 2022). Work in
the cohort from Pakistan (Saleheen et al. 2017) at risk for myocardial infarction
showed additional SNPs and that both, smaller apo(a) isoform size and high Lp(a)
levels, are independent and causal risk factors for CAD. In studies of diverse
cohorts, such as the Atherosclerosis Risk in Communities (ARIC), Lp(a) measured
at middle age of participants was significantly associated with valvular and vascular
calcification at older age, represented by aortic valve calcium, mitral valve calcifi-
cation, and other factors (Obisesan et al. 2022). In this cohort, plasma Lp(a) levels
and family history of cardiovascular disease had independent and additive joint
associations with cardiovascular risk (Mehta et al. 2020). Another study, looking at
mostly males from Southeast Asia, found that Lp(a) levels in plasma are a positive
predictor of coronary artery disease and acute myocardial infarction (Loh
et al. 2022).
166 A. Matveyenko et al.
Conclusions
Lp(a) is an atherogenic lipoprotein present in human plasma with higher levels cor-
responding to an elevated risks for ASCVD. It has a strong genetic predisposition
and its mechanisms of action have been linked to the propagation of atherogenic
cascade via alteration of macrophage gene expression. Although there are currently
no widely prescribed Lp(a)-lowering treatments in the United States, there are avail-
able therapies, whose utility in clinical practice, needs to be further studied.
References
Afanasieva OI, Ezhov MV, Tmoyan NA, Razova OA, Afanasieva MI, Matchin YG, Pokrovsky
SN. Low molecular weight apolipoprotein(a) phenotype rather than lipoprotein(a) is asso-
ciated with coronary atherosclerosis and myocardial infarction. Front Cardiovasc Med.
2022;9:843602.
Arora P, Kalra R, Callas PW, Alexander KS, Zakai NA, Wadley V, Arora G, Kissela BM, Judd SE,
Cushman M. Lipoprotein(a) and risk of ischemic stroke in the REGARDS study. Arterioscler
Thromb Vasc Biol. 2019;39:810–8.
Arsenault BJ, Kamstrup PR. Lipoprotein(a) and cardiovascular and valvular diseases: a genetic
epidemiological perspective. Atherosclerosis. 2022;349:7–16.
Arsenault BJ, Pelletier W, Kaiser Y, Perrot N, Couture C, Khaw KT, Wareham NJ, Bosse Y, Pibarot
P, Stroes ESG, Mathieu P, Theriault S, Boekholdt SM. Association of long-term exposure to
elevated lipoprotein(a) levels with parental life span, chronic disease-free survival, and mortal-
ity risk: a mendelian randomization analysis. JAMA Netw Open. 2020;3:e200129.
Bennet A, Di Angelantonio E, Erqou S, Eiriksdottir G, Sigurdsson G, Woodward M, Rumley A,
Lowe GD, Danesh J, Gudnason V. Lipoprotein(a) levels and risk of future coronary heart dis-
ease: large-scale prospective data. Arch Intern Med. 2008;168:598–608.
Berg K. A new serum type system in man-the LP system. Acta Pathol Microbiol Scand.
1963;59:369–82.
Berliner JA, Watson AD. A role for oxidized phospholipids in atherosclerosis. N Engl J Med.
2005;353:9–11.
Boffa MB. Beyond fibrinolysis: the confounding role of Lp(a) in thrombosis. Atherosclerosis.
2022;349:72–81. https://pubmed.ncbi.nlm.nih.gov/35606079/.
Bourgeois R, Bourgault J, Despres AA, Perrot N, Guertin J, Girard A, Mitchell PL, Gotti C,
Bourassa S, Scipione CA, Gaudreault N, Boffa MB, Koschinsky ML, Pibarot P, Droit
A, Theriault S, Mathieu P, Bosse Y, Arsenault BJ. Lipoprotein proteomics and aortic valve
transcriptomics identify biological pathways linking lipoprotein(a) levels to aortic stenosis.
Metabolites. 2021;11:459.
Burgess S, Ference BA, Staley JR, Freitag DF, Mason AM, Nielsen SF, Willeit P, Young R,
Surendran P, Karthikeyan S, Bolton TR, Peters JE, Kamstrup PR, Tybjaerg-Hansen A, Benn M,
Langsted A, Schnohr P, Vedel-Krogh S, Kobylecki CJ, Ford I, Packard C, Trompet S, Jukema
JW, Sattar N, Di Angelantonio E, Saleheen D, Howson JMM, Nordestgaard BG, Butterworth
AS, Danesh J, European Prospective Investigation Into C and Nutrition-Cardiovascular
Disease C. Association of LPA variants with risk of coronary disease and the implications
for lipoprotein(a)-lowering therapies: a mendelian randomization analysis. JAMA Cardiol.
2018;3:619–27.
Chemello K, Chan DC, Lambert G, Watts GF. Recent advances in demystifying the metabolism of
lipoprotein(a). Atherosclerosis. 2022;349:82–91.
168 A. Matveyenko et al.
Clarke R, Peden JF, Hopewell JC, Kyriakou T, Goel A, Heath SC, Parish S, Barlera S, Franzosi
MG, Rust S, Bennett D, Silveira A, Malarstig A, Green FR, Lathrop M, Gigante B, Leander K,
de Faire U, Seedorf U, Hamsten A, Collins R, Watkins H, Farrall M. Genetic variants associ-
ated with Lp(a) lipoprotein level and coronary disease. N Engl J Med. 2009;361:2518–28.
Coassin S, Kronenberg F. Lipoprotein(a) beyond the kringle IV repeat polymorphism: the com-
plexity of genetic variation in the LPA gene. Atherosclerosis. 2022;349:17–35.
Colantonio LD, Bittner V, Safford MM, Marcovina S, Brown TM, Jackson EA, Li M, Lopez JAG,
Monda KL, Plante TB, Kent ST, Muntner P, Rosenson RS. Lipoprotein(a) and the risk for
coronary heart disease and ischemic stroke events among black and white adults with cardio-
vascular disease. J Am Heart Assoc. 2022;11:e025397.
Emerging Risk Factors Collaboration, Erqou S, Kaptoge S, Perry PL, Di Angelantonio E,
Thompson A, White IR, Marcovina SM, Collins R, Thompson SG, Danesh J. Lipoprotein(a)
concentration and the risk of coronary heart disease, stroke, and nonvascular mortality.
JAMA. 2009;302:412–23.
Enkhmaa BAE, Berglund L. Lipoprotein(a): impact by ethnicity and environmental and medical
conditions. J Lipid Res. 2016;57:1111–25.
Erqou S, Kaptoge S, Perry PL, Emerging Risk Factors Collaboration. Lipoprotein(a) con-
centration and the risk of coronary heart disease, stroke, and nonvascular mortality.
JAMA. 2009;302:412–23.
Forbang NI, Criqui MH, Allison MA, Ix JH, Steffen BT, Cushman M, Tsai MY. Sex and ethnic
differences in the associations between lipoprotein(a) and peripheral arterial disease in the
Multi-Ethnic Study of Atherosclerosis. J Vasc Surg. 2016;63:453–8.
Heigl F, Hettich R, Lotz N, Reeg H, Pflederer T, Osterkorn D, Osterkorn K, Klingel R. Efficacy,
safety, and tolerability of long-term lipoprotein apheresis in patients with LDL- or Lp(a)
hyperlipoproteinemia: Findings gathered from more than 36,000 treatments at one center in
Germany. Atheroscler Suppl. 2015;18:154–62.
Jawi MM, Frohlich J, Chan SY. Lipoprotein(a) the insurgent: a new insight into the structure, func-
tion, metabolism, pathogenicity, and medications affecting lipoprotein(a) molecule. J Lipids.
2020;2020:3491764.
Kaiser Y, Daghem M, Tzolos E, Meah MN, Doris MK, Moss AJ, Kwiecinski J, Kroon J,
Nurmohamed NS, van der Harst P, Adamson PD, Williams MC, Dey D, Newby DE, Stroes
ESG, Zheng KH, Dweck MR. Association of lipoprotein(a) with atherosclerotic plaque pro-
gression. J Am Coll Cardiol. 2022;79:223–33.
Kamstrup PR, Nordestgaard BG. Elevated lipoprotein(a) levels, LPA risk genotypes, and increased
risk of heart failure in the general population. JACC Heart Fail. 2016;4:78–87.
Kamstrup PR, Benn M, Tybjaerg-Hansen A, Nordestgaard BG. Extreme lipoprotein(a) levels and
risk of myocardial infarction in the general population: the Copenhagen City Heart Study.
Circulation. 2008;117:176–84.
Kettunen J, Demirkan A, Wurtz P, Draisma HH, Haller T, Rawal R, Vaarhorst A, Kangas AJ,
Lyytikainen LP, Pirinen M, Pool R, Sarin AP, Soininen P, Tukiainen T, Wang Q, Tiainen M,
Tynkkynen T, Amin N, Zeller T, Beekman M, Deelen J, van Dijk KW, Esko T, Hottenga JJ, van
Leeuwen EM, Lehtimaki T, Mihailov E, Rose RJ, de Craen AJ, Gieger C, Kahonen M, Perola
M, Blankenberg S, Savolainen MJ, Verhoeven A, Viikari J, Willemsen G, Boomsma DI, van
Duijn CM, Eriksson J, Jula A, Jarvelin MR, Kaprio J, Metspalu A, Raitakari O, Salomaa V,
Slagboom PE, Waldenberger M, Ripatti S, Ala-Korpela M. Genome-wide study for circulat-
ing metabolites identifies 62 loci and reveals novel systemic effects of LPA. Nat Commun.
2016;7:11122.
Kobiyama K, Ley K. Atherosclerosis. Circ Res. 2018;123:1118–20.
Kosmas CE, Silverio D, Sourlas A, Peralta R, Montan PD, Guzman E, Garcia MJ. Role of
lipoprotein(a) in peripheral arterial disease. Ann Transl Med. 2019;7:S242.
Kostner KM, Kostner GM. Lipoprotein(a): a historical appraisal. J Lipid Res. 2017;58:1–14.
Kraaijenhof JM, Hovingh GK, Stroes ESG, Kroon J. The iterative lipid impact on inflammation in
atherosclerosis. Curr Opin Lipidol. 2021;32:286–92.
9 The Role of Lp(a) in Atherosclerosis: An Overview 169
Kraft HGSC, Menzel HJ, Utermann G. Apolipoprotein(a) alleles determine lipoprotein(a) particle
density and concentration in plasma. Atheroscler Thromb. 1992;12:302–6.
Kronenberg F. Lipoprotein(a). Handb Exp Pharmacol. 2022;270:201–32.
Kronenberg F, Utermann G. Lipoprotein(a): resurrected by genetics. J Intern Med. 2013;273:6–30.
Lackner C, Boerwinkle E, Leffert CC, Rahmig T, Hobbs HH. Molecular basis of apolipoprotein(a)
isoform size heterogeneity as revealed by pulsed-field get electrophoresis. J Clin Invest.
1991;87:2077–86.
Langsted A, Varbo A, Kamstrup PR, Nordestgaard BG. Elevated lipoprotein(a) does not cause low-
grade inflammation despite causal association with aortic valve stenosis and myocardial infarc-
tion: a study of 100,578 individuals from the general population. J Clin Endocrinol Metab.
2015;100:2690–9.
Langsted A, Kamstrup PR, Nordestgaard BG. High lipoprotein(a) and high risk of mortality. Eur
Heart J. 2019;40:2760–70.
Libby P. Mechanisms of acute coronary syndromes and their implications for therapy. N Engl J
Med. 2013;368:2004–13.
Libby P, Buring JE, Badimon L, Hansson GK, Deanfield J, Bittencourt MS, Tokgozoglu L, Lewis
EF. Atherosclerosis. Nat Rev Dis Primers. 2019;5:56.
Loh WJ, Chang X, Aw TC, Phua SK, Low AF, Chan MY, Watts GF, Heng CK. Lipoprotein(a) as
predictor of coronary artery disease and myocardial infarction in a multi-ethnic Asian popula-
tion. Atherosclerosis. 2022;349:160–5.
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer
GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen
TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O, Group
ESCSD. ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to
reduce cardiovascular risk. Eur Heart J. 2019;2020(41):111–88.
Marchini T, Mitre LS, Wolf D. Inflammatory cell recruitment in cardiovascular disease. Front Cell
Dev Biol. 2021;9:635527.
Markus MRP, Ittermann T, Schipf S, Bahls M, Nauck M, Volzke H, Santos RD, Peters A, Zeller T,
Felix SB, Vasan RS, Thorand B, Steinhagen-Thiessen E, Dorr M. Association of sex-specific
differences in lipoprotein(a) concentrations with cardiovascular mortality in individuals with
type 2 diabetes mellitus. Cardiovasc Diabetol. 2021;20:168.
McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors.
Pathology. 2018; https://doi.org/10.1016/j.pathol.2018.11.003.
Mehta A, Virani SS, Ayers CR, Sun W, Hoogeveen RC, Rohatgi A, Berry JD, Joshi PH, Ballantyne
CM, Khera A. Lipoprotein(a) and family history predict cardiovascular disease risk. J Am Coll
Cardiol. 2020;76:781–93. PMID: 32792075.
Moriarty PM, Gray JV, Gorby LK. Lipoprotein apheresis for lipoprotein(a) and cardiovascular
disease. J Clin Lipidol. 2019;13:894–900.
Nahrendorf M, Pittet MJ, Swirski FK. Monocytes: protagonists of infarct inflammation and repair
after myocardial infarction. Circulation. 2010;121:2437–45.
Nordestgaard BG, Langsted A. Lipoprotein(a) as a cause of cardiovascular disease: insights from
epidemiology, genetics, and biology. J Lipid Res. 2016;57:1953–75.
Nordestgaard BG, Chapman MJ, Ray K, Borén J, Andreotti F, Watts GF, Ginsberg H, Amarenco
P, Catapano A, Descamps OS, Fisher E, Kovanen PT, Lesnik P, Masana L, Reiner Z, Taskinen
M-R, Tokgözoglu L, Tybjærg-Hansen A, Panel aEASC. Lipoprotein(a) as a cardiovascular risk
factor: current status. Eur Heart J. 2010;31:2844–53.
Norgren L, Hiatt WR, Dormandy JA, Nehler MR, Harris KA, Fowkes FG, Group TIW. Inter-
society consensus for the management of peripheral arterial disease (TASC II). J Vasc Surg.
2007;45(Suppl S):S5–67.
Nugent AKGJ, Gorby LK, Moriarty PM. Lipoprotein apheresis: first FDA indicated treatment for
elevated lipoprotein(a). J Clin Cardiol. 2020;1:16–21.
Obisesan OH, Kou M, Wang FM, Boakye E, Honda Y, Uddin SMI, Dzaye O, Osei AD, Orimoloye
OA, Howard-Claudio CM, Coresh J, Blumenthal RS, Hoogeveen RC, Budoff MJ, Matsushita
K, Ballantyne CM, Blaha MJ. Lipoprotein(a) and subclinical vascular and valvular calcifica-
170 A. Matveyenko et al.
Simony SB, Mortensen MB, Langsted A, Afzal S, Kamstrup PR, Nordestgaard BG. Sex dif-
ferences of lipoprotein(a) levels and associated risk of morbidity and mortality by age: The
Copenhagen General Population Study. Atherosclerosis. 2022;355:76–82.
Sniderman AD, Thanassoulis G, Glavinovic T, Navar AM, Pencina M, Catapano A, Ference
BA. Apolipoprotein B particles and cardiovascular disease: a narrative review. JAMA Cardiol.
2019;4:1287–95.
Stefanutti C, Pisciotta L, Favari E, Di Giacomo S, Vacondio F, Zenti MG, Morozzi C, Berretti D,
Mesce D, Vitale M, Pasta A, Ronca A, Garuti A, Manfredini M, Angles-Cano E, Marcovina
SM, Watts GF, Multidisciplinary International Group for Hemapheresis TherapY and MEDIC.
ORG MEDC-M. Lipoprotein(a) concentration, genetic variants, apo(a) isoform size, and cel-
lular cholesterol efflux in patients with elevated Lp(a) and coronary heart disease submitted or
not to lipoprotein apheresis: an Italian case-control multicenter study on Lp(a). J Clin Lipidol.
2020;14:487–497.e1.
Syrovets T, Thillet J, Chapman MJ, Simmet T. Lipoprotein(a) is a potent chemoattractant for
human peripheral monocytes. Blood. 1997;90:2027–36.
Tada H, Takamura M, Kawashiri MA. Lipoprotein(a) as an old and new causal risk factor of ath-
erosclerotic cardiovascular disease. J Atheroscler Thromb. 2019;26:583–91.
Tmoyan NA, Ezhov MV, Afanasieva OI, Klesareva EA, Razova OA, Kukharchuk VV, Pokrovsky
SN. The association of lipoprotein(a) and apolipoprotein(a) phenotypes with peripheral artery
disease. Ter Arkh. 2018;90:31–6.
Tokgözoğlu L, Libby P. The dawn of a new era of targeted lipid-lowering therapies. Eur Heart
J. 2022;43(34):3198–208. PMID: 35051271.
Tomoi Y, Takahara M, Soga Y, Kodama K, Imada K, Hiramori S, Ando K. Impact of high
lipoprotein(a) levels on clinical outcomes following peripheral endovascular therapy. JACC
Cardiovasc Interv. 2022;15:1466–76.
Tsimikas S. A test in context: lipoprotein(a): diagnosis, prognosis, controversies, and emerging
therapies. J Am Coll Cardiol. 2017;69:692–711.
Tybjaerg-Hansen A. Using human genetics to predict the effects and side effects of lipoprotein(a)-
lowering drugs. J Am Coll Cardiol. 2016;68:2773–5.
Utermann G. The mysteries of lipoprotein(a). Science. 1989;246:904–10.
Utermann G. Genetic architecture and evolution of the lipoprotein(a) trait. Curr Opin Lipidol.
1999;10:133–41.
van Capelleveen JC, van der Valk FM, Stroes ES. Current therapies for lowering lipoprotein(a). J
Lipid Res. 2016;57:1612–8.
Waldmann E, Parhofer KG. Apheresis for severe hypercholesterolaemia and elevated lipoprotein(a).
Pathology. 2019;51:227–32.
Youssef A, Clark JR, Marcovina SM, Boffa MB, Koschinsky ML. Apo(a) and ApoB interact non-
covalently within hepatocytes: implications for regulation of Lp(a) levels by modulation of
ApoB secretion. Arterioscler Thromb Vasc Biol. 2022;42:289–304.
Zheng Z, Nakamura K, Gershbaum S, Wang X, Thomas S, Bessler M, Schrope B, Krikhely A,
Liu RM, Ozcan L, Lopez JA, Tabas I. Interacting hepatic PAI-1/tPA gene regulatory pathways
influence impaired fibrinolysis severity in obesity. J Clin Invest. 2020;130:4348–59.
Zhiduleva EV, Irtyuga OB, Shishkova AA, Ignat’eva EV, Kostina AS, Levchuk KA, Golovkin AS,
Rylov AY, Kostareva AA, Moiseeva OM, Malashicheva AB, Gordeev ML. Cellular mecha-
nisms of aortic valve calcification. Bull Exp Biol Med. 2018;164:371–5.
Chapter 10
Molecular Mechanisms of Lipoprotein(a)
Pathogenicity: Tantalizing Clues
and Unanswered Questions
Introduction
Although lipoprotein(a) (Lp(a)) was discovered almost 50 years ago (Berg and New
1963) and has been subsequently shown to be a causal and independent risk factor
for atherosclerotic cardiovascular disease (ASCVD) and calcific aortic valve dis-
ease (CAVD) (Arsenault and Kamstrup 2022), the mechanisms by which Lp(a)
mediates its pathogenic effects in vivo remain unclear. Lp(a) comprises an
apoB-100-containing lipoprotein to which is attached the unique apolipoprotein(a)
(apo(a)) moiety (Fig. 10.1). Amino acid analysis followed by complete sequencing
of the human apo(a) cDNA in 1987 revealed a high level of sequence identity with
the profibrinolytic enzyme plasminogen (Eaton et al. 1987; McLean et al. 1987).
Apo(a) contains a series of tri-looped structures called kringles that are similar to
the KIV domain of plasminogen, followed by sequences similar to the plasminogen
KV and protease domains (Fig. 10.1). Due to several critical amino acid substitu-
tions and a small deletion, the apo(a) protease-like domain has been shown to be
catalytically inactive (Gabel and Koschinsky 1995).
M. B. Boffa (*)
Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of
Western Ontario, London, ON, Canada
Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of
Western Ontario, London, ON, Canada
e-mail: [email protected]
M. L. Koschinsky
Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of
Western Ontario, London, ON, Canada
Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of
Western Ontario, London, ON, Canada
e-mail: [email protected]
Fig. 10.1 Structure and functional domains of Lp(a). Lp(a) consists of apo(a) covalently linked to
the apoB-100 moiety of an LDL-like lipoprotein particle. The lipid portion of the particle is a shell
of phospholipids (PL) and free cholesterol (FC) surrounding a neutral lipid core of cholesteryl
esters (CE) and triacylglycerols (TG). Apo(a) consists of ten types of KIV domains, a KV domain,
and an inactive protease domain. KIV2 is repeated different numbers of times in different apo(a)
isoforms. KIV5–KIV8 contain weak lysine-binding sites (wLBS), with those in KIV7 and KIV8
binding to specific lysine residues in apoB-1000 during the noncovalent step of Lp(a) assembly.
KIV9 contains a single-free cysteine that mediates disulfide bond formation with apoB-100.
KIV10 contains a strong lysine-binding site (sLBS) as well as covalently bound oxidized phospho-
lipid (OxPL). The sLBS is required for OxPL addition, and together these features promote several
pathogenic effects on vascular and immune cells. OxPL is also present noncovalently associated
with the lipid moiety of Lp(a), and accounts for up to 50% of the total OxPL on Lp(a). EC endo-
thelial cell, SMC smooth muscle cell, VIC valve interstitial cell
10 Molecular Mechanisms of Lipoprotein(a) Pathogenicity: Tantalizing Clues… 175
Apo(a) kringle IV sequences are present in ten types based on amino acid
sequence; these have been designated KIV1–KIV10 (McLean et al. 1987; van der
Hoek et al. 1993). The KIV2 sequence is present in a variable number of identically
repeated copies (from 3 to greater than 40) which is a hallmark of Lp(a) and reflects
allele size variation in LPA, the gene encoding apo(a) (Fig. 10.1) (Lackner et al.
1993; Marcovina et al. 1996). Interestingly, there is a strong inverse correlation
between apo(a) size and Lp(a) plasma levels, which likely arises due to less efficient
secretion of larger isoforms as a result of presecretory degradation of misfolded spe-
cies in the endoplasmic reticulum (Boffa and Koschinsky 2022). The KIV9 domain
houses the only unpaired cysteine in apo(a) and is involved in disulfide bond forma-
tion with a cysteine residue in the carboxyl-terminus of apoB-100 (Koschinsky
et al. 1993). The KIV5–8 domains each contain a weak lysine-binding site (wLBS);
the wLBS in KIV7 and KIV8 is required for intracellular noncovalent interaction
between apo(a) and apoB that precedes extracellular disulfide bond formation
(Fig. 10.1) (Becker et al. 2004; Youssef et al. 2022).
The apo(a) KIV10 domain contains a relatively strong lysine-binding site (sLBS)
that has been studied extensively in attempts to understand the pathophysiology of
Lp(a) in the vasculature. Lp(a) has been demonstrated to be the preferential lipopro-
tein carrier of proinflammatory oxidized phospholipids (OxPL), compared to LDL
(Bergmark et al. 2008). These species are present both on the lipid portion of Lp(a)
as well as covalently associated with apo(a) (Bergmark et al. 2008; Leibundgut
et al. 2013). Interestingly, in this regard, it has been shown that the KIV10 sLBS is
absolutely required for the covalent addition of oxidized phospholipid to this krin-
gle, likely involving addition of the OxPL adduct to a histidine side chain through
Michael reaction addition (Fig. 10.1) (Leibundgut et al. 2013; Scipione et al. 2015).
The proinflammatory effect of the OxPL on KIV10 has been demonstrated in many
studies, both in vitro and in vivo (Koschinsky and Boffa 2022; Dzobo et al. 2022).
In vitro studies have shown the role of OxPL on KIV10 in promoting proinflamma-
tory and phenotypes in a variety of vascular and inflammatory cells including valve
interstitial cells (VICs) (see below).
Many studies using a variety of vascular cell types have shown that the apo(a)
sLBS can compete with plasminogen for binding to cell surfaces, thereby inhibiting
plasminogen activation to the active enzyme plasmin (Boffa 2022). Downstream
effects on fibrin clot lysis have also been studied, with variable results, and the sig-
nificance of Lp(a) in promoting thrombosis in the arterial and venous circulation
remains controversial (Boffa 2022). Indeed, Lp(a) appears to confer risk for venous
thromboembolism only in individuals with extremely high Lp(a) levels (Kamstrup
et al. 2012). The role of Lp(a) in platelet function and coagulation, and in the lysis
of platelet-rich clots, is not clear (Boffa 2022). Importantly, it is difficult to assess
the contribution of Lp(a) to lysis of clots formed upon rupture of vulnerable athero-
sclerotic plaques (see below).
Despite the demonstration of elevated plasma Lp(a) levels as an independent and
causal risk factor for ASCVD and CAVD, the mechanism of action of Lp(a) in these
disease processes remains unclear. This reflects, in part, the complexity of the Lp(a)
structure, as well as the lack of suitable animal models for Lp(a); together these
176 M. B. Boffa and M. L. Koschinsky
Fig. 10.2 Overlapping pathogenic mechanisms of Lp(a) in atherosclerosis and calcific aortic
valve disease. There are several common mechanisms mediated by Lp(a) in the two disorders.
Compromised endothelial cell function leads to barrier permeability, infiltration of Lp(a) and
monocytes, expression of endothelial cell surface receptors for monocytes, and mural thromboses.
Lp(a) activates monocytes leading to cytokine secretion and enhanced potential for transendothe-
lial migration. Within the vessel wall or valve, Lp(a) promotes macrophage foam cell formation
and macrophage apoptosis, as well as stimulating the release of proinflammatory cytokines such as
interleukin-8 (IL-8) from macrophages. Lp(a) promotes smooth muscle cell (SMC) migration and
proliferation in the arterial intima. Lp(a) also promotes calcification of SMC in the atrial intima
and osteogenic differentiation and calcification of valve interstitial cells. Many functions of Lp(a),
indicated in red, are mediated by its bound oxidized phospholipid (OxPL). Lp(a) also transports
the phospholipase D enzyme autotaxin into the aortic valve leaflet, where it catalyzed generation
of the highly proinflammatory lysophosphatidic acid (lysoPA) using lysophosphatidylcholine
(lysoPC) as a substrate. The OxPL on Lp(a) thus helps to explain why elevated Lp(a) is a causal
risk factor for both atherothrombosis and aortic stenosis, despite the disease processes underlying
each of these disorders being distinct. TNF-α tumor necrosis factor-α
10 Molecular Mechanisms of Lipoprotein(a) Pathogenicity: Tantalizing Clues… 177
A number of early in vitro studies suggested that Lp(a) could contribute to cul-
tured vascular SMC migration and proliferation through the ability of apo(a) to
inhibit the plasmin-dependent activation of TGF-β (Fig. 10.2) (Grainger et al.
1993, 1994; O’Neil et al. 2004). More recently, the ability of OxPL on apo(a)
KIV10 to stimulate the expression of Klf-4, an important factor in phenotypic
switching of SMCs in atherosclerotic lesions, was attributed to the apo(a)-medi-
ated activation of the long noncoding RNA MIAT (Fig. 10.2) (Fasolo et al. 2021).
Lp(a) has also been implicated in the calcification of human aortic SMCs through
Notch1-NFκB and Notch1-BMP2-Smad1/5/9 pathways (Peng et al. 2022). The
Notch1-NFκB pathway resulted in increased osteopontin and inflammatory cyto-
kine expression, while Lp(a)-mediated Notch1-BMP2-Smad1/5/9 activation also
contributed to calcification of the cells. The ability of Lp(a) to increase VSMC
calcification is another mechanism by which Lp(a) contributes to vascular disease
(Fig. 10.2).
Recent studies have demonstrated a key role for Lp(a) in both the development and
progression of aortic valve disease (Thanassoulis et al. 2013; Capoulade et al. 2015).
This is likely mediated, in large part, by the ability of the OxPL component of
apo(a) to modify the phenotype of valve interstitial cells to resulting in proinflam-
matory and pro-osteogenic responses in these cells (Fig. 10.2). In a recent study by
Zheng and coworkers, treatment of VICs by Lp(a) or recombinant apo(a) resulted in
osteogenic differentiation in these cells through the induction of IL6, BMP2, and
RUNX2 expression (Zheng et al. 2019). The effects were attributed to the OxPL on
Lp(a) and apo(a): the anti-oxPL antibody E06 blocked the effects of Lp(a) as did
mutation of the KIV10 LBS which significantly reduced the effect of apo(a) (Zheng
et al. 2019). OxPLs transported by Lp(a) also increase the load of reactive oxygen
species in the aortic valve, loading to ROS-mediated activation of the NFκB path-
way, and induction of a program of inflammatory gene expression (Bouchareb et al.
2015; Mathieu et al. 2017). Additionally, Lp(a) binds to autotaxin and delivers it to
valves (Bouchareb et al. 2015; Mathieu et al. 2017); autotaxin promotes inflamma-
tion and osteogenic transdifferentiation of VICs through the production of LysoPA
which in turn binds and signals through the lysoPA receptor (Fig. 10.2). Taken
together, these studies suggest that Lp(a) can initiate a program of inflammation and
osteoblastic differentiation in valvular interstitial cells which is a major contributing
factor to AVS and CAVD. Lp(a) could also promote CAVD through promotion of
valve endothelial cell dysfunction, immune cell infiltration, and foam cell formation
(Fig. 10.2).
Fig. 10.3 Effects on imbalance between coagulation (formation of fibrin by thrombin cleavage of
fibrinogen) and fibrinolysis (degradation of insoluble fibrin into soluble fibrin degradation prod-
ucts) can cause thrombosis. Lp(a) and apo(a) promote this imbalance by favoring coagulation
(green mechanisms) and impeding fibrinolysis (red mechanisms). TFPI tissue factor pathway
inhibitor
and mouse pulmonary embolism models (Biemond et al. 1997; Palabrica et al.
1995); notably, however, Lp(a) itself was not tested in these studies. Indeed, the
available data from human epidemiological and genetic studies do not provide evi-
dence for a direct antifibrinolytic/prothrombotic impact of elevated Lp(a) (Boffa
2022). Moreover, in a recent study using subjects undergoing Lp(a) lowering with
antisense oligonucleotide therapy, we found that despite dramatic reductions in
plasma Lp(a) concentrations, ex vivo plasma clot lysis time was not affected (Boffa
et al. 2019). Furthermore, recombinant apo(a) had a potent antifibrinolytic effect
whereas Lp(a) purified from human plasma lacked this effect (Boffa et al. 2019).
Against this backdrop, we summarize below the clinical evidence with respect to
Lp(a) and thrombosis and thrombolysis, and we outline areas for future studies of
this issue.
risk factor. With the advent of genetic approaches to study the association of geneti-
cally elevated Lp(a) with disease—including Mendelian randomization studies—
the opportunity to assess a causal role for elevated Lp(a) in VTE and to eliminate
confounding variables has arisen. Several large studies using genetic approaches
have been published. All showed that genetically inherited elevated Lp(a) or geneti-
cal markers of high Lp(a) were not significant predictors of VTE (Kamstrup et al.
2012; Helgadottir et al. 2012; Danik et al. 2013; Larsson et al. 2020). Importantly,
in several of these studies, a causal role for elevated Lp(a) in the development of
atherothrombotic disorders was observed in the same population (Kamstrup et al.
2012; Helgadottir et al. 2012; Larsson et al. 2020). However, a posthoc observa-
tional study of one of these populations found that extremely high Lp(a) levels
(≥95th percentile) were significantly associated with VTE (Kamstrup et al. 2012).
A recent retrospective study of pulmonary embolism patients found no correlation
between Lp(a) levels and the severity of pulmonary embolism (Gressenberger
et al. 2022).
The general lack of association between elevated Lp(a) and risk for VTE (except
for extremely high Lp(a) concentrations) is consistent with a minimal or absent
antifibrinolytic ability of Lp(a). Venous thrombi are fibrin- and erythrocyte-rich and
form as a consequence of blood stasis, hypercoagulability, and endothelial damage.
Thrombi in the arterial tree are platelet-rich and fibrin-poor, and generally form as a
sequela of atherosclerotic plaque erosion or rupture. Thus, mechanistic implications
of associations between Lp(a) levels and ASCVD endpoints are confounded by the
possibility that Lp(a) may contribute to atherosclerosis, thrombosis, or both.
Interestingly, a consistent observation (albeit from relatively small sample sizes)
has been the association between elevated Lp(a) levels and risk of ischemic stroke
in children (Nowak-Gottl et al. 1999; Strater et al. 2002; Kenet et al. 2010;
Goldenberg et al. 2013). These events are frequently seen in patients with inherited
dispositions toward thrombophilia such as Factor V Leiden. That the events would
occur in the absence of underlying atherosclerosis are clear from the young age of
the patients, and this may speak to a prothrombotic or antifibrinolytic effect of Lp(a).
Early studies examined the proposition that elevated Lp(a) could reduce the effi-
cacy of thrombolytic therapy. Most of these occurred in the setting of acute myocar-
dial infarction (MBewu et al. 1994; Tranchesi et al. 1991; Armstrong et al. 1990;
von Hodenberg et al. 1991; Brugemann et al. 1994), although one examined isch-
emic stroke (Ribo et al. 2004). None of these studies showed that Lp(a) levels are a
significant predictor of successful recanalization, although all of them were limited
by small sample sizes (and thus few patients with high Lp(a)) and/or having sam-
pled blood for Lp(a) measurement in the postinfarct period where the acute phase
response may have increased Lp(a) levels (Boffa 2022).
Taken together, the jury is still out on whether Lp(a) inhibits fibrinolysis
in vivo. Further studies in animal models may be required to assess this question,
and further assessment of the impact of Lp(a) on thrombolytic therapy in the set-
ting of ischemic stroke, deep vein thrombosis, and pulmonary embolism may be
warranted.
182 M. B. Boffa and M. L. Koschinsky
Elevated Lp(a) is clearly and consistently associated with ASCVD events, though
less so with intermediate phenotypes such as intimal-medial thickness and coronary
calcium scores (Kivimaki et al. 2011; Raitakari et al. 1999; Razavi et al. 2021;
Mehta et al. 2022). This can be interpreted to mean that Lp(a) plays a more impor-
tant role in precipitating atherothrombotic events, rather than in promoting the
underlying atherosclerotic process. Two scenarios can be contemplated.
1. Lp(a) could be promoting thrombus formation directly through an impact on the
coagulation system or on platelet activation (Fig. 10.3). Little to no data on an
effect of Lp(a) on coagulation have been published, although early studies
showed that Lp(a) could exert a procoagulant effect by binding and inhibiting
tissue factor pathway inhibitor (Fig. 10.3) (Caplice et al. 2001). We and others
have shown that Lp(a) and apo(a) can impact fibrin clot structure, leading to a
form resistant to lysis (Scipione et al. 2017; Skuza et al. 2019; Rowland et al.
2014) (Fig. 10.3); we also demonstrated that apo(a) could accelerate the rate of
fibrin formation, which could also impact clot structure (Scipione et al. 2017).
Lp(a) does bind to platelets (Ezratty et al. 1993; Martinez et al. 2001), and Lp(a)
and apo(a) have been shown increase the proaggregant effect of certain agonists
(such as the protease-activated receptor-1 ligand peptide SFLLRN and arachi-
donic acid) in washed platelets (Martinez et al. 2001; Rand et al. 1998). However,
two recent studies in platelet-rich plasma showed that Lp(a) level did not predict
the aggregation response to several agonists including ADP, collagen, or arachi-
donic acid (Salsoso et al. 2020; Kille et al. 2021).
2. Lp(a) could be promoting a “vulnerable” plaque phenotype with a greater pro-
pensity to rupture and hence cause an atherothrombotic event (Fig. 10.3).
Consistent with this scenario, it was reported in a carotid ultrasound study that
Lp(a) level predicted the extent of stenosis but not total plaque area (Klein et al.
2008); the extent of stenosis could be interpreted to reflect ongoing rupture and
thrombus formation. The proinflammatory effects of Lp(a) owing to its OxPL
could result in a larger necrotic core and/or a thinner fibrous cap, both hallmarks
of rupture-prone plaques. Very little direct study of this question has been
attempted. In an immunohistochemical study of human coronary atherosclerotic
plaques of varying phenotypes, apo(a) immunostaining was found in proximity
to oxidation-specific epitopes—such as the OxPL recognized by E06—specifi-
cally in vulnerable or ruptured plaques (van Dijk et al. 2012).
It is clear from the above that, while some evidence for Lp(a)-promoting vul-
nerable plaque and/or arterial thrombosis exists, more research is necessary. This
will require both work in animal models, such as transgenic mice expressing
human Lp(a), as well as more imaging and biomarker studies in human patients.
Such research is important because it may help to stratify risk in patients with high
Lp(a) to identify those who would most benefit from emerging Lp(a)-lowering
therapies.
10 Molecular Mechanisms of Lipoprotein(a) Pathogenicity: Tantalizing Clues… 183
Concluding Remarks
These are exciting times in the Lp(a) field, with burgeoning interest in this causal
risk factor for CVD on from both basic researchers and clinicians. The quantitative
importance of elevated Lp(a) as a risk factor is now widely accepted, although at
present clinical adoption of Lp(a) measurement has lagged because of a lack, cur-
rently, of Lp(a)-lowering therapies. With Phase III cardiovascular outcomes trials in
progress on an antisense oligonucleotide-targeting LPA mRNA, we are potentially
on the cusp of having an effective treatment for lowering Lp(a) as well as definitive
proof that elevated Lp(a) is harmful. At the same time, our understanding of the
pathogenic mechanisms of Lp(a) continues to expand, with the role of Lp(a) as a
proinflammatory mediator emerging as a key factor. Further studies of these mecha-
nisms may lead to an alternative therapeutic strategy to mitigate the effect of ele-
vated Lp(a)—interference with its pathogenic effects in the vasculature.
References
Allen S, Khan S, Tam S, et al. Expression of adhesion molecules by lp(a): a potential novel mecha-
nism for its atherogenicity. FASEB J. 1998;12:1765–76.
Armstrong VW, Neubauer C, Schutz E, et al. Lack of association between raised serum Lp(a)
concentration and unsuccessful thrombolysis after acute myocardial infarction. Lancet.
1990;336:1077.
Arsenault BJ, Kamstrup PR. Lipoprotein(a) and cardiovascular and valvular diseases: a genetic
epidemiological perspective. Atherosclerosis. 2022;349:7–16.
Becker L, Cook PM, Wright TG, et al. Quantitative evaluation of the contribution of weak lysine-
binding sites present within apolipoprotein(a) kringle IV types 6-8 to lipoprotein(a) assembly.
J Biol Chem. 2004;279:2679–88.
Berg K, New A. Serum type system in man–the Lp system. Acta Pathol Microbiol Scand.
1963;59:369–82.
Bergmark C, Dewan A, Orsoni A, et al. A novel function of lipoprotein [a] as a preferential carrier
of oxidized phospholipids in human plasma. J Lipid Res. 2008;49:2230–9.
Biemond BJ, Friederich PW, Koschinsky ML, et al. Apolipoprotein(a) attenuates endogenous fibri-
nolysis in the rabbit jugular vein thrombosis model in vivo. Circulation. 1997;96:1612–5.
Boffa MB. Beyond fibrinolysis: the confounding role of Lp(a) in thrombosis. Atherosclerosis.
2022;349:72–81.
Boffa MB, Koschinsky ML. Lipoprotein (a): truly a direct prothrombotic factor in cardiovascular
disease? J Lipid Res. 2016;57:745–57.
Boffa MB, Koschinsky ML. Understanding the ins and outs of lipoprotein (a) metabolism. Curr
Opin Lipidol. 2022;33:185–92.
Boffa MB, Marar TT, Yeang C, et al. Potent reduction of plasma lipoprotein (a) with an anti-
sense oligonucleotide in human subjects does not affect ex vivo fibrinolysis. J Lipid Res.
2019;60:2082–9.
Bouchareb R, Mahmut A, Nsaibia MJ, et al. Autotaxin derived from lipoprotein(a) and valve
interstitial cells promotes inflammation and mineralization of the aortic valve. Circulation.
2015;132:677–90.
Brugemann J, van der Meer J, Hillege HL, et al. Lipoprotein(a) levels in patients with myocardial
infarction treated with anistreplase: no prediction of efficacy but inverse correlation with plas-
minogen activation in non-patency. Int J Cardiol. 1994;45:109–13.
184 M. B. Boffa and M. L. Koschinsky
Caplice NM, Panetta C, Peterson TE, et al. Lipoprotein (a) binds and inactivates tissue factor
pathway inhibitor: a novel link between lipoproteins and thrombosis. Blood. 2001;98:2980–7.
Capoulade R, Chan KL, Yeang C, et al. Oxidized phospholipids, lipoprotein(a), and progression of
calcific aortic valve stenosis. J Am Coll Cardiol. 2015;66:1236–46.
Cho T, Jung Y, Koschinsky ML. Apolipoprotein(a), through its strong lysine-binding site in
KIV(10′), mediates increased endothelial cell contraction and permeability via a Rho/Rho
kinase/MYPT1-dependent pathway. J Biol Chem. 2008;283:30503–12.
Cho T, Romagnuolo R, Scipione C, et al. Apolipoprotein(a) stimulates nuclear translocation of
beta-catenin: a novel pathogenic mechanism for lipoprotein(a). Mol Biol Cell. 2013;24:210–21.
Danik JS, Buring JE, Chasman DI, et al. Lipoprotein(a), polymorphisms in the LPA gene, and
incident venous thromboembolism among 21483 women. J Thromb Haemost. 2013;11:205–8.
Dzobo KE, Kraaijenhof JM, Stroes ESG, et al. Lipoprotein(a): an underestimated inflammatory
mastermind. Atherosclerosis. 2022;349:101–9.
Eaton DL, Fless GM, Kohr WJ, et al. Partial amino acid sequence of apolipoprotein(a) shows that
it is homologous to plasminogen. Proc Natl Acad Sci U S A. 1987;84:3224–8.
Edelstein C, Pfaffinger D, Hinman J, et al. Lysine-phosphatidylcholine adducts in kringle V impart
unique immunological and potential pro-inflammatory properties to human apolipoprotein(a).
J Biol Chem. 2003;278:52841–7.
Ezratty A, Simon DI, Loscalzo J. Lipoprotein(a) binds to human platelets and attenuates plasmino-
gen binding and activation. Biochemistry. 1993;32:4628–33.
Fasolo F, Jin H, Winski G, et al. Long noncoding RNA MIAT controls advanced atherosclerotic
lesion formation and plaque destabilization. Circulation. 2021;144:1567–83.
Gabel BR, Koschinsky MI. Analysis of the proteolytic activity of a recombinant form of
apolipoprotein(a). Biochemistry. 1995;34:15777–84.
Goldenberg NA, Bernard TJ, Hillhouse J, et al. Elevated lipoprotein (a), small apolipoprotein
(a), and the risk of arterial ischemic stroke in North American children. Haematologica.
2013;98:802–7.
Grainger DJ, Kirschenlohr HL, Metcalfe JC, et al. Proliferation of human smooth muscle cells
promoted by lipoprotein(a). Science. 1993;260:1655–8.
Grainger DJ, Kemp PR, Liu AC, et al. Activation of transforming growth factor-beta is inhibited in
transgenic apolipoprotein(a) mice. Nature. 1994;370:460–2.
Gressenberger P, Posch F, Pechtold M, et al. Lipoprotein(a) and pulmonary embolism severity-a
retrospective data analysis. Front Cardiovasc Med. 2022;9:808605.
Hajjar KA, Gavish D, Breslow JL, et al. Lipoprotein(a) modulation of endothelial cell surface
fibrinolysis and its potential role in atherosclerosis. Nature. 1989;339:303–5.
Hancock MA, Boffa MB, Marcovina SM, et al. 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;278:23260–9.
Haque NS, Zhang X, French DL, et al. CC chemokine I-309 is the principal monocyte che-
moattractant induced by apolipoprotein(a) in human vascular endothelial cells. Circulation.
2000;102:786–92.
Helgadottir A, Gretarsdottir S, Thorleifsson G, et al. Apolipoprotein(a) genetic sequence vari-
ants associated with systemic atherosclerosis and coronary atherosclerotic burden but not with
venous thromboembolism. J Am Coll Cardiol. 2012;60:722–9.
Kamstrup PR, Tybjaerg-Hansen A, Nordestgaard BG. Genetic evidence that lipoprotein(a) associ-
ates with atherosclerotic stenosis rather than venous thrombosis. Arterioscler Thromb Vasc
Biol. 2012;32:1732–41.
Keesler GA, Gabel BR, Devlin CM, et al. The binding activity of the macrophage lipoprotein(a)/
apolipoprotein(a) receptor is induced by cholesterol via a post-translational mechanism and
recognizes distinct kringle domains on apolipoprotein(a). J Biol Chem. 1996;271:32096–104.
Kenet G, Lutkhoff LK, Albisetti M, et al. Impact of thrombophilia on risk of arterial ischemic
stroke or cerebral sinovenous thrombosis in neonates and children: a systematic review and
meta-analysis of observational studies. Circulation. 2010;121:1838–47.
10 Molecular Mechanisms of Lipoprotein(a) Pathogenicity: Tantalizing Clues… 185
Kille A, Nuhrenberg T, Franke K, et al. Association of lipoprotein(a) with intrinsic and on-
clopidogrel platelet reactivity. J Thromb Thrombolysis. 2021; https://doi.org/10.1007/
s11239-021-02515-2.
Kivimaki M, Magnussen CG, Juonala M, et al. Conventional and Mendelian randomization analy-
ses suggest no association between lipoprotein(a) and early atherosclerosis: the Young Finns
Study. Int J Epidemiol. 2011;40:470–8.
Klein JH, Hegele RA, Hackam DG, et al. Lipoprotein(a) is associated differentially with carotid
stenosis, occlusion, and total plaque area. Arterioscler Thromb Vasc Biol. 2008;28:1851–6.
Koschinsky ML, Boffa MB. Oxidized phospholipid modification of lipoprotein(a): Epidemio
logy, biochemistry and pathophysiology. Atherosclerosis. 2022;349:92–100.
Koschinsky ML, Cote GP, Gabel B, et al. Identification of the cysteine residue in apolipoprotein(a)
that mediates extracellular coupling with apolipoprotein B-100. J Biol Chem. 1993;268:
19819–25.
Lackner C, Cohen JC, Hobbs HH. Molecular definition of the extreme size polymorphism in
apolipoprotein(a). Hum Mol Genet. 1993;2:933–40.
Larsson SC, Gill D, Mason AM, et al. Lipoprotein(a) in alzheimer, atherosclerotic, cerebrovas-
cular, thrombotic, and valvular disease: mendelian randomization investigation. Circulation.
2020;141:1826–8.
Leibundgut G, Scipione C, Yin H, et al. Determinants of binding of oxidized phospholipids on
apolipoprotein (a) and lipoprotein (a). J Lipid Res. 2013;54:2815–30.
Marcovina SM, Hobbs HH, Albers JJ. Relation between number of apolipoprotein(a) kringle 4
repeats and mobility of isoforms in agarose gel: basis for a standardized isoform nomenclature.
Clin Chem. 1996;42:436–9.
Martinez C, Rivera J, Loyau S, et al. Binding of recombinant apolipoprotein(a) to human platelets
and effect on platelet aggregation. Thromb Haemost. 2001;85:686–93.
Mathieu P, Arsenault BJ, Boulanger MC, et al. Pathobiology of Lp(a) in calcific aortic valve dis-
ease. Expert Rev Cardiovasc Ther. 2017;15:797–807.
MBewu AD, Durrington PN, Mackness MI, et al. Serum Lp(a) lipoprotein concentration and out-
come of thrombolytic treatment for myocardial infarction. Br Heart J. 1994;71:316–21.
McLean JW, Tomlinson JE, Kuang WJ, et al. cDNA sequence of human apolipoprotein(a) is
homologous to plasminogen. Nature. 1987;330:132–7.
Mehta A, Vasquez N, Ayers CR, et al. Independent association of lipoprotein(a) and coronary artery
calcification with atherosclerotic cardiovascular risk. J Am Coll Cardiol. 2022;79:757–68.
Miles LA, Fless GM, Levin EG, et al. A potential basis for the thrombotic risks associated with
lipoprotein(a). Nature. 1989;339:301–3.
Nowak-Gottl U, Strater R, Heinecke A, et al. Lipoprotein (a) and genetic polymorphisms of clot-
ting factor V, prothrombin, and methylenetetrahydrofolate reductase are risk factors of sponta-
neous ischemic stroke in childhood. Blood. 1999;94:3678–82.
O’Neil CH, Boffa MB, Hancock MA, et al. Stimulation of vascular smooth muscle cell proliferation
and migration by apolipoprotein(a) is dependent on inhibition of transforming growth factor-
beta activation and on the presence of kringle IV type 9. J Biol Chem. 2004;279:55187–95.
Palabrica TM, Liu AC, Aronovitz MJ, et al. Antifibrinolytic activity of apolipoprotein(a) in vivo:
human apolipoprotein(a) transgenic mice are resistant to tissue plasminogen activator-mediated
thrombolysis. Nat Med. 1995;1:256–9.
Pellegrino M, Furmaniak-Kazmierczak E, LeBlanc JC, et al. The apolipoprotein(a) component of
lipoprotein(a) stimulates actin stress fiber formation and loss of cell-cell contact in cultured
endothelial cells. J Biol Chem. 2004;279:6526–33.
Peng J, Liu MM, Liu HH, et al. Lipoprotein (a)-mediated vascular calcification: population-based
and in vitro studies. Metabolism. 2022;127:154960.
Raitakari OT, Adams MR, Celermajer DS. Effect of Lp(a) on the early functional and structural
changes of atherosclerosis. Arterioscler Thromb Vasc Biol. 1999;19:990–5.
Rand ML, Sangrar W, Hancock MA, et al. Apolipoprotein(a) enhances platelet responses
to the thrombin receptor-activating peptide SFLLRN. Arterioscler Thromb Vasc Biol.
1998;18:1393–9.
186 M. B. Boffa and M. L. Koschinsky
Rath M, Niendorf A, Reblin T, et al. Detection and quantification of lipoprotein(a) in the arterial
wall of 107 coronary bypass patients. Arteriosclerosis. 1989;9:579–92.
Razavi AC, Cardoso R, Dzaye O, et al. Risk markers for limited coronary artery calcium in persons
with significant aortic valve calcium (from the Multi-ethnic Study of Atherosclerosis). Am J
Cardiol. 2021;156:58–64.
Ribo M, Montaner J, Molina CA, et al. Admission fibrinolytic profile predicts clot lysis resistance in
stroke patients treated with tissue plasminogen activator. Thromb Haemost. 2004;91:1146–51.
Romagnuolo R, Marcovina SM, Boffa MB, et al. Inhibition of plasminogen activation by apo(a):
role of carboxyl-terminal lysines and identification of inhibitory domains in apo(a). J Lipid
Res. 2014;55:625–34.
Romagnuolo R, DeMarco K, Scipione CA, et al. Apolipoprotein(a) inhibits the conversion of Glu-
plasminogen to Lys-plasminogen on the surface of vascular endothelial and smooth muscle
cells. Thromb Res. 2018a;169:1–7.
Romagnuolo R, Scipione CA, Bazzi ZA, et al. Inhibition of pericellular plasminogen activation
by apolipoprotein(a): roles of urokinase plasminogen activator receptor and integrins alphaM-
beta2 and alphaVbeta3. Atherosclerosis. 2018b;275:11–21.
Rouy D, Koschinsky ML, Fleury V, et al. Apolipoprotein(a) and plasminogen interactions with
fibrin: a study with recombinant apolipoprotein(a) and isolated plasminogen fragments.
Biochemistry. 1992;31:6333–9.
Rowland CM, Pullinger CR, Luke MM, et al. Lipoprotein (a), LPA Ile4399Met, and fibrin clot
properties. Thromb Res. 2014;133:863–7.
Salsoso R, Dalcoquio TF, Furtado RHM, et al. Relation of high lipoprotein (a) concentrations
to platelet reactivity in individuals with and without coronary artery disease. Adv Ther.
2020;37:4568–84.
Sangrar W, Bajzar L, Nesheim ME, et al. Antifibrinolytic effect of recombinant apolipoprotein(a)
in vitro is primarily due to attenuation of tPA-mediated Glu-plasminogen activation.
Biochemistry. 1995;34:5151–7.
Sangrar W, Gabel BR, Boffa MB, et al. The solution phase interaction between apolipoprotein(a)
and plasminogen inhibits the binding of plasminogen to a plasmin-modified fibrinogen surface.
Biochemistry. 1997;36:10353–63.
Schnitzler JG, Hoogeveen RM, Ali L, et al. Atherogenic lipoprotein(a) increases vascular glycoly-
sis, thereby facilitating inflammation and leukocyte extravasation. Circ Res. 2020;126:1346–59.
Scipione CA, Sayegh SE, Romagnuolo R, et al. Mechanistic insights into Lp(a)-induced IL-8 expres-
sion: a role for oxidized phospholipid modification of apo(a). J Lipid Res. 2015;56:2273–85.
Scipione CA, McAiney JT, Simard DJ, et al. Characterization of the I4399M variant of
apolipoprotein(a): implications for altered prothrombotic properties of lipoprotein(a). J
Thromb Haemost. 2017;15:1834–44.
Seimon TA, Nadolski MJ, Liao X, et al. Atherogenic lipids and lipoproteins trigger CD36-TLR2-
dependent apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell Metab.
2010;12:467–82.
Skuza AA, Polak M, Undas A. Elevated lipoprotein(a) as a new risk factor of cerebral venous sinus
thrombosis: association with fibrin clot properties. J Thromb Thrombolysis. 2019;47:8–15.
Sotiriou SN, Orlova VV, Al-Fakhri N, et al. Lipoprotein(a) in atherosclerotic plaques recruits
inflammatory cells through interaction with Mac-1 integrin. FASEB J. 2006;20:559–61.
Strater R, Becker S, von Eckardstein A, et al. Prospective assessment of risk factors for recurrent
stroke during childhood—a 5-year follow-up study. Lancet. 2002;360:1540–5.
Takami S, Yamashita S, Kihara S, et al. Lipoprotein(a) enhances the expression of intercel-
lular adhesion molecule-1 in cultured human umbilical vein endothelial cells. Circulation.
1998;97:721–8.
Thanassoulis, G, Campbell, CY, Owens, DS, et al., CHARGE Extracoronary Calcium Working
Group. Genetic associations with valvular calcification and aortic stenosis. N Engl J Med.
2013;368:503–12.
10 Molecular Mechanisms of Lipoprotein(a) Pathogenicity: Tantalizing Clues… 187
Tranchesi B Jr, Chamone DF, Cobbaert C, et al. Coronary recanalization rate after intravenous
bolus of alteplase in acute myocardial infarction. Am J Cardiol. 1991;68:161–5.
van der Hoek YY, Wittekoek ME, Beisiegel U, et al. The apolipoprotein(a) kringle IV repeats
which differ from the major repeat kringle are present in variably-sized isoforms. Hum Mol
Genet. 1993;2:361–6.
van der Valk FM, Bekkering S, Kroon J, et al. Oxidized phospholipids on lipoprotein(a) elicit
arterial wall inflammation and an inflammatory monocyte response in humans. Circulation.
2016;134:611–24.
van Dijk RA, Kolodgie F, Ravandi A, et al. Differential expression of oxidation-specific epitopes
and apolipoprotein(a) in progressing and ruptured human coronary and carotid atherosclerotic
lesions. J Lipid Res. 2012;53:2773–90.
von Hodenberg E, Kreuzer J, Hautmann M, et al. Effects of lipoprotein (a) on success rate of
thrombolytic therapy in acute myocardial infarction. Am J Cardiol. 1991;67:1349–53.
Yeang C, Cotter B, Tsimikas S. Experimental animal models evaluating the causal role of
lipoprotein(a) in atherosclerosis and aortic stenosis. Cardiovasc Drugs Ther. 2016;30:75–85.
Youssef A, Clark JR, Marcovina SM, et al. Apo(a) and ApoB interact noncovalently within hepato-
cytes: implications for regulation of Lp(a) levels by modulation of ApoB secretion. Arterioscler
Thromb Vasc Biol. 2022;42:289–304.
Zheng KH, Tsimikas S, Pawade T, et al. Lipoprotein(a) and oxidized phospholipids promote valve
calcification in patients with aortic stenosis. J Am Coll Cardiol. 2019;73:2150–62.
Chapter 11
Thrombosis, Inflammation,
and Lipoprotein(a): Clinical Implications
M. S. Safarova
Atherosclerosis and Lipid Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
Department of Cardiovascular Medicine, University of Kansas Hospital and Medical Center,
Kansas City, KS, USA
e-mail: [email protected]
P. M. Moriarty (*)
Division of Endocrinology, Diabetes and Clinical Pharmacology, Department of Internal
Medicine, University of Kansas Medical Center, Kansas City, KS, USA
Atherosclerosis and Lipoprotein Apheresis Center, University of Kansas Medical Center,
Kansas City, KS, USA
e-mail: [email protected]
Fig. 11.1 Lp(a) mediates components of the endogenous fibrinolytic system and oxidation
LPA gene, IL-6 can induce apo(a) expression. As shown, transcription factor ELK1
mediates expression of apo(a) through the Ets domain in the LPA promoter; fibro-
blast growth factor 19 (FGF19) has inhibitory effects on apo(a) expression. The
oxidized phospholipids (oxPL)-Lp(a) complex can upregulate adhesion molecules,
increase secretion of chemo-attractants and cytokines, interact with various signal
transduction receptors on the cell surface, and modulate binding of leukocytes to
endothelial cells. Cells can recognize oxPL through scavenger receptors (SR) CD36
and SR class B type 1 (SR-B1), prostaglandin E2 receptor 2 subtype (EP2), vascular
endothelial growth factor receptor 2 (VEGFR2), and the platelet-activating factor
(PAF) receptor. Smaller Lp(a) isoforms have stronger association with
oxPL. Oxidation of Lp(a) increases clot density. Role of Lp(a)-targeted therapies in
mediating levels of pro-inflammatory, pro-thrombotic, and antifibrinolytic markers
and its effects on clinical outcomes needs further investigation. In this chapter, we
discuss clinical relevance and implications of pro-thrombotic and pro-inflammatory
states associated with Lp(a).
Clinical Vignette
A previously healthy 11-year-old boy was admitted for a 5-day history of head-
aches, dizziness, and lethargy. His exam showed visual field defects and memory
impairment. Magnetic resonance imaging of the brain demonstrated acute strokes in
the right parietal, left cerebellar, left thalamic, and bilateral frontal lobes. An
11 Thrombosis, Inflammation, and Lipoprotein(a): Clinical Implications 191
Introduction
The point of life is to find the delicate equilibrium between dream and reality.—Lillian
Eugenia Smith
A high plasma level of Lp(a) is a causal risk factor for atherosclerotic cardiovas-
cular disease (ASCVD) through various pathways associated with increased athero-
genesis, inflammation, and thrombosis (Reyes-Soffer et al. 2022; Tsimikas 2017).
Lp(a) affects endothelial function and mediates inflammation and oxidative stress,
fibrinolysis, and plaque stability, leading to accelerated atherothrombosis. Such life-
threatening and debilitating events as described in the above clinical vignettes moti-
vate to continue building the foundation of understanding clinical implications of
192 M. S. Safarova and P. M. Moriarty
Lp(a) in: (1) (athero)thrombosis, (2) platelet and coagulation cascade, and (3)
inflammation. According to the 2018 National Heart Lung and Blood Institute
(NHLBI) report, an estimated 1.4 billion people globally have Lp(a) levels ≥50 mg/
mL (>100–125 nmol/L) with a prevalence ranging from 10% to 30% (Tsimikas
et al. 2018). The prevalence is higher in patients with established ASCVD, calcific
aortic valve disease, and chronic kidney disease (Tsimikas et al. 2018).
Lp(a) is a lipoprotein with a density between 1.06 and 1.11 g/mL that can bind to
lysine-rich components of the coagulation system and several components of extra-
cellular and subendothelial matrix of the vascular wall via its apoB and
apolipoprotein(a) (apo(a)), including binding to fibronectin, fibrinogen, glycosami-
noglycans, and proteoglycans (Kostner and Bihari-Varga 1990; Klezovitch et al.
1998). Lp(a) can be retained in the arterial intima, localizing preferentially to ath-
erosclerotic plaques (Boffa and Koschinsky 2016). The low-density lipoprotein
(LDL) moiety of the Lp(a) particle is covalently linked to the plasminogen-like
glycoprotein apo(a) through a single disulfide link between apolipoprotein B100
Cys 3734 and apo(a) kringle IV type 9 Cys67. Lp(a) is almost completely confined
to a subset of primates. It has been proposed that the duplication of the PLG gene
evolved into the LPA gene (Lawn et al. 1995).
Similarities between the two genes (LPA and PLG) include 5′-flanking and
untranslated regions, multiple copies of kringle IV-, a single copy kringle V-like
and protease-like domains, as well as a related 3′-untranslated region (Lawn et al.
1995; McLean et al. 1987). An Lp(a)-like complex is found in hedgehogs which is
thought to have evolved independently from that of humans. While individuals
with low concentrations of plasma Lp(a) typically show no syndromic features or
pathologic conditions, the physiological role of Lp(a) in humans is not entirely
clear. An analysis of individual-level data of 112,338 UK Biobank participants
demonstrated that one standard deviation of genetically lowered Lp(a) level was
associated with reduction in risk of coronary heart disease (CHD) and peripheral
vascular disease by 30% (odds ratio, 0.71; 95% confidence interval, 0.69–0.73 and
0.69; 0.59–0.80, respectively), ischemic stroke by 13% (0.87; 0.79–0.96), aortic
valve stenosis by 37% (0.63; 0.47–0.83) (Emdin et al. 2016). These findings were
reproduced with the LPA-rs41272114 (null allele frequency, 3%) associated with
low Lp(a), providing a significant protective effect in carriers with a ~20% risk
reduction in CHD (Kyriakou et al. 2014). No association of genetically predicted
low Lp(a) levels with type 2 diabetes, malignancy, or venous thromboembolism
(VTE) was observed in this study (Emdin et al. 2016). Current hypothesis of the
evolutionary advantages of synthesizing Lp(a)-like particles includes accelerated
repair of vascular lesions, tissue injuries, healing of wounds, reduced bleeding, as
well as induction and participation in different forms of acute phase responses
(Lippi and Guidi 2000; Brown and Goldstein 1987; Kronenberg and Utermann
2013; Caplice et al. 2001; Boffa et al. 2004; von Zychlinski et al. 2011). However,
these properties can become pathogenic in the setting of increased concentrations
of the lipoprotein and homeostatic imbalance.
11 Thrombosis, Inflammation, and Lipoprotein(a): Clinical Implications 193
It is hard to imagine that nature is only teasing us and the structural resemblance between
apo(a) and plasminogen has no clinical consequences.—Michael S. Brown and Joseph
L. Goldstein (1987)
Lp(a) has been shown to inhibit endogenous fibrinolysis through: (1) competitive
inhibition of pericellular plasminogen activation on vascular and blood cells medi-
ated by tissue plasminogen activator (tPA), (2) mediation of plasminogen binding to
platelets, (3) competition with plasminogen for binding to fibrin, mononuclear cells,
annexin II on endothelial cells, (4) inhibition of fibrinogen binding to platelets acti-
vated with platelet-activating factor (PAF) (Loscalzo et al. 1990; Edelberg et al.
1989; Hajjar et al. 1989; Simon et al. 1991; Ezratty et al. 1993; Edelberg and Pizzo
1995; Moliterno et al. 1993; Romagnuolo et al. 2014). In mice resistant to tPA-
mediated thrombolysis, apo(a) was reported to reduce clot lysis in vivo (Palabrica
et al. 1995). Clot lysis was attenuated in the setting of apo(a) transgene when com-
pared to their sex-matched normal littermates. Experiments with removal of kringle
V and the lysine binding site in kringle IV type 10, respectively, negated and sub-
stantially reduced the inhibitory effect of apo(a) (Romagnuolo et al. 2014). In cul-
tured endothelial cells, Lp(a) was shown to induce rearrangements of actin filaments
(Pellegrino et al. 2004) and upregulate the expression of PAI-1, thereby reducing the
amount of tPA available for plasminogen activation (Levin et al. 1994). Lp(a)-
mediated plasmin recognition of fibrin clots was demonstrated in experiments
196 M. S. Safarova and P. M. Moriarty
showing that apo(a) upregulated α2-antiplasmin (Edelberg and Pizzo 1992), thereby
impairing fibrinolysis.
In the presence of Lp(a), washed human platelets demonstrate significant
enhancement of aggregation and release of granule contents (Rand et al. 1998). In
direct binding experiments, specific and reversible binding of Lp(a) to platelets was
observed (Ezratty et al. 1993). On the other hand, activation of platelets with ade-
nosine diphosphate (ADP) halted Lp(a) binding capacity. Further, in a dose-
dependent manner, Lp(a) was shown to inhibit PAF-induced platelet activation as
well as primary and secondary platelet aggregation induced by ADP and calcium.
When apo(a) was completely removed from the Lp(a) particle, its inhibitory effect
on PAC-1 (a mouse monoclonal antibody indicative of platelet activation) binding
to activated platelets significantly enhanced the antiaggregatory effects in compari-
son with the “unreduced” Lp(a) (Tsironis et al. 2004).
There is controversy surrounding apo(a) isoform size dependent versus indepen-
dent inhibition of plasminogen activation by Lp(a). In the clinical setting, it has
been demonstrated that the size of apo(a) isoforms was inversely associated with an
up to eightfold increase in the risk of thromboembolic events (Espinosa et al. 2001;
Falco et al. 1998). In a case-control study, copy number variation of LPA KIV-2 was
an independent determinant of VTE (Sticchi et al. 2016). The KIV type 2 repeat
number was significantly lower in patients with VTE than in healthy controls,
including an observed higher frequency of the KIV-2 repeat number of less than 8
(Sticchi et al. 2016). Levels of Lp(a) may vary up to 200-fold for a given apo(a)
isoform (Perombelon et al. 1994). Single nucleotide polymorphisms mapped to LPA
and KIV-2 copy number have been shown to provide complementary information,
explaining the variation in plasma Lp(a) concentrations (Lanktree et al. 2010).
Thus, while some research has shown that small size apo(a) isoforms display higher
affinity to bind fibrin (Angles-Cano et al. 2001), there are data demonstrating a lack
of association between the LPA score comprising rs10455872 and rs3798220 vari-
ants (Helgadottir et al. 2012; Danik et al. 2013), number of KIV type 2 repeats, and
the level of plasminogen inhibition (Romagnuolo et al. 2014; Hancock et al. 2003),
highlighting a need for further research in this area.
Lp(a) is the preferential lipoprotein carrier for oxidized phospholipids (OxPL), pro-
atherogenic and pro-inflammatory markers. In vitro experiments demonstrated that
products of oxidation make fibrin clot less permeable (Hoffman 2008), another
mechanism by which Lp(a) may be affecting clot lysis. In individuals with elevated
Lp(a) levels (>50 mg/dL), a local increase in the arterial wall inflammation in vivo,
enhanced peripheral blood mononuclear cells trafficking as well as transendothelial
migration and accumulation in the arterial wall with and without plaquing was
noted (van der Valk et al. 2016). Further, it was shown that patients with familial
hypercholesterolemia (average LDL-C, 236 mg/dL) and elevated Lp(a) levels
11 Thrombosis, Inflammation, and Lipoprotein(a): Clinical Implications 197
(range, 43–401 nmol/L) elicit markedly increased local arterial wall inflammation
as compared to healthy control subjects (van Wijk et al. 2014). Following lipopro-
tein apheresis, a significant reduction of the arterial wall inflammation estimated
using the target-to-background ratio of a fluorodeoxyglucose (FDG) uptake on the
PET/CT scan was demonstrated (van Wijk et al. 2014). This anti-inflammatory
effect was observed after a single session of apheresis. Milder reduction in the Lp(a)
levels with PCSK9 inhibitors did not show any significant change in target-to-
background ratio of the index arterial vessel as compared to placebo (Stiekema
et al. 2019).
Mechanism by which cells recognize oxPL include recognition and interactions
with the cell receptors such as CD36, SRB1, EP2, VEGFR2, Toll-like receptor-4,
and the PAF receptors (Zimman et al. 2007; Berliner et al. 2009). In the middle-
aged patients with type 2 diabetes, oxidative stress enhanced pro-thrombotic state
by unfavorably affecting the fibrin network structure and impairing fibrin clot sus-
ceptibility to lysis regardless of diabetes severity and duration (Lados-Krupa et al.
2015). Oxidized apoB100-containing lipoproteins were independently associated
with the clot lysis time in diabetes, suggesting that oxidized forms might directly
impair plasmin-mediated fibrin degradation (Lados-Krupa et al. 2015). An aggre-
gation of Lp(a) particles on the surface of the aortic smooth muscle cells was
observed in the presence lipoprotein lipase and sphingomyelinase (Tabas et al.
1993). In the same series of experiments, coincubation of Lp(a)-coated smooth
muscle cells with macrophages resulted in formation of lipid-laden macrophages
(Tabas et al. 1993). The role of macrophages in Lp(a) pathophysiology is discussed
in Chap. 10.
Several mechanisms of Lp(a)-mediated plaque instability have been described,
including increased production of interleukin-8 (IL-8). IL-8 among other effects
disinhibits matrix metalloproteinases (Moreau et al. 1999; Ezhov et al. 2019),
which increases the intensity of plaque inflammation and likelihood of plaque rup-
ture. Oxidized Lp(a) fractions induce a dose-dependent reduction of nitric oxide
synthase expression and, as a result, reduce nitric oxide production (Moeslinger
et al. 2006), thereby adversely affecting vascular homeostasis. In an assessment of
vasomotor response to acetylcholine, an association between elevated Lp(a) levels
and impaired endothelium-dependent vasodilatation in coronary arteries with and
without angiographically detectable atherosclerotic lesions was observed (Tsurumi
et al. 1995). In a dose-dependent manner, Lp(a) promotes intercellular cell adhe-
sion molecule-1 (ICAM1) expression (Takami et al. 1998), upregulation of which
has a key role in the inflammatory response (Libby et al. 2011). Proteomic analysis
of Lp(a) revealed its association with the histidine-rich glycoprotein (HRG) known
to interact with heparin, plasminogen, fibrinogen, and complement components
(von Zychlinski et al. 2011). Further there was a strong signal for the C3 comple-
ment component in the Lp(a) position in the plasma protein fraction, highlighting
an association of C3 with the Lp(a) particle (von Zychlinski et al. 2011; Garcia-
Arguinzonis et al. 2021).
In critically ill patients with COVID-19 infection, treatment with monoclonal
antibodies against IL-6 receptor (such as tocilizumab and sarilumab) was shown to
198 M. S. Safarova and P. M. Moriarty
Medicine is the art of addition and subtraction. The subtraction of all that is excessive, and
the addition of all that is missing. And he who might be the best at doing this — will be the
best doctor.—Hippocrates Asclepiades.
11 Thrombosis, Inflammation, and Lipoprotein(a): Clinical Implications 199
Among carriers of the LPA-rs3798220 variant (median Lp(a) level >80 mg/dL) in
the Women’s Health Study, aspirin intake was associated with a 56% (hazard ratio,
0.44; 0.20–0.94) reduction in the risk of major cardiovascular events, including
myocardial infarction, ischemic stroke, and cardiovascular death (Chasman et al.
2009). After a median of 9.9 years, there was a twofold decrease in the absolute risk
in the placebo group: the event rate was 2.14% (0.81–3.45%) in the aspirin group
and 4.83% (2.74–6.87%) with placebo (Chasman et al. 2009). In Japanese patients,
treatment with aspirin was associated with a significant reduction in Lp(a) levels
(Akaike et al. 2002). These findings were reproduced in the South Asian population
aged 21–60 years with aspirin significantly reducing Lp(a) levels (Ranga et al.
2007). Supporting experimental data in mice suggested that aspirin can suppress
apo(a) mRNA expression (Kagawa et al. 1999). In healthy individuals, lower Lp(a)
values were measured in EDTA-treated plasma, citrated, and heparinized plasma as
compared to the serum (Lippi et al. 1996). However, there are no studies investigat-
ing effects of anticoagulation on Lp(a).
Apo-B Lowering
In patients with CHD with on-statin LDL above 130 mg/dL, elevated Lp(a) levels
>80th percentile additionally increased the risk of recurrent ischemic events by 40%
(odds ratio, 1.40; 1.15–1.71) (O’Donoghue et al. 2014). In a meta-analysis includ-
ing primary and secondary ASCVD prevention randomized clinical trials similar
odds ratios (1.43; 1.15–1.76) were observed in statin-treated patients with Lp(a)
levels >50 mg/dL (Willeit et al. 2018). In the primary prevention JUPITER trial
(Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating
Rosuvastatin) independent of the LDL-C levels, for each standard deviation change
in Lp(a) while on a statin there was a 27% (hazard ratio, 1.27; 1.01–1.59) increase
in the relative risk of incident ASCVD (Khera et al. 2014). Among 25,096 partici-
pants in the secondary prevention FOURIER trial (Further Cardiovascular Outcomes
Research with PCSK9 Inhibition in Subjects with Elevated Risk), patients with
higher baseline Lp(a) (>120 nmol/L) had a 25% relative risk reduction (0.64–0.88)
and a 2.4% absolute risk reduction with PCSK9 inhibition, translating into an esti-
mated number needed to treat of 41 to prevent one fatal or non-fatal myocardial
infarction or urgent revascularization (O’Donoghue et al. 2019). There is ample
evidence that selective lipoprotein apheresis improves clinical outcomes related to
atherothrombosis in patients with elevated Lp(a) (Jaeger et al. 2009; Leebmann
et al. 2013; Safarova et al. 2013). Lipoprotein apheresis results in greater than 60%
reduction of apoB-containing lipoproteins following a single apheresis procedure. It
has been shown to improve blood rheology, rapidly and efficiently reduce Lp(a)
levels and inflammatory markers, including IL-6 and oxPL (Moriarty 2015). In
patients with CHD, familial hypercholesterolemia, and elevated Lp(a) levels, treat-
ment with lipoprotein apheresis demonstrated a significant downregulation of
200 M. S. Safarova and P. M. Moriarty
mRNA expression for IL-1α, IL-6, and TNF-α (Stefanutti et al. 2017). Since 2020,
the US Food and Drug Administration (FDA) approved criteria for apheresis in
secondary prevention of ASCVD with LDL-C levels >100 mg/dL and Lp(a) >60 mg/
dL on maximally tolerated lipid-lowering therapy (Nugent et al. 2020).
The importance of assessing Lp(a) in patients in the primary prevention setting
as well as in those with prior thrombotic events relates to its ability to identify sub-
jects at increased risk, and the potential to modulate impaired fibrinolysis and
inflammation and as a result, improve outcomes. To date, extensive evidence exists
in support of a causal association of elevated Lp(a) levels with the risk of atheroscle-
rosis development and progression in different vascular beds. Likely, the presence
of the atherosclerotic plaque is in fact an inciting event for Lp(a) to promote throm-
bus formation in the unstable milieu. Current data suggest that except for Lp(a)
levels ≥90–95th percentile, small isoforms (KIV-2 repeats <10–6th percentile) and
in individuals with existing pro-inflammatory and pro-thrombotic conditions, Lp(a)
does not initiate venous thrombus formation, but rather contributes to its propaga-
tion and density. Lp(a) has a complex genetic architecture. For instance, studies
within or outside the LPA gene region demonstrated that in carriers of the Lp(a)-
raising genetic variants (i.e., rs10455872), the presence of a rare missense
rs41267813[A] variant was associated with substantially lower Lp(a) concentra-
tions (Said et al. 2021), potentially offering a protective effect against CHD.
Discovery of new mechanisms elucidating pathways affecting interplay between
Lp(a) and acute thrombotic events will continue to provide potential therapeutic tar-
gets addressing current gaps in residual risk. Genetic studies evaluating LPA interac-
tion using functional studies and its clinical relevance on the clinical outcomes
involving diverse cohorts are needed. Further research is warranted to assess differ-
ences in the markers of the coagulation system activation and platelet activation before
and after Lp(a)-targeted treatment to address pathophysiological relevance of pro-
thrombotic and atherogenic properties of Lp(a) in humans. Studies investigating
effects of aspirin in primary prevention and other antiplatelet agents in secondary
prevention in patients with elevated Lp(a) are needed. Polygenic risk scores can assist
in unraveling the interplay between thrombosis and inflammation in patients with
elevated Lp(a), especially when tested in racially diverse populations (Table 11.1).
Table 11.1 Proposed research questions to bridge gaps in knowledge of clinical relevance of
elevated Lp(a) in atherothrombosis and venous thrombosis
Design Proposed study question
Novel experimental Mechanisms and pathways beyond plasminogen activation and tPA-
models mediated fibrinolysis affecting interaction between high Lp(a), acute
Functional studies thrombosis, and unstable atherosclerotic lesion development.
Biomarker research Markers of the coagulation system activation, fibrinolytic potential, and
platelet activation before and after Lp(a)-targeted treatment, with and
without other conditions precipitating thrombosis.
Assessment of anti-inflammatory effects of Lp(a)-lowering therapies on
vulnerable plaques identified using novel imaging modalities
Genetic studies Clinical impact of genetic architecture of elevated Lp(a) on outcomes in
cohorts with diverse ethnic/racial background.
Clinical/pragmatic Effects of aspirin in primary prevention of ASCVD in individuals with
trials elevated Lp(a).
Effects of dual antiplatelet agents/more potent antiplatelet agents in
secondary prevention of ASCVD in patients with elevated Lp(a).
Role of elevated Lp(a) in determining duration of anticoagulation in
patients with VTE.
Role of elevated Lp(a) in addressing the risk of stroke in patients with
atrial fibrillation.
Assessing the role of Lp(a) and targeted treatment in patients undergoing
aortic valve replacement.
Clinical trials/ Lp(a)-targeted therapies and prognosis in patients with elevated Lp(a),
outcome research identifying race-specific therapeutic targets
References
Chasman DI, Shiffman D, Zee RY, et al. Polymorphism in the apolipoprotein(a) gene, plasma
lipoprotein(a), cardiovascular disease, and low-dose aspirin therapy. Atherosclerosis.
2009;203:371–6.
Danik JS, Buring JE, Chasman DI, Zee RY, Ridker PM, Glynn RJ. Lipoprotein(a), polymorphisms
in the LPA gene, and incident venous thromboembolism among 21483 women. J Thromb
Haemost. 2013;11:205–8.
Dentali F, Gessi V, Marcucci R, Gianni M, Grandi AM, Franchini M. Lipoprotein(a) as a risk factor
for venous thromboembolism: a systematic review and meta-analysis of the literature. Semin
Thromb Hemost. 2017;43:614–20.
Di Maio S, Lamina C, Coassin S, et al. Lipoprotein(a) and SARS-CoV-2 infections: Susceptibility
to infections, ischemic heart disease and thromboembolic events. J Intern Med. 2021; https://
doi.org/10.1111/joim.13338.
Edelberg JM, Pizzo SV. Lipoprotein (a) promotes plasmin inhibition by alpha 2-antiplasmin.
Biochem J. 1992;286(Pt 1):79–84.
Edelberg JM, Pizzo SV. Lipoprotein (a) in the regulation of fibrinolysis. J Atheroscler Thromb.
1995;2(Suppl 1):S5–7.
Edelberg JM, Gonzalez-Gronow M, Pizzo SV. Lipoprotein a inhibits streptokinase-mediated acti-
vation of human plasminogen. Biochemistry. 1989;28:2370–4.
Emdin CA, Khera AV, Natarajan P, et al. Phenotypic characterization of genetically lowered human
lipoprotein(a) levels. J Am Coll Cardiol. 2016;68:2761–72.
Espinosa G, Tassies D, Font J, et al. Antiphospholipid antibodies and thrombophilic factors in
giant cell arteritis. Semin Arthritis Rheum. 2001;31:12–20.
Etingin OR, Hajjar DP, Hajjar KA, Harpel PC, Nachman RL. Lipoprotein (a) regulates plasmino-
gen activator inhibitor-1 expression in endothelial cells. A potential mechanism in thrombogen-
esis. J Biol Chem. 1991;266:2459–65.
Ezhov M, Safarova M, Afanasieva O, Mitroshkin M, Matchin Y, Pokrovsky S. Matrix metallopro-
teinase 9 as a predictor of coronary atherosclerotic plaque instability in stable coronary heart
disease patients with elevated lipoprotein(a) levels. Biomolecules. 2019;9(4):129. https://doi.
org/10.3390/biom9040129.
Ezratty A, Simon DI, Loscalzo J. Lipoprotein(a) binds to human platelets and attenuates plasmino-
gen binding and activation. Biochemistry. 1993;32:4628–33.
Falco C, Estelles A, Dalmau J, Espana F, Aznar J. Influence of lipoprotein (a) levels and isoforms
on fibrinolytic activity—study in families with high lipoprotein (a) levels. Thromb Haemost.
1998;79:818–23.
Garcia-Arguinzonis M, Diaz-Riera E, Pena E, et al. Alternative C3 complement system: lipids and
atherosclerosis. Int J Mol Sci. 2021;22:5122.
Goldenberg NA, Bernard TJ, Hillhouse J, et al. Elevated lipoprotein (a), small apolipoprotein
(a), and the risk of arterial ischemic stroke in North American children. Haematologica.
2013;98:802–7.
Grainger DJ, Kirschenlohr HL, Metcalfe JC, Weissberg PL, Wade DP, Lawn RM. Proliferation of
human smooth muscle cells promoted by lipoprotein(a). Science. 1993;260:1655–8.
Gregson J, Kaptoge S, Bolton T, et al. Cardiovascular risk factors associated with venous throm-
boembolism. JAMA Cardiol. 2019;4:163–73.
Hajjar KA, Gavish D, Breslow JL, Nachman RL. Lipoprotein(a) modulation of endothelial cell
surface fibrinolysis and its potential role in atherosclerosis. Nature. 1989;339:303–5.
Hancock MA, Boffa MB, Marcovina SM, Nesheim ME, Koschinsky ML. Inhibition of plasmino-
gen activation by lipoprotein(a): critical domains in apolipoprotein(a) and mechanism of inhi-
bition on fibrin and degraded fibrin surfaces. J Biol Chem. 2003;278:23260–9.
Heit JA, Armasu SM, Asmann YW, et al. A genome-wide association study of venous throm-
boembolism identifies risk variants in chromosomes 1q24.2 and 9q. J Thromb Haemost.
2012;10:1521–31.
Helgadottir A, Gretarsdottir S, Thorleifsson G, et al. Apolipoprotein(a) genetic sequence vari-
ants associated with systemic atherosclerosis and coronary atherosclerotic burden but not with
venous thromboembolism. J Am Coll Cardiol. 2012;60:722–9.
11 Thrombosis, Inflammation, and Lipoprotein(a): Clinical Implications 203
Libby P, Ridker PM, Hansson GK. Progress and challenges in translating the biology of athero-
sclerosis. Nature. 2011;473:317–25.
Lippi G, Guidi G. Lipoprotein(a): from ancestral benefit to modern pathogen? QJM. 2000;93:75–84.
Lippi G, Giampaolo L, Guidi G. Effects of anticoagulants on lipoprotein(a) measurements with
four commercial assays. Eur J Clin Chem Clin Biochem. 1996;34:251–5.
Loscalzo J, Weinfeld M, Fless GM, Scanu AM. Lipoprotein(a), fibrin binding, and plasminogen
activation. Arteriosclerosis. 1990;10:240–5.
Ma GS, Wilkinson MJ, Reeves RR, et al. Lipoprotein(a) in patients undergoing transcatheter aortic
valve replacement. Angiology. 2019;70:332–6.
McLean JW, Tomlinson JE, Kuang WJ, et al. cDNA sequence of human apolipoprotein(a) is
homologous to plasminogen. Nature. 1987;330:132–7.
Miller PE, Martin SS, Toth PP, et al. Screening and advanced lipid phenotyping in familial hyper-
cholesterolemia: The Very Large Database of Lipids Study-17 (VLDL-17). J Clin Lipidol.
2015;9:676–83.
Moeslinger T, Friedl R, Spieckermann PG. Inhibition of inducible nitric oxide synthesis by aza-
thioprine in a macrophage cell line. Life Sci. 2006;79:374–81.
Moliterno DJ, Lange RA, Meidell RS, et al. Relation of plasma lipoprotein(a) to infarct artery
patency in survivors of myocardial infarction. Circulation. 1993;88:935–40.
Moreau M, Brocheriou I, Petit L, Ninio E, Chapman MJ, Rouis M. Interleukin-8 mediates downreg-
ulation of tissue inhibitor of metalloproteinase-1 expression in cholesterol-loaded human mac-
rophages: relevance to stability of atherosclerotic plaque. Circulation. 1999;99:420–6. PMID:
30934954. Matrix Metalloproteinase 9 as a Predictor of Coronary Atherosclerotic Plaque
Instability in Stable Coronary Heart Disease Patients with Elevated Lipoprotein(a) Levels.
Moriarty PM. Lipoprotein apheresis: present and future uses. Curr Opin Lipidol. 2015;26:544–52.
Moriarty PM, Tennant H, Sehar N, et al. Case report of male child with elevated lipoprotein (a)
leading to acute ischemic stroke. J Clin Apher. 2017;32:574–8.
Nguyen S, Ilano L, Oluoha N, Pakbaz Z. Lipoprotein(a) a risk factor for venous thrombosis and
pulmonary embolism in patients younger than 50 years of age. Blood. 2018;132:5055.
Nordestgaard BG, Chapman MJ, Ray K, et al. Lipoprotein(a) as a cardiovascular risk factor: cur-
rent status. Eur Heart J. 2010;31:2844–53.
Nowak-Gottl U, Debus O, Findeisen M, et al. Lipoprotein (a): its role in childhood thromboembo-
lism. Pediatrics. 1997;99:E11.
Nugent AK, Gray JV, Gorby LK, Moriarty PM. Lipoprotein apheresis: first FDA indicated treat-
ment for elevated lipoprotein(a). J Clin Cardiol. 2020;1:16–21.
O’Donoghue ML, Morrow DA, Tsimikas S, et al. Lipoprotein(a) for risk assessment in patients
with established coronary artery disease. J Am Coll Cardiol. 2014;63:520–7.
O’Donoghue ML, Fazio S, Giugliano RP, et al. Lipoprotein(a), PCSK9 inhibition, and cardiovas-
cular risk. Circulation. 2019;139:1483–92.
Paciullo F, Giannandrea D, Virgili G, Cagini C, Gresele P. Role of increased lipoprotein (a) in
retinal vein occlusion: a systematic review and meta-analysis. TH Open. 2021;5:e295–302.
Palabrica TM, Liu AC, Aronovitz MJ, Furie B, Lawn RM, Furie BC. Antifibrinolytic activity of
apolipoprotein(a) in vivo: human apolipoprotein(a) transgenic mice are resistant to tissue plas-
minogen activator-mediated thrombolysis. Nat Med. 1995;1:256–9.
Pellegrino M, Furmaniak-Kazmierczak E, LeBlanc JC, et al. The apolipoprotein(a) component of
lipoprotein(a) stimulates actin stress fiber formation and loss of cell-cell contact in cultured
endothelial cells. J Biol Chem. 2004;279:6526–33.
Perombelon YF, Soutar AK, Knight BL. Variation in lipoprotein(a) concentration associated with
different apolipoprotein(a) alleles. J Clin Invest. 1994;93:1481–92.
Pierini FS, Botta E, Soriano ER, et al. Effect of tocilizumab on LDL and HDL characteristics in
patients with rheumatoid arthritis. An observational study. Rheumatol Ther. 2021;8:803–15.
Rand ML, Sangrar W, Hancock MA, et al. Apolipoprotein(a) enhances platelet responses
to the thrombin receptor-activating peptide SFLLRN. Arterioscler Thromb Vasc Biol.
1998;18:1393–9.
11 Thrombosis, Inflammation, and Lipoprotein(a): Clinical Implications 205
Ranga GS, Kalra OP, Tandon H, Gambhir JK, Mehrotra G. Effect of aspirin on lipoprotein(a) in
patients with ischemic stroke. J Stroke Cerebrovasc Dis. 2007;16:220–4.
Reyes-Soffer G, Ginsberg HN, Berglund L, et al. Lipoprotein(a): a genetically determined, causal,
and prevalent risk factor for atherosclerotic cardiovascular disease: a scientific statement from
the American Heart Association. Arterioscler Thromb Vasc Biol. 2022;42(1):e48–e60.
Rodger MA, Le Gal G, Carrier M, et al. Serum lipoprotein (a) levels in patients with first
unprovoked venous thromboembolism is not associated with subsequent risk of recurrent
VTE. Thromb Res. 2010;126:222–6.
Romagnuolo R, Marcovina SM, Boffa MB, Koschinsky ML. Inhibition of plasminogen activation
by apo(a): role of carboxyl-terminal lysines and identification of inhibitory domains in apo(a).
J Lipid Res. 2014;55:625–34.
Safarova MS, Ezhov MV, Afanasieva OI, et al. Effect of specific lipoprotein(a) apheresis on coro-
nary atherosclerosis regression assessed by quantitative coronary angiography. Atheroscler
Suppl. 2013;14:93–9.
Said MA, Yeung MW, van de Vegte YJ, et al. Genome-wide association study and identification of
a protective missense variant on lipoprotein(a) concentration: protective missense variant on
lipoprotein(a) concentration-brief report. Arterioscler Thromb Vasc Biol. 2021;41:1792–800.
Satterfield BA, Dikilitas O, Safarova MS, et al. Associations of genetically predicted lipoprotein
(a) levels with cardiovascular traits in individuals of European and African Ancestry. Circ
Genom Precis Med. 2021;14(4):e003354.
Scipione CA, McAiney JT, Simard DJ, et al. Characterization of the I4399M variant of
apolipoprotein(a): implications for altered prothrombotic properties of lipoprotein(a). J
Thromb Haemost. 2017;15:1834–44.
Simon DI, Fless GM, Scanu AM, Loscalzo J. Tissue-type plasminogen activator binds to and
is inhibited by surface-bound lipoprotein(a) and low-density lipoprotein. Biochemistry.
1991;30:6671–7.
Skuza AA, Polak M, Undas A. Elevated lipoprotein(a) as a new risk factor of cerebral venous sinus
thrombosis: association with fibrin clot properties. J Thromb Thrombolysis. 2019;47:8–15.
Sofi F, Marcucci R, Abbate R, Gensini GF, Prisco D. Lipoprotein (a) and venous thromboembo-
lism in adults: a meta-analysis. Am J Med. 2007;120:728–33.
Stefanutti C, Mazza F, Pasqualetti D, et al. Lipoprotein apheresis downregulates IL-1 alpha, IL-6
and TNF-alpha mRNA expression in severe dyslipidaemia. Atheroscler Suppl. 2017;30:200–8.
Sticchi E, Magi A, Kamstrup PR, et al. Apolipoprotein(a) kringle-IV type 2 copy number variation
is associated with venous thromboembolism. PLoS One. 2016;11:e0149427.
Stiekema LCA, Stroes ESG, Verweij SL, et al. Persistent arterial wall inflammation in patients with
elevated lipoprotein(a) despite strong low-density lipoprotein cholesterol reduction by pro-
protein convertase subtilisin/kexin type 9 antibody treatment. Eur Heart J. 2019;40:2775–81.
Strang AC, Bisoendial RJ, Kootte RS, et al. Pro-atherogenic lipid changes and decreased
hepatic LDL receptor expression by tocilizumab in rheumatoid arthritis. Atherosclerosis.
2013;229:174–81.
Tabas I, Li Y, Brocia RW, Xu SW, Swenson TL, Williams KJ. Lipoprotein lipase and sphingo-
myelinase synergistically enhance the association of atherogenic lipoproteins with smooth
muscle cells and extracellular matrix. A possible mechanism for low density lipoprotein and
lipoprotein(a) retention and macrophage foam cell formation. J Biol Chem. 1993;268:20419–32.
Takami S, Yamashita S, Kihara S, et al. Lipoprotein(a) enhances the expression of intercel-
lular adhesion molecule-1 in cultured human umbilical vein endothelial cells. Circulation.
1998;97:721–8.
Tsimikas S. A test in context: lipoprotein(a): diagnosis, prognosis, controversies, and emerging
therapies. J Am Coll Cardiol. 2017;69:692–711.
Tsimikas S, Fazio S, Ferdinand KC, et al. NHLBI Working Group recommendations to reduce
lipoprotein(a)-mediated risk of cardiovascular disease and aortic stenosis. J Am Coll Cardiol.
2018;71:177–92.
206 M. S. Safarova and P. M. Moriarty
Tsironis LD, Mitsios JV, Milionis HJ, Elisaf M, Tselepis AD. Effect of lipoprotein (a) on plate-
let activation induced by platelet-activating factor: role of apolipoprotein (a) and endogenous
PAF-acetylhydrolase. Cardiovasc Res. 2004;63:130–8.
Tsurumi Y, Nagashima H, Ichikawa K, Sumiyoshi T, Hosoda S. Influence of plasma lipoprotein (a)
levels on coronary vasomotor response to acetylcholine. J Am Coll Cardiol. 1995;26:1242–50.
Undas A, Stepien E, Tracz W, Szczeklik A. Lipoprotein(a) as a modifier of fibrin clot permeability
and susceptibility to lysis. J Thromb Haemost. 2006;4:973–5.
van der Valk FM, Bekkering S, Kroon J, et al. Oxidized phospholipids on lipoprotein(a) elicit
arterial wall inflammation and an inflammatory monocyte response in humans. Circulation.
2016;134:611–24.
van Wijk DF, Sjouke B, Figueroa A, et al. Nonpharmacological lipoprotein apheresis reduces arte-
rial inflammation in familial hypercholesterolemia. J Am Coll Cardiol. 2014;64:1418–26.
von Depka M, Nowak-Gottl U, Eisert R, et al. Increased lipoprotein (a) levels as an independent
risk factor for venous thromboembolism. Blood. 2000;96:3364–8.
von Zychlinski A, Kleffmann T, Williams MJ, McCormick SP. Proteomics of Lipoprotein(a) identi-
fies a protein complement associated with response to wounding. J Proteome. 2011;74:2881–91.
Wade DP, Clarke JG, Lindahl GE, et al. 5′ control regions of the apolipoprotein(a) gene and mem-
bers of the related plasminogen gene family. Proc Natl Acad Sci U S A. 1993;90:1369–73.
Willeit P, Ridker PM, Nestel PJ, 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;392:1311–20.
Zimman A, Mouillesseaux KP, Le T, et al. Vascular endothelial growth factor receptor 2 plays a
role in the activation of aortic endothelial cells by oxidized phospholipids. Arterioscler Thromb
Vasc Biol. 2007;27:332–8.
Chapter 12
The Kidney Is the Heart of the
Organs: Its Role in Lp(a) Physiology
and Pathophysiology
Hans Dieplinger
Introduction
Definition of Lipoprotein(a)
H. Dieplinger (*)
Department of Genetics, Institute of Genetic Epidemiology, Medical University of Innsbruck,
Innsbruck, Austria
e-mail: [email protected]
Metabolism of Lipoprotein(a)
Since the discovery of Lp(a) 60 years ago, tremendous effort has been invested in
the elucidation of its molecular, cellular, and metabolic pathways. Despite an over-
whelming body of experimental evidence from various in vitro and in vivo systems,
the metabolism of this enigmatic lipoprotein still remains poorly understood
(Chemello et al. 2022).
Kinetic studies in vivo using stable isotope tracers and compartmental modeling
demonstrated that in individuals with a wide range of plasma Lp(a) concentrations,
the isotopic tracer curves for Lp(a)-apoB-100 and Lp(a)-apo(a) were essentially
identical, with similar contour and area-under-curve suggesting intracellular assem-
bly of Lp(a) (Watts et al. 2018). This notion was confirmed in a further in vivo study
in healthy individuals (Frischmann et al. 2012) and another kinetic study of statin-
treated patients with elevated and normal Lp(a) concentrations (Ma et al. 2019).
These kinetic data therefore generally support an intracellular assembly of Lp(a)
suggesting that newly synthesized Lp(a)-apoB-100 and Lp(a)-apo(a) are secreted as
a holoparticle with coupled apo(a) and apoB100 residence times in the circulation.
Controversial results of an extracellular Lp(a) assembly are possibly due to inap-
propriate cellular or animal models.
The mechanisms of Lp(a) clearance from the circulation also remain unclear and
controversial (McCormick and Schneider 2019). Without doubt is the liver the
major site of Lp(a) clearance followed to a much lesser extent by the kidney and the
arterial wall (Cain et al. 2005; Hrzenjak et al. 2003). Renal arterio-venous differ-
ences in Lp(a) concentrations suggest that the kidney can extract substantial amounts
of Lp(a) from the circulation (Kronenberg et al. 1997). A metabolic role for the
kidney is further supported by the inverse correlation between plasma Lp(a) and
glomerular filtration rate (GFR), with a significant increase in Lp(a) in patients with
more advanced chronic kidney disease (CKD) (Kronenberg 2014a) and by in vivo
kinetic studies demonstrating diminished clearance of Lp(a) in CKD patients treated
with hemodialysis (Frischmann et al. 2007). Further support for a possibly direct
catabolic function of the kidney for Lp(a) came from the discovery of fragments of
apo(a) in human urine and the decreased urinary excretion of apo(a) in patients with
renal dysfunction (Kostner et al. 1996; Mooser et al. 1996).
Immune-Histochemical Studies
a b c
d e f
Fig. 12.1 Immunoreactivity of apo(a) in glomeruli of different tissue sections. Lp(a) plasma con-
centrations and apo(a) isoforms are indicated. The intensity of apo(a) staining in the glomeruli
depends on Lp(a) concentrations (a–f). HMW, high molecular weight apo(a) phenotype; LMW,
low molecular weight apo(a) phenotype; bars 20 μm
12 The Kidney Is the Heart of the Organs: Its Role in Lp(a) Physiology… 211
a b
c d
Fig. 12.2 Immunoreactivity of apo(a) in the glomerulum shows apo(a) in mesangial cells (a).
Higher magnification of rectangle from picture a (b). Granular pattern of apo(a) staining in main
cell body and processes of mesangial cells, (c, d) Mesangial cells with typical staining. Bars 10 μm
found on the walls of the capillaries between tubuli and the blood vessels (Fig. 12.3e–
g) and on the membranes of erythrocytes. Similar to apo(a), no positive staining was
ever observed for apoB in proximal and distal convolute tubuli (Fig. 12.3h), as well
as in collecting tubuli. The negative controls did not show any staining (not shown),
but staining of a tissue sample from a patient with very low Lp(a) plasma concentra-
tion demonstrated apoB immunoreactivity (not shown).
Apo(a) and apoB colocalized in mesangial cells (Fig. 12.4), capillaries, and
blood vessels (data not shown). The negative controls did not show any non-specific
staining (not shown).
Mesangial cells contribute to the regulation of glomerular filtration, produce
mesangial matrix, and are continuously exposed to the plasma compartment, only
separated from the capillary lumen by a fenestrated endothelium without interven-
ing basement membrane (Latta 1992). Mesangial cells participate in a number of
glomerular diseases and it is still a matter of debate whether lipids/lipoproteins
selectively enhance mesangial matrix synthesis, proliferation of human mesangial
cells, and foam cell formation—that may induce renal damage or diseases caused
by lipid/lipoprotein deposition—induce injuries of mesangial cells (Gyebi et al.
2012; Mondorf et al. 1999; Wheeler et al. 1994). It has been postulated that the
interaction between plasma lipoproteins and mesangial cells plays a major role in
212 H. Dieplinger
a e
b f
c g
d h
Fig. 12.3 Immuno-peroxidase staining of apo(a) (a–d) and apoB (e–h) in different regions of the
human kidney tissue: (a, e) capillaries in glomeruli, (b, f) capillaries between tubuli, (c, g) blood
vessel, (d, h) proximal (p) and distal (d) convoluted tubuli (DIC image). Bars: (a, b, e, f) 10 μm;
(c, d, g, h) 20 μm
12 The Kidney Is the Heart of the Organs: Its Role in Lp(a) Physiology… 213
c
214 H. Dieplinger
Due to the structural similarities between LDL and Lp(a), the LDL receptor (LDLR)
has been investigated and discussed extensively as a candidate receptor for Lp(a)
over the past decades. Strong arguments again a role of the LDLR for Lp(a) uptake
and degradation came from numerous reports that statins which enhance LDLR
expression and thereby markedly reduce LDL, have a neutral, or an even elevating
effect on plasma Lp(a) concentrations (Khera et al. 2014; Tsimikas et al. 2020;
Willeit et al. 2018). Unexpectedly, PCSK9 inhibitors, which also increase the cell
surface expression of the LDLR, via an inhibition of LDLR intracellular degrada-
tion, not only lower LDL-C by 50–60%, but also reduce plasma Lp(a) concentra-
tions by 20–30% (McKenney et al. 2012). This observation has renewed research
into the roles of PCSK9 and of the LDLR in mediating the clearance of Lp(a) from
the circulation.
Several earlier reports from in vitro studies suggested, however, a role of the
LDLR for Lp(a) uptake, as well. Initial reports showed that Lp(a) can bind to the
LDLR, albeit with a lower affinity than LDL (Armstrong et al. 1990; Havekes et al.
1981; Reblin et al. 1997; Snyder et al. 1992). Lp(a) was also proposed to associate
with LDL and undergo LDLR-mediated clearance via a “hitch-hiking” mechanism
(Hofer et al. 1997; Kostner 1993). In HepG2 and primary human fibroblasts, PCSK9
was shown to reduce the binding and the cellular uptake of Lp(a) via the LDLR
12 The Kidney Is the Heart of the Organs: Its Role in Lp(a) Physiology… 215
(Raal et al. 2014). In contrast, other studies found no significant role for the LDLR
in mediating the cellular uptake of Lp(a) in primary human hepatocytes and HepG2
cells (Sharma et al. 2017; Villard et al. 2016). Moreover, no significant difference
was found in the cellular uptake of Lp(a) in primary lymphocytes derived from
normolipemic individuals compared with patients with homozygous familial hyper-
cholesterolemia (FH) and complete absence of LDLR function (Chemello
et al. 2020).
Multiple alternative pathways for Lp(a) clearance using other receptors have
been proposed, as elegantly reviewed by McCormick and Schneider (2019). For
instance, in macrophages, the Toll-like receptor 2 (TLR2) acts as a receptor for
Lp(a)-bound oxidized phospholipids (oxPL). This observation is in line with a large,
although non-significant, genome-wide association study (GWAS) showing that
TLR2 is the only receptor associated with circulating Lp(a) concentrations (Mack
et al. 2017; Seimon et al. 2010). Likewise, the scavenger receptor BI (SR-BI) has
been shown to promote the selective uptake of Lp(a) cholesterol esters in cells and
in SR-BI transgenic and knockout mice (Yang et al. 2013).
Due to the high degree of glycosylation of apo(a), carbohydrate-binding proteins
(lectins), such as the asialoglycoprotein receptor (ASGPR), have also been shown
to act as Lp(a) receptors in mice (Hrzenjak et al. 2003), but not all findings are con-
sistent (Cain et al. 2005; Sharma et al. 2017). Given the strong homology between
apo(a) and plasminogen, the role of plasminogen receptors in mediating Lp(a)
clearance has been investigated (Tam et al. 1996). One of such receptors, the plas-
minogen receptor presenting a C terminal lysine (PLGRKT), was shown to mediate
the cellular uptake of Lp(a) by human hepatoma cells and primary human fibro-
blasts. This study also showed that the LDL component of Lp(a) undergoes lyso-
somal degradation whereas apo(a) traffics through recycling endosomes and is
re-secreted into the medium (Sharma et al. 2017). However, the concentration of
free apo(a) in human plasma is relatively low, which suggests minimal to no recy-
cling of apo(a) in the circulation.
Several other members of the LDLR family of receptors have also been proposed
to mediate whole Lp(a) particle cellular uptake. Thus, the VLDL receptor binds
apo(a) and allows the internalization and subsequent degradation of Lp(a) in mac-
rophages (Argraves et al. 1997). The LDLR-related protein 1 (LPR1) and megalin/
gp330 (known as LRP2) also play a role in Lp(a) binding, cellular uptake, and deg-
radation in vitro (Reblin et al. 1997; März et al. 1993; Niemeier et al. 1999). LRP8
is also able to bind Lp(a) at the plasma membrane, but it remains to be shown
whether this promotes cellular uptake and degradation of Lp(a) particles (Steyrer
and Kostner 1990). The cellular uptake of Lp(a) in HepG2 hepatoma cells was,
however, recently shown to be unaffected overexpressing either the VLDLR, LRP1,
or LRP8 (Romagnuolo et al. 2017).
Interestingly, GWAS studies could not identify a significant association between
any of the proposed receptors and Lp(a) concentrations except the LDL receptor.
This might be explained by the fact that Lp(a) also contains cholesterol and the
signal with the LDL receptor might stem from the cholesterol content of Lp(a)
(Mack et al. 2017; Hoekstra et al. 2021).
216 H. Dieplinger
In vivo kinetic studies using stable isotopes have been performed to investigate pos-
sible mechanisms underlying elevated Lp(a) concentrations in two different groups
of kidney disease (Fig. 12.5). These studies also revealed insights into a possible
role of the kidney in Lp(a) metabolism.
Patients with nephrotic syndrome (NS) revealed increased synthesis rates of
Lp(a) without changes in the fractional catabolic rate indicating an increased pro-
duction of Lp(a)—along with many other proteins—rather than a decreased catabo-
lism (de Sain-van der Velden et al. 1998). These results have to be, however, taken
cautiously since they are based on kinetic data from only five patients and five
controls with widely varying single values. Therefore, although mean values
between patients and controls look impressively different (see Fig. 12.5), produc-
tion rates do not differ significantly between these two groups after statistical evalu-
ation of the provided single data. Therefore, human kinetic studies should be
repeated with higher numbers of included patients/controls to confirm the previ-
ously published analysis. It has been demonstrated that in NS, patients lose a signifi-
cant amount of proteins via urine, and that the increased synthesis of Lp(a) might be
a result of compensation to keep up the oncotic pressure in the circulating blood.
In contrast, CKD patients treated by hemodialysis (HD, essentially lacking renal
function) showed similar synthesis rates for both apo(a) and apoB from Lp(a) between
hemodialysis and healthy controls (Fig. 12.5). The fractional catabolic rates (FCR)
for both components of Lp(a), however, were significantly lower in HD patients com-
pared with controls. This resulted in a much longer residence time of 8.9 days for
Lp(a)-apo(a) and 12.9 days for Lp(a)-apoB in HD patients compared to controls (4.4
Fig. 12.5 In vivo kinetic studies using stable isotopes reveal different mechanisms leading to
increased Lp(a) concentrations in patients with nephrotic syndrome (NS) and chronic kidney dis-
ease (CKD) treated with hemodialysis (HD): whereas in NS patients Lp(a) production rates are
increased with no changes in catabolic rates, the situation in HD patients is opposite: Lp(a) con-
centrations are increased due to diminished catabolic rates and not synthesis. For NS patients,
kinetic parameters are given as Lp(a) total protein, for HD patients as Lp(a)-apo(a). Each bar rep-
resents mean ± standard error. Data for NS patients are taken from de Sain-van der Velden et al.
(1998), those for HD patients from Frischmann et al. (2007)
12 The Kidney Is the Heart of the Organs: Its Role in Lp(a) Physiology… 217
and 3.9 days, respectively) (Frischmann et al. 2007). These results suggest, together
with the discovery of apo(a) immunostaining in healthy human kidney tissue (see
section “Immune-Histochemical Studies”), a possible catabolic function for Lp(a) of
the human kidney. The prolonged residence time of Lp(a) in HD patients might sub-
stantially contribute to the high risk of atherosclerosis in these patients (see below).
Parra et al. reported for the first time in 1987 elevated Lp(a) concentrations in hemo-
dialysis patients (Parra et al. 1987). Since then, interest in the role of the kidney in
the metabolism of Lp(a) has steadily increased as documented in the comprehensive
review articles (Kronenberg et al. 1996; Kronenberg 2014a; Hopewell et al. 2018).
The earliest report related to this topic came from Papadopoulos et al., who described
a higher frequency of a second pre-beta band in agarose gel electrophoresis in
hemodialysis patients than in controls (Papadopoulos et al. 1980). Numerous stud-
ies have since then been published related to Lp(a) in nephrotic syndrome, CKD, or
kidney transplantation (see a recent review by Enkhmaa and Berglund (2022)).
Increased Lp(a) concentrations have been observed in patients with reduced kidney
function characterized by impaired glomerular filtration rates (GFR) (Catena et al.
2015; Kovesdy et al. 2002; Kronenberg et al. 2000; Lin et al. 2014; Milionis et al.
218 H. Dieplinger
1999; Sechi et al. 1998). In only three of these studies, apo(a) isoforms were ana-
lyzed in addition to plasma Lp(a) concentrations (Kronenberg et al. 2000; Milionis
et al. 1999; Sechi et al. 1998). Kronenberg et al. examined the association between
kidney function, Lp(a) plasma concentrations, and apo(a) isoform size in multi-
center design in 227 non-nephrotic patients with different degrees of kidney impair-
ment. Lp(a) concentrations were significantly higher in patients with kidney disease
compared with 227 age-, sex- and apo(a)-isoform-matched controls (Kronenberg
et al. 2000). Lp(a) were increased already in the earliest stages of kidney impair-
ment before GFR starts to decrease. Kidney function was inversely related with
Lp(a) concentrations, independent of the initial kidney disease. Most remarkably,
the inverse association between Lp(a) values and kidney function was only seen in
the subgroup of patients with HMW apo(a) isoforms, in line with observations in
hemodialyzed CKD patients (see section “Chronic Kidney Disease Treated by
Hemodialysis or Peritoneal Dialysis” (Dieplinger et al. 1993; Kronenberg
et al. 1995)).
Inverse correlations between Lp(a) concentrations and GFR were also found in
the Penn Diabetes Heart Study based on 1.852 patients with mild kidney impair-
ment (Lin et al. 2014) and in a population study involving 7.675 individuals from
different ethnic backgrounds, particularly in non-Hispanic blacks, eventually sug-
gesting ethnic differences (Kovesdy et al. 2002).
However, the observed association between Lp(a) and GFR in the above-
mentioned studies could not be confirmed by others: there was no significant asso-
ciation described in 804 individuals with stage 3–4 CKD and also no suggestion of
an interaction with apo(a) isoform size (Uhlig et al. 2005). Furthermore, a study of
87 kidney donors whose average kidney function was reduced from a GFR of 112
before donation to 72 mL/min/1.73 m2 one year later showed no significant differ-
ence in Lp(a) plasma values as a result of donation (Doucet et al. 2016). The reason
for the described discrepancies remains unclear; an explanation for the findings in
kidney transplant patients may be caused by an influence of immunosuppressive
medications on Lp(a) concentrations.
et al. 1993; Kronenberg et al. 1995; Barbagallo et al. 1993; Gault et al. 1995; Shoji
et al. 1992; Webb et al. 1993; Buggy et al. 1993; Irish et al. 1992; Anwar et al. 1993;
Murphy et al. 1992; Thillet et al. 1994; Wanner et al. 1995). Only one study reported
lower Lp(a) values in CAPD patients (Kandoussi et al. 1992). In these studies, the
range of differences between controls and patients was extremely broad. These
inconsistent findings can be explained by the low number of patients and controls in
several studies together with an up to 1.000-fold inter-individual variation in Lp(a)
concentrations and the otherwise strong genetic control of Lp(a) concentrations.
These circumstances require large numbers of investigated individuals to reveal
reliable results as discussed earlier (Kronenberg et al. 1996; Kronenberg 2014b).
Only few studies with an adequate number of patients have determined apo(a)
isoforms in addition to Lp(a) concentrations (Dieplinger et al. 1993; Auguet et al.
1993; Hirata et al. 1993; Wanner et al. 1995).
To overcome the limitations of small sample size, Kronenberg et al. performed a
large multicenter study that included 534 hemodialysis and 168 CAPD patients
(Kronenberg et al. 1995). Both patient groups showed significantly elevated Lp(a)
levels in comparison with the controls. Lp(a) values were significantly higher in
patients treated with CAPD than with hemodialysis. Notably, the elevations on
Lp(a) in hemodialysis and CAPD patients were less pronounced than in several
other small studies. Consideration of apo(a) phenotypes revealed that the increased
concentration of Lp(a) was not explained by different frequencies of apo(a) iso-
forms between patients and controls confirming and extending earlier findings
(Dieplinger et al. 1993). Therefore, elevated Lp(a) values in CKD are caused by the
disease and are not due a higher frequency of LMW apo(a) phenotypes in patients.
The reason for the selective elevation of Lp(a) levels in HMW isoforms in both
treatment groups is presently unclear.
Similar to nephrotic syndrome, the markedly elevated Lp(a) concentrations in
CAPD patients are probably caused by the high loss of protein, in this case through
the dialysate fluid as demonstrated by Kronenberg et al. (1995). A generally
increased hepatic synthesis and secretion of lipoproteins including Lp(a) is the most
likely reason for their higher Lp(a) values. This increased synthesis of Lp(a) might
be responsible for the trend to higher Lp(a) values in CAPD patients with LMW
apo(a) isoforms, which, however, did not reach statistical significance.
Once the final CKD stage is reached, the cause of kidney disease has no influ-
ence on Lp(a) plasma concentrations (Kronenberg et al. 1995). This observation
was confirmed by a study including hemodialysis, CAPD, and renal transplant
patients by reporting similar Lp(a) values in patients with and without insulin-
dependent diabetes mellitus (Gault et al. 1995).
The mechanism underlying Lp(a) elevation in CKD is still not fully understood.
The rapid decrease of Lp(a) following renal transplantation, as outlined in section
“Kidney Transplantation”, argues against an elevation induced by an acute phase
reaction, as was suggested earlier (Levine and Gordon 1995). Human kinetic studies
in various groups of these patients have been performed to examine whether this
elevation is caused by synthesis or catabolism. Further mainly kinetic studies are
necessary to shed light on the apo(a)-isoform-specific elevation of Lp(a) and its pos-
sible clinical impact.
220 H. Dieplinger
Kidney Transplantation
Findings of elevated Lp(a) concentrations in various patient groups with CKD led
several researchers to study the influence of kidney transplantation on Lp(a) plasma
concentrations. Black and Wilcken were the first to observe a highly significant
decrease in Lp(a) in 20 patients following renal transplantation (Black and Wilcken
1992). The results from several subsequent studies were not consistent, probably
reflecting differences in the study design. All prospective longitudinal studies
clearly showed a decrease in Lp(a) following transplantation (Gault et al. 1995;
Azrolan et al. 1994; Kronenberg et al. 1993, 1994a; Murphy and McNamee 1992;
Murphy et al. 1995; Segarra et al. 1995; Yang et al. 1994). Lp(a) changes were inde-
pendent of the modality of immunosuppressive therapy.
Lp(a) decreased after kidney transplantation in CKD patients, previously treated
by CAPD, independently of their apo(a) isoform. In contrast, in previously hemo-
dialyzed patients, Lp(a) declined after kidney transplantation only in those with
large apo(a) isoforms (Enkhmaa et al. 2016; Kerschdorfer et al. 1999; Kronenberg
et al. 1994a, 2003; Rosas et al. 2008). These findings are in line with the previously
described increased Lp(a) concentrations depending on the apo(a) size.
These results, together with those of kinetic studies in hemodialysis patients
(Frischmann et al. 2007), are a further convincing indication of a metabolic role of
the kidney in Lp(a) catabolism and that the observed Lp(a) changes are due to loss
of functional kidney tissue.
Acknowledgments Work from the author described in this overview has been supported by
grants from the Austrian Science Fund (P12358-MED) and the Jubiläumsfonds of the Austrian
National Bank (6721/4).
The author highly appreciates the critical reading of this review manuscript by Florian
Kronenberg as well as fruitful and long-lasting collaborations with many scientific colleagues and
technical assistants including H Andersson, M Auinger, D Bach, U Beisiegel, C Bösmüller, JH
Bräsen, B Dieplinger, N Donarski, E Dosch, C Drechsler, A von Eckardstein, G Friedrich, ME
Frischmann, B Grabensee, H Graf, E Gröchenig, A Gruber, M Hohenegger, F Hoppichler, E Hoye,
K Huber, K Ikewaki, H Kathrein, L Kerschdorfer, B Kollerits, P König, GM Kostner, KM Kostner,
V Krane, F Kronenberg, MF Kronenberg, C Lackner, C Lamina, U Lang, I Leiter, K Lhotta, A
Lingenhel, EM Lobentanz, R Margreiter, W März, G Maurer, HJ Menzel, N Moes, U Neyer, D
Öfner, G Pinter, A Pribasnig, B Rantner, T Reblin, J Reitinger, P Riegler, E Ritz, H Salmhofer, W
Salvenmoser, C Sandholzer, JR Schäfer, K Scheiber, U Scheidle, M Schober, JP Schwaiger, H
Schweer, RA Stahl, T Stefenelli, W Sturm, F Thaiss, J Thiery, E Trenkwalder, G Utermann, C
Wanner, M Wieshofer, G Wolf.
My special thanks go to Linda Fineder for a tremendous 30-year-long collaboration.
12 The Kidney Is the Heart of the Organs: Its Role in Lp(a) Physiology… 223
References
Anwar N, Bhatnagar D, Short CD, Mackness MI, Durrington PN, Prais H, et al. Serum lipoprotein
(a) concentrations in patients undergoing continuous ambulatory peritoneal dialysis. Nephrol
Dial Transplant. 1993;8:71–4.
Argraves KM, Kozarsky KF, Fallon JT, Harpel PC, Strickland DK. The atherogenic lipoprotein
Lp(a) is internalized and degraded in a process mediated by the VLDL receptor. J Clin Invest.
1997;100:2170–81.
Armstrong VW, Harrach B, Robenek H, Helmhold M, Walli AK, Seidel D. Heterogeneity of
human lipoprotein Lp[a]: cytochemical and biochemical studies on the interaction of two Lp[a]
species with the LDL receptor. J Lipid Res. 1990;31:429–41.
Arsenault BJ, Kamstrup PR. Lipoprotein(a) and cardiovascular and valvular diseases: a genetic
epidemiological perspective. Atherosclerosis. 2022;349:7–16. https://doi.org/10.1016/j.
atherosclerosis.2022.04.015.
Auguet T, Sentí M, Rubies-Prat J, Pelegrí A, Pedro-Botet J, Nogués X, et al. Serum lipoprotein(a)
concentration in patients with chronic renal failure receiving hemodialysis: influence of
apolipoprotein(a) genetic polymorphism. Nephrol Dial Transplant. 1993;8:1099–103.
Azrolan N, Brown CD, Thomas L, Hayek T, Zhao ZH, Roberts KG, et al. Cyclosporin A has
divergent effects on plasma LDL cholesterol (LDL-C) and lipoprotein(a) [Lp(a)] levels in renal
transplant recipients: evidence for renal involvement in the maintenance of LDL-C and the ele-
vation of Lp(a) concentrations in hemodialysis patients. Arterioscler Thromb. 1994;14:1393–8.
Bajaj A, Damrauer SM, Anderson AH, Xie D, Budoff MJ, Go AS, et al. Lipoprotein(a) and risk
of myocardial infarction and death in chronic kidney disease: findings from the CRIC study
(chronic renal insufficiency cohort). Arterioscler Thromb Vasc Biol. 2017;37(10):1971–8.
https://doi.org/10.1161/ATVBAHA.117.309920.
Barbagallo CM, Averna MR, Scafidi V, Galione A, Notarbartolo A. Increased lipoprotein (a) levels
in subjects with chronic renal failure on hemodialysis. Nephron. 1992;62:471–2.
Barbagallo CM, Averna MR, Sparacino V, Galione A, Caputo F, Scafidi V, et al. Lipoprotein (a)
levels in end-stage renal failure and renal transplantation. Nephron. 1993;64:560–4.
Black IW, Wilcken DEL. Decreases in apolipoprotein(a) after renal transplantation: implications
for lipoprotein(a) metabolism. Clin Chem. 1992;38:353–7.
Bonen DK, Hausman AML, Hadjiagapiou C, Skarosi SF, Davidson NO. Expression of a recombi-
nant apolipoprotein(a) in HepG2 cells. Evidence for intracellular assembly of lipoprotein(a). J
Biol Chem. 1997;272:5659–67.
Brown CD, Azrolan N, Thomas L, Roberts KG, Bostom A, Zhao ZH, et al. Reduction of
lipoprotein(a) following treatment with lovastatin in patients with unremitting nephrotic syn-
drome. Am J Kidney Dis. 1995;26:170–7.
Brunner C, Lobentanz EM, Petho-Schramm A, Ernst A, Kang C, Dieplinger H, et al. The num-
ber of identical kringle IV repeats in apolipoprotein(a) affects its processing and secretion by
HepG2 cells. J Biol Chem. 1996;271:32403–10.
Buggy D, Breathnach A, Keogh B, Cooke T, Feely J. Lipoprotein(a) and treatment of chronic renal
disease. J Intern Med. 1993;234:453–5.
Cain WJ, Millar JS, Himebauch AS, Tietge UJ, Maugeais C, Usher D, et al. Lipoprotein [a] is
cleared from the plasma primarily by the liver in a process mediated by apolipoprotein [a]. J
Lipid Res. 2005;46(12):2681–91. https://doi.org/10.1194/jlr.M500249-JLR200.
Catena C, Colussi G, Nait F, Pezzutto F, Martinis F, Sechi LA. Early renal failure as a cardiovascu-
lar disease: focus on lipoprotein(a) and prothrombotic state. World J Nephrol. 2015;4(3):374–8.
https://doi.org/10.5527/wjn.v4.i3.374.
Cauza E, Kletzmaier J, Bodlaj G, Dunky A, Herrmann W, Kostner K. Relationship of non-LDL-
bound apo(a), urinary apo(a) fragments and plasma Lp(a) in patients with impaired renal func-
tion. Nephrol Dial Transplant. 2003;18(8):1568–72. https://doi.org/10.1093/ndt/gfg181.
224 H. Dieplinger
Chemello K, Beeske S, Trang Tran TT, Blanchard V, Villard EF, Poirier B, et al. Lipoprotein(a)
cellular uptake ex vivo and hepatic capture in vivo is insensitive to PCSK9 inhibition
with alirocumab. JACC Basic Transl Sci. 2020;5(6):549–57. https://doi.org/10.1016/j.
jacbts.2020.03.008.
Chemello K, Chan DC, Lambert G, Watts GF. Recent advances in demystifying the metab-
olism of lipoprotein(a). Atherosclerosis. 2022;349:82–91. https://doi.org/10.1016/j.
atherosclerosis.2022.04.002.
Chiesa G, Hobbs HH, Koschinsky ML, Lawn RM, Maika SD, Hammer RE. Reconstitution of
lipoprotein(a) by infusion of human low density lipoprotein into transgenic mice expressing
human apolipoprotein(a). J Biol Chem. 1992;267:24369–74.
Chronic Kidney Disease Prognosis Consortium, Matsushita K, van der Velde M, Astor BC,
Woodward M, Levey AS, et al. Association of estimated glomerular filtration rate and albu-
minuria with all-cause and cardiovascular mortality in general population cohorts: a col-
laborative meta-analysis. Lancet. 2010;375(9731):2073–81. https://doi.org/10.1016/
S0140-6736(10)60674-5.
Coassin S, Kronenberg F. Lipoprotein(a) beyond the kringle IV repeat polymorphism: the com-
plexity of genetic variation in the LPA gene. Atherosclerosis. 2022;349:17–35. https://doi.
org/10.1016/j.atherosclerosis.2022.04.003.
Cressman MD, Heyka RJ, Paganini EP, O’Neil J, Skibinski CI, Hoff HF. Lipoprotein(a) is an
independent risk factor for cardiovascular disease in hemodialysis patients. Circulation.
1992;86:475–82.
Cressman MD, Abood D, O’Neil J, Hoff HF. Lp(a) and premature mortality during chronic hemo-
dialysis treatment. Chem Phys Lipids. 1994;67-68:419–27.
de Sain-van der Velden MG, Reijngoud DJ, Kaysen GA, Gadellaa MM, Voorbij H, Stellaard F,
et al. Evidence for increased synthesis of lipoprotein(a) in the nephrotic syndrome [in process
citation]. J Am Soc Nephrol. 1998;9:1474–81.
Dieplinger H, Lackner C, Kronenberg F, Sandholzer C, Lhotta K, Hoppichler F, et al. Elevated
plasma concentrations of lipoprotein(a) in patients with end-stage renal disease are not related
to the size polymorphism of apolipoprotein(a). J Clin Invest. 1993;91:397–401.
Docci D, Baldrati L, Capponcini C, Feletti C. Serum lipoprotein(a) (Lpa(a)) in haemodialysis
patients. Nephrol Dial Transplant. 1994;9(6):733–4. https://doi.org/10.1093/ndt/9.6.733b.
Doucet B, Kostner K, Kaiser O, Hawley C, Isbel N. Live donor study—implications of kidney
donation on cardiovascular risk with a focus on lipid parameters including lipoprotein a.
Nephrology (Carlton). 2016;21(10):901–4. https://doi.org/10.1111/nep.12792.
Emanuele E, Lusignani LS, Peros E, Montagna G, D’Angelo A, Montagna L, et al. Lipoprotein(a)-
associated atherothrombotic risk in hemodialysis patients. Am J Nephrol. 2004;24(2):221–9.
https://doi.org/10.1159/000077293.
Enkhmaa B, Berglund L. Non-genetic influences on lipoprotein(a) concentrations. Atherosclerosis.
2022;349:53–62. https://doi.org/10.1016/j.atherosclerosis.2022.04.006.
Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and
medical conditions. J Lipid Res. 2016;57(7):1111–25. https://doi.org/10.1194/jlr.R051904.
Erqou S, Kaptoge S, Perry PL, Di Angelantonio E, Thompson A, White IR, et al. Lipoprotein(a)
concentration and the risk of coronary heart disease, stroke, and nonvascular mortality.
JAMA. 2009;302(4):412–23. https://doi.org/10.1001/jama.2009.1063.
Faucher C, Doucet C, Baumelou A, Chapman J, Jacobs C, Thillet J. Elevated lipoprotein (a) levels
in primary nephrotic syndrome. Am J Kidney Dis. 1993;22:808–13.
Fiorini F, Masturzo P, Mij M, Bertolini S. Lipoprotein(a) levels in hemodialysis patients: relation
to glucose intolerance and hemodialysis duration [letter]. Nephron. 1995;70:500–1.
Frischmann ME, Kronenberg F, Trenkwalder E, Schaefer JR, Schweer H, Dieplinger B, et al.
In vivo turnover study demonstrates diminished clearance of lipoprotein(a) in hemodialysis
patients. Kidney Int. 2007;71(10):1036–43.
Frischmann ME, Ikewaki K, Trenkwalder E, Lamina C, Dieplinger B, Soufi M, et al. In vivo
stable-isotope kinetic study suggests intracellular assembly of lipoprotein(a). Atherosclerosis.
2012;225(2):322–7. https://doi.org/10.1016/j.atherosclerosis.2012.09.031.
12 The Kidney Is the Heart of the Organs: Its Role in Lp(a) Physiology… 225
Gambhir JK, Kalra OP, Khaira A, Kaur H. Association between high molecular weight apolipopro-
tein isoforms and lipoprotein levels in advanced chronic kidney disease and the effect of hemo-
dialysis. Indian J Nephrol. 2013;23(1):18–23. https://doi.org/10.4103/0971-4065.107189.
Gault MH, Longerich LL, Purchase L, Harnett J, Breckenridge C. Comparison of Lp(a) concentra-
tions and some potential effects in hemodialysis, CAPD, transplantation, and control groups,
and review of the literature. Nephron. 1995;70:155–70.
Greiber S, Krämer-Guth A, Pavenstädt H, Gutenkunst M, Schollmeyer P, Wanner C. Effects
of lipoprotein(a) on mesangial cell proliferation and viability. Nephrol Dial Transplant.
1996;11:778–85.
Gröne EF, Abboud HE, Höhne M, Walli AK, Gröne H-J, Stüker D, et al. Actions of lipoproteins
in cultured human mesangial cells: modulation by mitogenic vasoconstrictors. Am J Physiol.
1992;263:F686–F96.
Gyebi L, Soltani Z, Reisin E. Lipid nephrotoxicity: new concept for an old disease. Curr Hypertens
Rep. 2012;14(2):177–81. https://doi.org/10.1007/s11906-012-0250-2.
Haffner SM, Gruber KK, Aldrete G Jr, Morales PA, Stern MP, Tuttle KR. Increased lipoprotein(a)
concentrations in chronic renal failure. J Am Soc Nephrol. 1992;3:1156–62.
Havekes L, Vermeer BJ, Brugman T, Emeis J. Binding of Lp (a) to the low density lipoprotein
receptor of human fibroblast. FEBS Lett. 1981;132:169–73.
Heimann P, Josephson MA, Fellner SK, Thistlethwaite JR Jr, Stuart FP, Dasgupta A. Elevated
lipoprotein(a) levels in renal transplantation and hemodialysis patients. Am J Nephrol.
1991;11:470–4.
Hirata K, Kikuchi S, Saku K, Jimi S, Zhang B, Naito S, et al. Apolipoprotein(a) phenotypes and
serum lipoprotein(a) levels in maintenance hemodialysis patients with/without diabetes mel-
litus. Kidney Int. 1993;44:1062–70.
Hoekstra M, Chen HY, Rong J, Dufresne L, Yao J, Guo X, et al. Genome-wide association study
highlights APOH as a novel locus for lipoprotein(a) levels-brief report. Arterioscler Thromb
Vasc Biol. 2021;41(1):458–64. https://doi.org/10.1161/ATVBAHA.120.314965.
Hofer G, Steyrer E, Kostner GM, Hermetter A. LDL-mediated interaction of Lp[a] with HepG2
cells: a novel fluorescence microscopy approach. J Lipid Res. 1997;38:2411–21.
Hong SY, Yang DH. Lipoprotein(a) levels and fibrinolytic activity in patients with nephrotic syn-
drome. Nephron. 1995;69:125–30.
Hopewell JC, Haynes R, Baigent C. The role of lipoprotein (a) in chronic kidney disease. J Lipid
Res. 2018;59(4):577–85. https://doi.org/10.1194/jlr.R083626.
Hrzenjak A, Frank S, Wo X, Zhou Y, Van Berkel T, Kostner GM. Galactose-specific asialoglyco-
protein receptor is involved in lipoprotein (a) catabolism. Biochem J. 2003;376(Pt 3):765–71.
https://doi.org/10.1042/BJ20030932.
Irish AB, Hayes JM, Simons LA, Simons J, Savdie E. Lipoprotein(a) levels in chronic renal dis-
ease states, dialysis and transplantation. Aust N Z J Med. 1992;22:243–8.
Joven J, Simo JM, Vilella E, Camps J, Espinel E, Villabona C. Accumulation of atherogenic rem-
nants and lipoprotein(a) in the nephrotic syndrome: relation to remission of proteinuria. Clin
Chem. 1995;41:908–13.
Kandoussi A, Cachera C, Pagniez D, Dracon M, Fruchart JC, Tacquet A. Plasma level of lipopro-
tein Lp(a) is high in predialysis or hemodialysis, but not in CAPD. Kidney Int. 1992;42:424–5.
Karádi I, Romics L, Palos G, Doman J, Kaszas I, Hesz A, et al. Lp(a) lipoprotein concentration
in serum of patients with heavy proteinuria of different origin. Clin Chem. 1989;35:2121–3.
Kerschdorfer L, Konig P, Neyer U, Bosmuller C, Lhotta K, Auinger M, et al. Lipoprotein(a) plasma
concentrations after renal transplantation: a prospective evaluation after 4 years of follow-up.
Atherosclerosis. 1999;144(2):381–91.
Khera AV, Everett BM, Caulfield MP, Hantash FM, Wohlgemuth J, Ridker PM, et al.
Lipoprotein(a) concentrations, rosuvastatin therapy, and residual vascular risk: an analysis
from the JUPITER Trial (Justification for the Use of Statins in Prevention: an Intervention
Trial Evaluating Rosuvastatin). Circulation. 2014;129(6):635–42. https://doi.org/10.1161/
CIRCULATIONAHA.113.004406.
226 H. Dieplinger
McLean JW, Tomlinson JE, Kuang W-J, Eaton DL, Chen EY, Fless GM, et al. cDNA sequence of
human apolipoprotein (a) is homologous to plasminogen. Nature. 1987;300:132–7.
Menzel H-J, Dieplinger H, Lackner C, Hoppichler F, Lloyd JK, Muller DR, et al.
Abetalipoproteinemia with an ApoB-100-lipoprotein(a) glycoprotein complex in plasma.
Indication for an assembly defect. J Biol Chem. 1990;265:981–6.
Milionis HJ, Elisaf MS, Tselepis A, Bairaktari E, Karabina SA, Siamopoulos KC. Apolipoprotein(a)
phenotypes and lipoprotein(a) concentrations in patients with renal failure. Am J Kidney Dis.
1999;33:1100–6.
Mondorf UF, Piiper A, Herrero M, Olbrich HG, Bender M, Gross W, et al. Lipoprotein(a) stimu-
lates growth of human mesangial cells and induces activation of phospholipase C via pertussis
toxin-sensitive G proteins [in process citation]. Kidney Int. 1999;55:1359–66.
Mooser V, Marcovina S, White A, Hobbs HH. Kringle-containing fragments of apolipoprotein(a)
circulate in human plasma and are excreted into the urine. J Clin Invest. 1996;98:2414–24.
Murphy BG, McNamee PT. Apolipoprotein(a) concentration decreases following renal transplan-
tation. Nephrol Dial Transplant. 1992;7:174–5.
Murphy BG, McNamee P, Duly E, Henry W, Archbold P, Trinick T. Increased serum
apolipoprotein(a) in patients with chronic renal failure treated with continuous ambulatory
peritoneal dialysis. Atherosclerosis. 1992;93:53–7.
Murphy BG, Yong A, Brown JH, McNamee PT. Effect of immunosuppressive drug regime on car-
diovascular risk profile following kidney transplantation. Atherosclerosis. 1995;116(2):241–5.
https://doi.org/10.1016/0021-9150(95)05552-8.
Nakahara C, Nagata M, Watanabe T, Kobayashi K, Hamaguchi H, Kanemoto K, et al. Clinical
significance of apolipoprotein (a) deposition in kidney diseases of children. Clin Nephrol.
1999;52:131–8.
Niemeier A, Willnow T, Dieplinger H, Jacobsen C, Meyer N, Hilpert J, et al. Identification of
megalin/gp330 as a receptor for lipoprotein(a) in vitro. Arterioscler Thromb Vasc Biol.
1999;19:552–61.
Nordestgaard BG, Langsted A. Lipoprotein (a) as a cause of cardiovascular disease: insights
from epidemiology, genetics, and biology. J Lipid Res. 2016;57(11):1953–75. https://doi.
org/10.1194/jlr.R071233.
Ohashi H, Oda H, Ohno M, Watanabe S, Sakata S. Lipoprotein(a) as a risk factor for coronary
artery disease in hemodialysis patients. Kidney Int Suppl. 1999;71:S242–S4.
Oida K, Takai H, Maeda H, Takahashi S, Shimada A, Suzuki J, et al. Apolipoprotein(a) is present in
urine and its excretion is decreased in patients with renal failure. Clin Chem. 1992;38:2244–8.
Okura Y, Saku K, Hirata K, Zhang B, Liu R, Ogahara S, et al. Serum lipoprotein (a) levels in main-
tenance hemodialysis patients. Nephron. 1993;65:46–50.
Papadopoulos NM, Borer WZ, Elin RJ, Diamond LH. An abnormal lipoprotein in the serum of
uremic patients maintained on chronic hemodialysis. Ann Intern Med. 1980;92:634–5.
Parra HJ, Mezdour H, Cachera C, Dracon M, Tacquet A, Fruchard JC. Lp(a) lipoprotein in patients
with chronic renal failure treated by hemodialysis. Clin Chem. 1987;33:721.
Parsi D, Dracon M, Cachera C. Lipoprotein abnormalities in chronic hemodialysis patients.
Nephrol Dial Transplant. 1988;3:51–6.
Parsons DS, Reaveley DA, Pavitt DV, Misra M, Brown EA. Lipoprotein (a) levels in those with
high molecular weight apo (a) isoforms may remain low in a significant proportion of patients
with end-stage renal disease. Nephrol Dial Transplant. 2003;18(9):1848–53. https://doi.
org/10.1093/ndt/gfg276.
Querfeld U, Lang M, Friedrich JB, Kohl B, Fiehn W, Schärer K. Lipoprotein(a) serum lev-
els and apolipoprotein(a) phenotypes in children with chronic renal disease. Pediatr Res.
1993;34:772–6.
Raal FJ, Giugliano RP, Sabatine MS, Koren MJ, Langslet G, Bays H, et al. Reduction in lipo-
protein (a) with the PCSK9 monoclonal antibody evolocumab (AMG 145): a pooled analysis
of over 1300 patients in 4 phase 2 trials. J Am Coll Cardiol. 2014;63:1278–88. https://doi.
org/10.1016/j.jacc.2014.01.006.
12 The Kidney Is the Heart of the Organs: Its Role in Lp(a) Physiology… 229
Reblin T, Niemeier A, Meyer N, Willnow TE, Kronenberg F, Dieplinger H, et al. Cellular uptake
of lipoprotein[a] by mouse embryonic fibroblasts via the LDL receptor and the LDL receptor-
related protein. J Lipid Res. 1997;38:2103–10.
Reblin T, Donarski N, Fineder L, Brasen JH, Dieplinger H, Thaiss F, et al. Renal handling of
human apolipoprotein(a) and its fragments in the rat. Am J Kidney Dis. 2001;38:619–30.
Reyes-Soffer G, Ginsberg HN, Ramakrishnan R. The metabolism of lipoprotein (a): an ever-
evolving story. J Lipid Res. 2017;58(9):1756–64. https://doi.org/10.1194/jlr.R077693.
Romagnuolo R, Scipione CA, Marcovina SM, Gemin M, Seidah NG, Boffa MB, et al. Roles of
the low density lipoprotein receptor and related receptors in inhibition of lipoprotein(a) inter-
nalization by proprotein convertase subtilisin/kexin type 9. PLoS One. 2017;12(7):e0180869.
https://doi.org/10.1371/journal.pone.0180869.
Rosas S, Joffe M, Wolfe M, Brayman K, Rader DJ. Effects of renal replacement therapy on plasma
lipoprotein(a) levels. Am J Nephrol. 2008;28(3):361–5. https://doi.org/10.1159/000112225.
Sandholzer C, Saha N, Kark JD, Rees A, Jaross W, Dieplinger H, et al. Apo(a) isoforms predict risk
for coronary heart disease: a study in six populations. Arterioscler Thromb. 1992;12:1214–26.
Sato H, Suzuki S, Ueno M, Shimada H, Karasawa R, Nishi S-I, et al. Localization of
apolipoprotein(a) and B-100 in various renal diseases. Kidney Int. 1993;43:430–5.
Sechi LA, Zingaro L, De Carli S, Sechi G, Catena C, Falleti E, et al. Increased serum lipoprotein(a)
levels in patients with early renal failure. Ann Intern Med. 1998;129:457–61.
Segarra A, Chacón P, Martin M, Vilardell M, Vila J, Cotrina M, et al. Serum lipoprotein (a) levels
in patients with chronic renal failure—evolution after renal transplantation and relationship
with other parameters of lipoprotein metabolism: a prospective study. Nephron. 1995;69:9–13.
Seimon TA, Nadolski MJ, Liao X, Magallon J, Nguyen M, Feric NT, et al. Atherogenic lipids and
lipoproteins trigger CD36-TLR2-dependent apoptosis in macrophages undergoing endoplasmic
reticulum stress. Cell Metab. 2010;12(5):467–82. https://doi.org/10.1016/j.cmet.2010.09.010.
Sharma M, Redpath GM, Williams MJ, McCormick SP. Recycling of apolipoprotein(a) after
PlgRKT-mediated endocytosis of lipoprotein(a). Circ Res. 2017;120(7):1091–102. https://doi.
org/10.1161/CIRCRESAHA.116.310272.
Shlipak MG, Fried LF, Cushman M, Manolio TA, Peterson D, Stehman-Breen C, et al.
Cardiovascular mortality risk in chronic kidney disease: comparison of traditional and novel
risk factors. JAMA. 2005;293(14):1737–45. https://doi.org/10.1001/jama.293.14.1737.
Shoji T, Nishizawa Y, Nishitani H, Yamakawa M, Morii H. High serum lipoprotein(a) concentra-
tions in uremic patients treated with continuous ambulatory peritoneal dialysis. Clin Nephrol.
1992;38:271–6.
Snyder ML, Polacek D, Scanu AM, Fless GM. Comparative binding and degradation of
lipoprotein(a) and low density lipoprotein by human monocyte-derived macrophages. J Biol
Chem. 1992;267:339–46.
Stenvinkel P, Berglund L, Heimbürger O, Pettersson E, Alvestrand A. Lipoprotein(a) in nephrotic
syndrome. Kidney Int. 1993;44:1116–23.
Steyrer E, Kostner GM. Interaction of lipoprotein Lp[a] with the B/E-receptor: a study using iso-
lated bovine adrenal cortex and human fibroblast receptors. J Lipid Res. 1990;31:1247–53.
Suzuki S, Takahashi H, Sato H, Takashima N, Arakawa M, Gejyo F. Significance of glomerular
deposition of apolipoprotein (a) in various glomerulopathies. Am J Nephrol. 1997;17:499–504.
Takegoshi T, Kitoh C, Haba T, Hirai J, Wakasugi T, Saga T, et al. A study of the clinical signifi-
cance of lipoprotein (a) in nephrotic syndrome. Jpn J Med. 1991;30:21–5.
Takemura T, Yoshioka K, Aya N, Murakami K, Matumoto A, Itakura H, et al. Apolipoproteins
and lipoprotein receptors in glomeruli in human kidney diseases. Kidney Int. 1993;43:918–27.
Tam S-P, Zhang X, Koschinsky ML. Interaction of a recombinant form of apolipoprotein(a) with
human fibroblasts and with the human hepatoma cell line HepG2. J Lipid Res. 1996;37:518–33.
Thanassoulis G. Lipoprotein (a) in calcific aortic valve disease: from genomics to novel drug target
for aortic stenosis. J Lipid Res. 2016;57(6):917–24. https://doi.org/10.1194/jlr.R051870.
Thillet J, Doucet C, Issad B, Allouache M, Chapman JM, Jacobs C. Elevated LP(A) levels in
patients with end-stage renal disease. Am J Kidney Dis. 1994;23:620–1.
230 H. Dieplinger
Thomas ME, Freestone A, Varghese Z, Persaud JW, Moorhead JF. Lipoprotein(a) in patients with
proteinuria. Nephrol Dial Transplant. 1992;7:597–601.
Trenkwalder E, Gruber A, Konig P, Dieplinger H, Kronenberg F. Increased plasma concentrations
of LDL-unbound apo(a) in patients with end-stage renal disease. Kidney Int. 1997;52:1685–92.
Tsimikas S, Gordts P, Nora C, Yeang C, Witztum JL. Statin therapy increases lipoprotein(a) levels.
Eur Heart J. 2020;41(24):2275–84. https://doi.org/10.1093/eurheartj/ehz310.
Uhlig K, Wang SR, Beck GJ, Kusek JW, Marcovina SM, Greene T, et al. Factors associated with
lipoprotein(a) in chronic kidney disease. Am J Kidney Dis. 2005;45(1):28–38.
Utermann G. The mysteries of lipoprotein(a). Science. 1989;246:904–10.
Utermann G, Menzel HJ, Kraft HG, Duba HC, Kemmler HG, Seitz C. Lp(a) glycoprotein phe-
notypes. Inheritance and relation to Lp(a)-lipoprotein concentrations in plasma. J Clin Invest.
1987;80(2):458–65. https://doi.org/10.1172/JCI113093.
Vaziri ND. Disorders of lipid metabolism in nephrotic syndrome: mechanisms and consequences.
Kidney Int. 2016;90(1):41–52. https://doi.org/10.1016/j.kint.2016.02.026.
Villard EF, Thedrez A, Blankenstein J, Croyal M, Tran TT, Poirier B, et al. PCSK9 modulates the
secretion but not the cellular uptake of lipoprotein(a) ex vivo: an effect blunted by alirocumab.
JACC Basic Transl Sci. 2016;1(6):419–27. https://doi.org/10.1016/j.jacbts.2016.06.006.
Wanner C, Rader D, Bartens W, Krämer J, Brewer HB, Schollmeyer P, et al. Elevated plasma
lipoprotein(a) in patients with the nephrotic syndrome. Ann Intern Med. 1993;119:263–9.
Wanner C, Bartens W, Walz G, Nauck M, Schollmeyer P. Protein loss and genetic polymorphism of
apolipoprotein(a) modulate serum lipoprotein(a) in CAPD patients. Nephrol Dial Transplant.
1995;10:75–81.
Watts GF, Chan DC, Somaratne R, Wasserman SM, Scott R, Marcovina SM, et al. Controlled
study of the effect of proprotein convertase subtilisin-kexin type 9 inhibition with evolocumab
on lipoprotein(a) particle kinetics. Eur Heart J. 2018;39(27):2577–85. https://doi.org/10.1093/
eurheartj/ehy122.
Webb AT, Reaveley DA, O’Donnell M, O’Connor B, Seed M, Brown EA. Lipoprotein (a) in
patients on maintenance haemodialysis and continuous ambulatory peritoneal dialysis [see
comments]. Nephrol Dial Transplant. 1993;8:609–13.
Webb AT, Reaveley DA, O’Donnell M, O’Connor B, Seed M, Brown EA. Lipids and Apoprotein(a)
as risk factors for vascular disease in patients on renal replacement therapy. Nephrol Dial
Transplant. 1995;10:354–7.
Wheeler DC, Chana RS, Topley N, Petersen MM, Davies M, Williams JD. Oxidation of low density
lipoprotein by mesangial cells may promote glomerular injury. Kidney Int. 1994;45:1628–36.
White AL, Lanford RE. Cell surface assembly of lipoprotein(a) in primary cultures of baboon
hepatocytes. J Biol Chem. 1994;269:28716–23.
White AL, Rainwater DL, Hixson JE, Estlack LE, Lanford RE. Intracellular processing of apo(a)
in primary baboon hepatocytes. Chem Phys Lipids. 1994;67–68:123–33.
White AL, Guerra B, Wang J, Lanford RE. Presecretory degradation of apolipoprotein[a] is medi-
ated by the proteasome pathway. J Lipid Res. 1999;40:275–86.
Wilkinson J, Munro LH, Higgins JA. Apolipoprotein[a] is not associated with apolipoprotein B in
human liver. J Lipid Res. 1994;35:1896–901.
Willeit P, Ridker PM, Nestel PJ, Simes J, Tonkin AM, Pedersen TR, 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(10155):1311–20. https://doi.
org/10.1016/S0140-6736(18)31652-0.
Yang CW, Kim YS, Kim SY, Choi EJ, Chang YS, Yoon YS, et al. Lipoprotein(a) concentrations
in chronic renal failure and following renal transplantation. Transplant Proc. 1994;26:2082–3.
Yang XP, Amar MJ, Vaisman B, Bocharov AV, Vishnyakova TG, Freeman LA, et al. Scavenger
receptor-BI is a receptor for lipoprotein(a). J Lipid Res. 2013;54(9):2450–7. https://doi.
org/10.1194/jlr.M038877.
Chapter 13
Lp(a) as a Cardiovascular Risk Factor
Introduction
Epidemiological and genetic evidence has clearly shown that elevated levels of
lipoprotein(a) [Lp(a)] are causally linked with an increased risk of cardiovascular
disease (CVD). This has led to renewed interest in an “old” lipoprotein that, although
sharing structural similarities with LDL, is endowed with exclusive properties due
to the presence of apolipoprotein(a) [apo(a)], a protein with homology to plasmino-
gen (McLean et al. 1987). Lp(a) exerts multiple effects in CVD, as it can act simi-
larly to an LDL particle and enter the intima of the arterial wall, thus contributing to
atherosclerosis, but it can also inhibit fibrinolysis due to its homology with plas-
minogen and contribute to inflammation by mean of Lp(a)-associated oxidized
phospholipids (Koutsogianni et al. 2021).
Apo(a), encoded by the LPA gene, is a highly heterogeneous protein containing
multiple repeats of kringle 4 type 2 (KIV2). The number of repeats is genetically
determined by common copy-number variation within the LPA gene and is inversely
related to the plasma concentration of Lp(a), with isoforms containing fewer KIV2
repeats being associated with smaller Lp(a) lipoprotein size and higher circulating
levels. Two unique features of Lp(a) are its wide range of plasma level variation
A. Pirillo
Center for the Study of Atherosclerosis, E. Bassini Hospital, Milan, Italy
IRCCS MultiMedica, Milan, Italy
e-mail: [email protected]
A. L. Catapano (*)
IRCCS MultiMedica, Milan, Italy
Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano,
Milan, Italy
e-mail: [email protected]
(from <0.2 to >200 mg/dL, or <0.5 to 500 nmol/L), which for the most part reflects
genetic variations in LPA, and its profile of distribution in the population, which is
highly skewed with a long tail toward extremely high values, with ~20% of indi-
viduals showing Lp(a) levels >50 mg/dL (Nordestgaard et al. 2010). Elevated Lp(a)
levels can occur in individuals with otherwise normal lipid levels; the risk threshold
for Lp(a) is set at 50 mg/dL.
Being mostly genetically determined, circulating levels of Lp(a) are relatively
stable throughout life. Based on this observation, current European guidelines for
the management of dyslipidemias recommend measuring Lp(a) at least once in life
(Mach et al. 2020).
A large number of studies have established a causal relationship between Lp(a) and
CVD (Emerging Risk Factors Collaboration et al. 2009); above all, elevated Lp(a)
increases the risk of myocardial infarction (MI), stroke, and peripheral arterial dis-
ease, but its pathophysiological role appears to be more complex than that of
LDL. In fact, the mechanisms beyond this association likely involve both its LDL
particle-like features (promoting atherosclerosis) and plasminogen-like particle
(inhibiting fibrinolysis).
A stepwise increase in the risk of MI with increasing levels of Lp(a) was reported
in both genders in a general European population, with extreme levels of Lp(a)
(>95th percentile) predicting a threefold to fourfold increased MI risk (Kamstrup
et al. 2008). In agreement with this observation, another study reported a threefold
to fourfold higher prevalence of ASCVD and MI in adults having Lp(a) >99th per-
centile [median Lp(a) 460 nmol/L] compared to those with Lp(a) levels ≤20th per-
centile [median Lp(a) 7 nmol/L] (Nurmohamed et al. 2021). The incorporation of
Lp(a) into algorithms for CV risk assessment led to the increase of mean estimated
10-year risk and the upward reclassification of substantial percentages of patients,
both in primary and in secondary prevention (Nurmohamed et al. 2021). An inter-
esting observation reported in this study was a higher LDL-C goal attainment among
individuals with lower Lp(a) levels compared with those with high Lp(a) levels,
which may suggest that measured LDL-C in these patients mainly is the result of
high Lp(a) levels (Nurmohamed et al. 2021). It has been observed that a 15 mg/dL
(~0.39 nmol/L) increase in Lp(a) cholesterol determines a higher hazard ratio of CV
mortality compared with a corresponding increase in LDL cholesterol (1.18 vs
1.05), which may suggest that not only the cholesterol content in lipoprotein(a) is
pathogenic, but likely its unique protein apo(a) may play a relevant role as well
(Langsted et al. 2019).
Mendelian randomization studies have substantiated the causal role of Lp(a) in
CVD (Clarke et al. 2009; Kamstrup et al. 2009; Burgess et al. 2018; Lamina et al.
2019). Two common single-nucleotide polymorphisms (SNPs) have been strongly
associated with both increased levels of Lp(a) and increased risk of coronary
13 Lp(a) as a Cardiovascular Risk Factor 233
disease (Clarke et al. 2009); on the other hand, a 10 mg/dL lower genetically deter-
mined Lp(a) level was associated with a 5.8% lower risk of coronary heart disease
(CHD) (Burgess et al. 2018). The observation that a similar reduction in LDL-C will
translate into a 14.5% lower CHD risk has a relevant consequence on the magnitude
of Lp(a) level reduction required to provide a clinical benefit: a ~100 mg/dL Lp(a)
reduction concentration anticipates a CHD risk reduction similar to that achieved
with a ~39 mg/dL change in LDL-C level (Burgess et al. 2018) (Fig. 13.1). A similar
finding has been reported by another study (Lamina et al. 2019) (Fig. 13.1).
Elevated Lp(a) levels can also explain, at least in part, the residual CV risk com-
monly observed in patients with well-controlled LDL-C levels. A meta-analysis of
data from 29,069 patients included in seven placebo-controlled statin trials showed
that elevated baseline and on-statin Lp(a) levels conferred a significantly higher CV
risk, suggesting that statins do not impact the residual risk in patients with elevated
Lp(a) (Willeit et al. 2018) (Fig. 13.2). Among patients with recent ACS receiving
intensive or maximum-tolerated statin treatment, baseline Lp(a) levels predicted the
risk of MACE, nonfatal MI, and CHD and CV death, independent of baseline
LDL-C (Bittner et al. 2020). Although the reduction of MACE is mainly attributable
to the reduction in LDL-C (referred to as “corrected LDL-C”) across the range of
baseline Lp(a) levels, in patients with the highest baseline Lp(a) levels the contribu-
tion of Lp(a) reduction in reducing the risk of MACE was substantial (Bittner et al.
2020). Interestingly, moderately elevated plasma Lp(a) levels (≥15 mg/dL) appear
to confer an increased risk of all-cause mortality in patients with CAD (Liu
et al. 2021).
The role of Lp(a) in determining the residual CV risk is further supported by the
results of most recent trials evaluating therapies able to achieve considerable reduc-
tions in LDL-C levels. The FOURIER trial, which evaluated the clinical impact of
60
50
Genetic estimate
% CHD risk reduction
30 LDL-C Lp(a)
38,67 mg/dL 65,7-101,5 mg/dL
Trial estimate
20 (short-term clinical trials for
LDL-C)
10
0
0 20 40 60 80 100 120
Fig. 13.1 Estimates of CHD risk reduction with lowering of LDL-C or Lp(a) level
234 A. Pirillo and A. L. Catapano
1.8
Baseline
1.6 On-statin
Hazard ratio (95% CI)
1.4
1.2
0.8
15 to <30 mg/dL 30 to <50 mg/dL ≥50 mg/dL Lp(a) level
Fig. 13.2 Association of baseline and on-statin Lp(a) levels with incident CVD, age-adjusted, and
sex-adjusted according to different Lp(a) levels
evolocumab in patients with ASCVD, found that higher baseline levels of Lp(a)
were associated with an increased risk of coronary events, independent of LDL-C
levels (O'Donoghue et al. 2019). Furthermore, a post hoc analysis of the ODYSSEY
OUTCOMES trial observed that while patients having LDL-C levels ≥70 mg/dL
derive a clinical benefit from alirocumab treatment independent of Lp(a) levels, in
those having LDL-C ~70 mg/dL alirocumab treatment provides incremental clinical
benefit only when Lp(a) is at least mildly elevated (≥13.7 mg/dL) (Schwartz et al.
2021). This observation suggests that Lp(a) reduction translates into a clinical ben-
efit, or at least allows to identify patients who may benefit from PCSK9 inhibition.
It must be acknowledged that the response of Lp(a) to a PCSK9 inhibitor is highly
variable and related to the size of apo(a), since each additional kringle domain is
associated with an additional 3% reduction in Lp(a) (Blanchard et al. 2021).
A large number of studies have shown that the prevalence of elevated Lp(a) in
patients with CVD is higher than in the general population. As an example, the fre-
quency of elevated Lp(a) among patients admitted to the coronary care unit was
27%, and it was even higher (32%) among patients with premature CAD (Ellis et al.
2018). Similarly, patients with relatively early onset CAD had a median Lp(a) of
29 mg/dL; levels ≥30 mg/dL were observed in half of the patients, 36.1% had an
13 Lp(a) as a Cardiovascular Risk Factor 235
Lp(a) 50 ≥ mg/dL, and 16.5% had Lp(a) level ≥100 mg/dL, a level that was shown
to increase CV risk by about threefold (Oo et al. 2020). Likewise, among patients
undergoing percutaneous coronary intervention, Lp(a) was elevated in 48% of indi-
viduals; it is worth remarking that elevated Lp(a) was observed among 45% of
patients with LDL-C ≤70 mg/dL, suggesting a contribution to residual CV risk
(Weiss et al. 2017).
The direct association between Lp(a) levels and the risk of myocardial infarction
has been clearly established in populations of European ancestry (Kamstrup et al.
2008, 2009; Clarke et al. 2009). On the other hand, the heritability of apo(a) and
Lp(a) levels varies across ethnicities, with, as an example, African Americans exhib-
iting the highest Lp(a) level despite having a lower heritability compared with
236 A. Pirillo and A. L. Catapano
% Individuals with
Lp(a) >50 mg/dL
Controls Cases
Controls Cases
2.0-53.6
7.0 13.0 Southeast Asians
2.4-74.2
2.1-61.5
9.0 18.0 South Asians 3.2-88.2
2.0-77.2
14.0 21.0 Latin Americans
2.0-100.0
2.0-89.3
14.0 18.0 Europeans
2.0-99.1
1.9-39.9
3.0 6.0 Chinese 2.3-53.4
2.0-66.8
12.0 15.0 Arabs 2.0-82.6
3.3-102.4
27.0 26.0 Africans 4.1-110.6
0 4 8 12 16 20 24 28
Median Lp(a) (mg/dL)
Fig. 13.3 Median levels (fifth and 95th percentile) of Lp(a) and prevalence of Lp(a) levels >50 mg/
dL in individuals from diverse ancestries (in healthy individuals-CONTROLS- and patients with
myocardial infarction-CASES)
Caucasians (Enkhmaa et al. 2019). Relative to Blacks, South Asians exhibit the
second highest median Lp(a) level, followed by Whites, Hispanics, and East Asians;
however, the causal relationship between Lp(a) and CVD extends to all racial and
ethnic groups (Virani et al. 2022).
An interesting analysis of data from the INTERHEART study, involving 52
countries, showed differences in Lp(a) levels in individuals from diverse ancestries,
with Africans having higher levels compared with other populations (Pare et al.
2019) (Fig. 13.3). Despite the observed differences, high Lp(a) levels were overall
associated with an increased risk of MI (OR 1.48), independently of other estab-
lished MI risk factors (Pare et al. 2019). When analyzed in single ethnic groups,
elevated Lp(a) levels (>50 mg/dL) were associated with increased MI risk in all
populations, except Arabs and Africans; in these two groups, however, the small
sample size might have limited the relevance of the observation compared with
other groups (Pare et al. 2019). In fact, a comparison of the association between
Lp(a) and incident CV events between African Americans and Caucasians in the
ARIC study showed that the hazard ratios for incident CVD and CHD were signifi-
cantly higher in the highest quintile of Lp(a) (>13.5 mg/dL) in both ethnic groups;
it must be acknowledged that Lp(a) levels in the highest quintile ranged from >24 to
81.7 mg/dL (median 32.1 mg/dL) in African Americans and from 13.5 to 80.3 mg/
dL (median 20.4 mg/dL) in Caucasians (Virani et al. 2012). These findings need to
be validated but suggest that, while Lp(a) thresholds designated on the basis of stud-
ies mainly performed in Europeans apply to different ethnic groups, they might
require an adjustment for other ethnic groups having higher mean Lp(a)
concentrations.
13 Lp(a) as a Cardiovascular Risk Factor 237
Conclusions
Although epidemiological and genetic studies have clearly established a causal role
for Lp(a) in CVD, to date the evidence that reducing Lp(a) levels translates into a
clinical benefit is still lacking. While substantial reductions in Lp(a) levels are
required to observe a clinical benefit, new agents that potently lower Lp(a) are under
clinical development. Ongoing trials will tell whether this reduction translates into
reduced CVD events. A phase III study will assess the impact of Lp(a)-lowering
with the antisense oligonucleotide Pelacarsen on major CV events in patients with
CVD and elevated Lp(a) levels (≥70 mg/dL), with a planned follow-up of 4 years
(NCT04023552).
A recent study using samples from the UK Biobank has shown that elevated
Lp(a) levels are associated with an increased risk for incident CAD in individuals
without a family history of heart disease, suggesting that Lp(a) evaluation may be
beneficial in refining CAD risk in primary prevention patients (Finneran et al. 2021).
While waiting for the results of outcome trials and looking for resolutions of major
issues in the measurement of Lp(a) (Virani et al. 2022), Lp(a) assessment should be
regarded as a plus in clinical practice and measured at least once in life (Mach et al.
2020; Reyes-Soffer et al. 2022), with the aim to improve risk stratification and iden-
tify individuals that might be at increased CV risk due to the presence of ele-
vated Lp(a).
References
Vongpromek R, Bos S, Ten Kate GJ, Yahya R, Verhoeven AJ, de Feyter PJ, et al. Lipoprotein(a)
levels are associated with aortic valve calcification in asymptomatic patients with familial
hypercholesterolaemia. J Intern Med. 2015;278(2):166–73.
Weiss MC, Berger JS, Gianos E, Fisher E, Schwartzbard A, Underberg J, et al. Lipoprotein(a)
screening in patients with controlled traditional risk factors undergoing percutaneous coronary
intervention. J Clin Lipidol. 2017;11(5):1177–80.
Willeit P, Ridker PM, Nestel PJ, Simes J, Tonkin AM, Pedersen TR, 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(10155):1311–20.
Yeang C, Willeit P, Tsimikas S. The interconnection between lipoprotein(a), lipoprotein(a) cho-
lesterol and true LDL-cholesterol in the diagnosis of familial hypercholesterolemia. Curr Opin
Lipidol. 2020;31(6):305–12.
Chapter 14
Lp(a) and Aortic Valve Stenosis, Stroke,
and Other Noncoronary Cardiovascular
Diseases
Introduction
High lipoprotein(a) [Lp(a)] levels are now, based on more than three decades of
accumulating evidence from mechanistic, epidemiological, and genetic studies,
widely recognized as an important and likely causal risk factor for ischemic cardio-
vascular and, in particular, coronary artery disease (CAD) (Kamstrup 2021; Reyes-
Soffer et al. 2022). High Lp(a) levels in the top 10% of the concentration distribution
(Fig. 14.1) associate with a two to threefold increase in risk of myocardial infarction
independent of conventional risk factors (Kamstrup 2021). More recently, high
Lp(a) levels, found in an estimated >1 billion individuals globally, have also been
identified as a risk factor for calcific aortic valve stenosis (AVS) with risk estimates
of at least the same magnitude as those found for CAD (Thanassoulis et al. 2013;
Anne Langsted and Pia R. Kamstrup contributed equally with all other contributors.
A. Langsted (*)
Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte,
Herlev, Denmark
Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of
Copenhagen, Copenhagen, Denmark
e-mail: [email protected]
P. R. Kamstrup (*)
Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte,
Herlev, Denmark
The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and
Gentofte, Herlev, Denmark
e-mail: [email protected]
Fig. 14.1 Lp(a) concentration distribution. Plasma levels of Lp(a) (total mass and particle num-
ber) in the Copenhagen General Population Study (N = 79,718 White individuals of Danish
descent, 118 measurements >200 mg/dL not displayed). All measurement values were calibrated
to fresh sample measurements by the latex-enhanced Denka Seiken (Denka Seiken, Tokyo, Japan)
immunoturbidimetric assay with traceability to an international calibrator (WHO SRM 2B).
Conversion to nmol/L was done according to the following equation based on ~13,900 individuals
with measurements in both mg/dL and nmol/L (Denka Seiken Roche distributed assay using dif-
ferent calibrations for mg/dL and nmol/L): lipoprotein(a) nmol/L = 2.18*lipoprotein(a) mg/
dL–3.83. (Adapted from Clin Chem 2021;67:154–166 with permission)
Calcific AVS is a chronic, progressive disease which shares risk factors with athero-
sclerotic disease and is estimated to affect 3% of adults older than 75 years of age
and with a steeply increasing disease burden in high-income countries (Otto and
Prendergast 2014; Yadgir et al. 2020). Up to 50% of patients progress to severe
disease within 2–4 years, and valve replacement represents the only treatment
option for severe disease characterized by obstructive heart failure and increased
risk of sudden death (Coisne et al. 2021). While familial aggregation exists for both
bi- and tricuspid diseases, up until 2013, no specific genetic risk factors had been
identified (Otto and Prendergast 2014). However, in a landmark study from 2013,
and using a hypothesis-free genome-wide association study (GWAS) approach, the
LPA gene was identified as the only genome-wide significant locus for the presence
of aortic valve calcification (AVC) in a White European ancestry cohort (N = 6942)
and with replication also in African American and Hispanic American cohorts
(Thanassoulis et al. 2013). The LPA rs10455872 single nucleotide polymorphism
(SNP), previously found to be strongly associated with Lp(a) levels (Clarke et al.
2009), associated with a twofold increase per allele in risk of AVC considered an
early phenotype for AVS (Thanassoulis et al. 2013). Also, based on Lp(a) levels
available in a subgroup (N = 3670), an odds ratio of 1.62 (1.27–2.06) for AVC was
found per log-unit increase in genetically determined Lp(a) levels. Finally, in two
cohort studies, an association with incident AVS and valve replacement was found
with hazard ratios per allele of 1.7 (95% confidence interval: 1.3–2.2) and 1.5
(1.1–2.3). The combined findings of the study provided strong genetic evidence of
a causal association of Lp(a) with AVC and likely also AVS.
Prior to 2013, associations of high Lp(a) levels with increased risk of AVC or
AVS had only been sporadically reported in smaller epidemiological studies (Gotoh
et al. 1995; Stewart et al. 1997; Glader et al. 2003; Bozbas et al. 2007). However,
the 2013 GWAS study generated considerable interest in high Lp(a) levels as a pos-
sible causal risk factor for calcific AVS. Thus, in 2014, risk estimates for incident
AVS at different levels of Lp(a) were reported from large general population stud-
ies. The first was a combined analysis of the historic Copenhagen City Heart Study
and the contemporary Copenhagen General Population Study (Kamstrup et al.
2014). Risk of AVS was increased for Lp(a) levels in the top third of the concentra-
tion distribution (≥20 mg/dL, ≥40 nmol/L), and individuals with levels ≥90th per-
centile (≥65 mg/dL, ≥138 nmol/L) had a two to threefold increased risk of AVS as
compared to individuals with levels <5 mg/dL (N = 29,016, Fig. 14.2). Notably, the
risk estimates appeared independent of the presence or absence of CAD. On a con-
tinuous scale, a tenfold increase in plasma Lp(a) levels is associated with a hazard
ratio of 1.4 (1.2–1.7) comparable to the 1.6 (1.2–2.1)-fold increase in risk found for
a similar increase in genetically determined levels based on three LPA genotypes
244 A. Langsted and P. R. Kamstrup
1 2 3 4 5 1 2 3 4 5
Hazard ratio (95% CI) for aortic Hazard ratio (95% CI) for aortic
valve stenosis valve stenosis
Fig. 14.2 Risk of aortic valve stenosis by Lp(a) levels. Analyses were adjusted for age, sex, total
cholesterol, HDL (high-density lipoprotein) cholesterol, systolic blood pressure, smoking, and
diabetes. Lipoprotein(a) in mg/dL is shown as median (interquartile range). (Adapted from J Am
Coll Cardiol 2014;63:470–7 with permission)
disease. The totality of evidence pointing to high Lp(a) levels as a potentially modi-
fiable cause of calcific AVS thus holds great promise for improved prevention of
symptomatic AVS upon future availability of effective Lp(a)-lowering drugs.
Stroke
Ischemic Stroke
Lp(a) is a firmly established risk factor for myocardial infarction and AVS by pro-
posed pathophysiological mechanisms such as atherosclerosis and thrombosis. The
association of high Lp(a) levels with risk of ischemic stroke (IS) is not as firmly
established as results from several studies are unclear and somewhat conflicting.
Most studies did find increased risk of IS with high Lp(a) levels such as in the large
prospective Atherosclerosis Risk in Communities (ARIC) study from the USA,
which included both White and Black individuals with a 79% higher risk ratio for
high versus low levels (Ohira et al. 2006). Another large prospective contemporary
study from Denmark, the Copenhagen General Population Study (N = 46,699),
found increased risk of IS per 50 mg/dL higher Lp(a) levels with a hazard ratio of
1.2 (95% CI: 1.1–1.3) in observational analyses and with genetic estimates of 1.2
(1.0–1.4) via KIV2 and 1.3 (1.1–1.5) via rs10455872, indicating a causal role
(Fig. 14.3) (Langsted et al. 2019a). In support of a genetic association, the previ-
ously mentioned large UK Biobank study found an odds ratio of 0.87 (0.79–0.96)
for risk of IS for one standard deviation genetically lower Lp(a) levels (Emdin et al.
2016). Also, meta-analyses of observational studies, for example, from the Emerging
Risk Factors Collaboration including data from 24 studies (Emerging Risk Factors
Collaboration et al. 2009) and from India including data from 41 studies (Kumar
et al. 2021) on IS, find an association of high Lp(a) levels with increased risk of
IS. In the Prospective Epidemiological Study of Myocardial Infarction (PRIME)
study, a large prospective study from Northern Ireland and France, the association
was not significant, but the point estimate indicated an increased risk of IS with high
Lp(a) levels (Canoui-Poitrine et al. 2010). On the contrary, data from the Physicians’
Health Study including White middle-aged males from the USA found no associa-
tion between high Lp(a) levels and risk of IS (Ridker et al. 1995).
Notably, the risk estimates for IS for high Lp(a) levels or corresponding genetic
variants are lower than those reported for myocardial infarction and AVS, perhaps
indicating that the pathophysiology for IS might be different.
Hemorrhagic Stroke
In most cases, the pathophysiology of hemorrhagic stroke differs greatly from the
causes of IS. High Lp(a) levels have previously been associated with a low risk of
bleeding (Langsted et al. 2017) perhaps due to the proposed antifibrinolytic
246 A. Langsted and P. R. Kamstrup
effects of Lp(a) because of its homology with plasminogen. Most studies examin-
ing the role of Lp(a) in stroke have focused on ischemic or overall stroke as
described above, and results on hemorrhagic stroke are even more mixed with
both protective and pathological effects of high Lp(a) levels reported. In a study
from Japan, it was found that high Lp(a) levels were associated with low risk of
hemorrhagic stroke, most significantly in men with a hazard ratio of 0.44
(0.21–0.96) for highest versus lowest tertile of Lp(a) levels (Ishikawa et al. 2013).
The meta-analysis from the Emerging Risk Factors Collaboration et al. (2009)
including nine studies on hemorrhagic stroke found no association of Lp(a) with
hemorrhagic stroke, in contrast to findings from two Chinese studies (Sun et al.
2003; Fu et al. 2020) which found high Lp(a) levels to be associated with increased
risk of hemorrhagic stroke.
The complex nature and fundamentally different causes of hemorrhagic stroke
compared to ischemic stroke might be one of the reasons for these highly conflicting
results. Studies including subtypes of hemorrhagic stroke based on the underlying
pathophysiology are needed to find meaningful associations.
levels and risk of arterial IS in the young. A meta-analysis including 4 studies and a
total of 90 events found that children with high Lp(a) levels have an odds ratio of 4.2
(2.9–6.1) for risk of arterial IS (Sultan et al. 2014). Another meta-analysis including
five studies (with two studies also included in the former meta-analysis) found an
odds ratio for high Lp(a) levels of 6.3 (4.5–8.7) in children with arterial IS (Kenet
et al. 2010).
Of note, as arterial IS is much less prevalent in children than in adults and is often
associated with underlying medical conditions, most of the studies on Lp(a) have
excluded children with other risk factors which may, therefore, limit the generaliz-
ability of the study findings; however, the risk estimates are substantial and should
be investigated further.
Heart Failure
The two to threefold increased risk of myocardial infarction and calcific AVS found
in individuals with Lp(a) levels in the top decile is consistent with Lp(a) also being
a possible risk factor for heart failure (HF), representing a major and increasing
health-economic burden in aging populations (Kamstrup 2021; Heidenreich et al.
2013). In 2015, a clear stepwise association of Lp(a) levels with risk of incident HF
was reported from the combined Copenhagen general population studies including
>98,000 adult individuals of Danish descent (Kamstrup and Nordestgaard 2016).
Lp(a) levels >90th percentile (>67 mg/dL) are associated with a 1.6–1.8-fold
increased risk as compared to individuals with levels in the lower third of the con-
centration distribution and with comparable genetic risk estimates in support of a
causal association. Notably, the association appeared largely driven by the likely
causal associations of Lp(a) with myocardial infarction and/or AVS, with 63% of
the Lp(a)-driven HF risk mediated by these two conditions (Kamstrup and
Nordestgaard 2016). Further, a 9% population attributable risk of HF was estimated
for high Lp(a) levels indicating that, given the development of future effective
Lp(a)-lowering treatments, a notable decrease in HF incidence may also be achieved.
The observational association of Lp(a) levels with HF was since replicated in
both the Atherosclerosis Risk in Communities (ARIC) study and in the Multi-
Ethnic Study of Atherosclerosis (MESA) (Agarwala et al. 2017; Steffen et al.
2018). Findings from the large 2019 Icelandic case-control study with informa-
tion on measured and genetically imputed Lp(a) plasma levels also provided
additional evidence of a causal association of Lp(a) with HF (Gudbjartsson et al.
2019). Thus, a 50 nM (~25 mg/dL) increase in genetically determined Lp(a) lev-
els is associated with a 5% increase in risk of HF. This is in addition to the more
pronounced risk increases reported for CAD, AVS, and peripheral arterial dis-
ease (PAD).
248 A. Langsted and P. R. Kamstrup
Summary
Lp(a) has since its discovery been a lipoprotein particle of high interest in cardiovascu-
lar research due to a composition consistent with both proatherosclerotic and prothrom-
botic effects. It is now well established that high levels are associated both observationally
and genetically, and therefore likely causally, with increased risk of CAD, calcific AVS,
HF, IS, PAD, and mortality (Langsted et al. 2019b). The exact pathophysiology of high
Lp(a) levels has not yet been elucidated and may involve, in addition to proatheroscle-
rotic and prothrombotic effects, also proinflammatory and procalcific effects, and the
exact mechanisms behind different CVD manifestations may differ.
Guidelines today are transitioning from recommending measurement of Lp(a)
only in individuals at increased risk of cardiovascular disease to once-in-a-lifetime
measurement in all. Currently, promising lipoprotein(a)-lowering agents are being
tested, and studies will hopefully show that lowering of lipoprotein(a) will lower the
risk of CVD.
References
Agarwala A, Pokharel Y, Saeed A, Sun W, Virani SS, Nambi V, et al. The association of
lipoprotein(a) with incident heart failure hospitalization: atherosclerosis risk in communities
study. Atherosclerosis. 2017;262:131–7.
Arsenault BJ, Boekholdt SM, Dube MP, Rheaume E, Wareham NJ, Khaw KT, et al. 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(3):304–10.
14 Lp(a) and Aortic Valve Stenosis, Stroke, and Other Noncoronary Cardiovascular… 249
Bozbas H, Yildirir A, Atar I, Pirat B, Eroglu S, Aydinalp A, et al. Effects of serum levels of novel
atherosclerotic risk factors on aortic valve calcification. J Heart Valve Dis. 2007;16(4):387–93.
Canoui-Poitrine F, Luc G, Bard JM, Ferrieres J, Yarnell J, Arveiler D, et al. Relative contribution of
lipids and apolipoproteins to incident coronary heart disease and ischemic stroke: the PRIME
study. Cerebrovasc Dis. 2010;30(3):252–9.
Capoulade R, Yeang C, Chan KL, Pibarot P, Tsimikas S. Association of mild to moderate aortic
valve stenosis progression with higher lipoprotein(a) and oxidized phospholipid levels: second-
ary analysis of a randomized clinical trial. JAMA Cardiol. 2018;3(12):1212–7.
Clarke R, Peden JF, Hopewell JC, Kyriakou T, Goel A, Heath SC, et al. Genetic variants associ-
ated with Lp(a) lipoprotein level and coronary disease. N Engl J Med. 2009;361(26):2518–28.
Coisne A, Montaigne D, Aghezzaf S, Ridon H, Mouton S, Richardson M, et al. Association of
mortality with aortic stenosis severity in outpatients: results from the VALVENOR study.
JAMA Cardiol. 2021;6(12):1424–31.
Despres AA, Perrot N, Poulin A, Tastet L, Shen M, Chen HY, et al. Lipoprotein(a), oxidized phos-
pholipids, and aortic valve microcalcification assessed by 18F-sodium fluoride positron emis-
sion tomography and computed tomography. CJC Open. 2019;1(3):131–40.
Emdin CA, Khera AV, Natarajan P, Klarin D, Won HH, Peloso GM, et al. Phenotypic characterization
of genetically lowered human lipoprotein(a) levels. J Am Coll Cardiol. 2016;68(25):2761–72.
Emerging Risk Factors Collaboration, Erqou S, Kaptoge S, Perry PL, Di Angelantonio E,
Thompson A, et al. Lipoprotein(a) concentration and the risk of coronary heart disease, stroke,
and nonvascular mortality. JAMA. 2009;302(4):412–23.
Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children
and Adolescents, National Heart, Lung, and Blood Institute. Expert panel on integrated guide-
lines for cardiovascular health and risk reduction in children and adolescents: summary report.
Pediatrics. 2011;128(Suppl 5):S213–56.
Fu H, Zhang D, Zhu R, Cui L, Qiu L, Lin S, et al. Association between lipoprotein(a) concentration
and the risk of stroke in the Chinese Han population: a retrospective case-control study. Ann
Transl Med. 2020;8(5):212.
Glader CA, Birgander LS, Soderberg S, Ildgruben HP, Saikku P, Waldenstrom A, et al.
Lipoprotein(a), Chlamydia pneumoniae, leptin and tissue plasminogen activator as risk mark-
ers for valvular aortic stenosis. Eur Heart J. 2003;24(2):198–208.
Gotoh T, Kuroda T, Yamasawa M, Nishinaga M, Mitsuhashi T, Seino Y, et al. Correlation between
lipoprotein(a) and aortic valve sclerosis assessed by echocardiography (the JMS Cardiac Echo
and Cohort Study). Am J Cardiol. 1995;76(12):928–32.
Gudbjartsson DF, Thorgeirsson G, Sulem P, Helgadottir A, Gylfason A, Saemundsdottir J, et al.
Lipoprotein(a) concentration and risks of cardiovascular disease and diabetes. J Am Coll
Cardiol. 2019;74(24):2982–94.
Heidenreich PA, Albert NM, Allen LA, Bluemke DA, Butler J, Fonarow GC, et al. Forecasting
the impact of heart failure in the United States: a policy statement from the American Heart
Association. Circ Heart Fail. 2013;6(3):606–19.
Ishikawa S, Kotani K, Kario K, Kayaba K, Gotoh T, Nakamura Y, et al. Inverse association
between serum lipoprotein(a) and cerebral hemorrhage in the Japanese population. Thromb
Res. 2013;131(2):e54–8.
Kamstrup PR. Lipoprotein(a) and cardiovascular disease. Clin Chem. 2021;67(1):154–66.
Kamstrup PR, Nordestgaard BG. Elevated lipoprotein(a) levels, LPA risk genotypes, and increased
risk of heart failure in the general population. JACC Heart Fail. 2016;4(1):78–87.
Kamstrup PR, Tybjaerg-Hansen A, Nordestgaard BG. Genetic evidence that lipoprotein(a) associ-
ates with atherosclerotic stenosis rather than venous thrombosis. Arterioscler Thromb Vasc
Biol. 2012;32(7):1732–41.
Kamstrup PR, Tybjaerg-Hansen A, Nordestgaard BG. Elevated lipoprotein(a) and risk of aortic
valve stenosis in the general population. J Am Coll Cardiol. 2014;63(5):470–7.
Kenet G, Lutkhoff LK, Albisetti M, Bernard T, Bonduel M, Brandao L, et al. Impact of throm-
bophilia on risk of arterial ischemic stroke or cerebral sinovenous thrombosis in neonates
and children: a systematic review and meta-analysis of observational studies. Circulation.
2010;121(16):1838–47.
250 A. Langsted and P. R. Kamstrup
Kumar P, Swarnkar P, Misra S, Nath M. Lipoprotein(a) level as a risk factor for stroke and its sub-
type: a systematic review and meta-analysis. Sci Rep. 2021;11(1):15660.
Langsted A, Kamstrup PR, Nordestgaard BG. High lipoprotein(a) and low risk of major bleeding
in brain and airways in the general population: a Mendelian randomization study. Clin Chem.
2017;63(11):1714–23.
Langsted A, Nordestgaard BG, Kamstrup PR. Elevated lipoprotein(a) and risk of ischemic stroke.
J Am Coll Cardiol. 2019a;74(1):54–66.
Langsted A, Kamstrup PR, Nordestgaard BG. High lipoprotein(a) and high risk of mortality. Eur
Heart J. 2019b;40(33):2760–70.
Laschkolnig A, Kollerits B, Lamina C, Meisinger C, Rantner B, Stadler M, et al. Lipoprotein(a)
concentrations, apolipoprotein(a) phenotypes, and peripheral arterial disease in three indepen-
dent cohorts. Cardiovasc Res. 2014;103(1):28–36.
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, et al. 2019 ESC/EAS
Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular
risk. Eur Heart J. 2020;41(1):111–88.
Ohira T, Schreiner PJ, Morrisett JD, Chambless LE, Rosamond WD, Folsom AR. Lipoprotein(a)
and incident ischemic stroke: the atherosclerosis risk in communities (ARIC) study. Stroke.
2006;37(6):1407–12.
Otto CM, Prendergast B. Aortic-valve stenosis—from patients at risk to severe valve obstruction.
N Engl J Med. 2014;371(8):744–56.
Pearson GJ, Thanassoulis G, Anderson TJ, Barry AR, Couture P, Dayan N, et al. 2021 Canadian
Cardiovascular Society Guidelines for the management of dyslipidemia for the prevention of
cardiovascular disease in adults. Can J Cardiol. 2021;37(8):1129–50.
Reyes-Soffer G, Ginsberg HN, Berglund L, Duell PB, Heffron SP, Kamstrup PR, et al.
Lipoprotein(a): a genetically determined, causal, and prevalent risk factor for atherosclerotic
cardiovascular disease: a scientific statement from the American Heart Association. Arterioscler
Thromb Vasc Biol. 2022;42(1):e48–60.
Ridker PM, Stampfer MJ, Hennekens CH. Plasma concentration of lipoprotein(a) and the risk of
future stroke. JAMA. 1995;273(16):1269–73.
Schwartz GG, Steg PG, Szarek M, Bittner VA, Diaz R, Goodman SG, et al. Peripheral artery dis-
ease and venous thromboembolic events after acute coronary syndrome: role of lipoprotein(a)
and modification by alirocumab: prespecified analysis of the ODYSSEY OUTCOMES ran-
domized clinical trial. Circulation. 2020;141(20):1608–17.
Steffen BT, Duprez D, Bertoni AG, Guan W, Tsai MY. Lp(a) [Lipoprotein(a)]-related risk of heart
failure is evident in whites but not in other racial/ethnic groups. Arterioscler Thromb Vasc Biol.
2018;38(10):2498–504.
Stewart BF, Siscovick D, Lind BK, Gardin JM, Gottdiener JS, Smith VE, et al. Clinical factors
associated with calcific aortic valve disease. Cardiovascular Health Study. J Am Coll Cardiol.
1997;29(3):630–4.
Sultan SM, Schupf N, Dowling MM, Deveber GA, Kirton A, Elkind MS. Review of lipid and
lipoprotein(a) abnormalities in childhood arterial ischemic stroke. Int J Stroke. 2014;9(1):79–87.
Sun L, Li Z, Zhang H, Ma A, Liao Y, Wang D, et al. Pentanucleotide TTTTA repeat polymorphism
of apolipoprotein(a) gene and plasma lipoprotein(a) are associated with ischemic and hemor-
rhagic stroke in Chinese: a multicenter case-control study in China. Stroke. 2003;34(7):1617–22.
Thanassoulis G, Campbell CY, Owens DS, Smith JG, Smith AV, Peloso GM, et al. Genetic asso-
ciations with valvular calcification and aortic stenosis. N Engl J Med. 2013;368(6):503–12.
Yadgir S, Johnson CO, Aboyans V, Adebayo OM, Adedoyin RA, Afarideh M, et al. Global,
regional, and National Burden of calcific aortic valve and degenerative mitral valve diseases,
1990–2017. Circulation. 2020;141(21):1670–80.
Zheng KH, Tsimikas S, Pawade T, Kroon J, Jenkins WSA, Doris MK, et al. Lipoprotein(a) and
oxidized phospholipids promote valve calcification in patients with aortic stenosis. J Am Coll
Cardiol. 2019;73(17):2150–62.
Chapter 15
Lipoprotein(a) in Cardiovascular Disease:
Evidence from Large Epidemiological
Studies
Introduction
Fig. 15.1 Overview of scientific publications on lipoprotein(a) from the bibliographic database
Scopus since its discovery in 1963 and up until 2021. Lp(a) lipoprotein(a). (Illustration by Børge
G. Nordestgaard)
extreme lipoprotein(a) levels, that is, ≥120 mg/dL (the 95th percentile) (Kamstrup
et al. 2008). The study also demonstrated that the risk of myocardial infarction
increased in a stepwise manner with increasing levels of lipoprotein(a). Further,
when combining data from two large prospective cohorts of the adult Danish popula-
tion, the Copenhagen City Heart Study and the Copenhagen General Population
Study, a stepwise increase in the risk of incident heart failure was observed with
increasing levels of lipoprotein(a) (Kamstrup and Nordestgaard 2016). For use spe-
cifically in this book, we have updated our former epidemiological studies based on
the Copenhagen General Population Study on the association between elevated
lipoprotein(a) and risk of myocardial infarction and heart failure in Fig. 15.2
(Kamstrup et al. 2008, 2009; Kamstrup and Nordestgaard 2016; Langsted et al.
2015). As can be observed, individuals with lipoprotein(a) in the 23rd–65th percen-
tile (5–17 mg/dL or 7–33 nmol/L) have an age- and sex-adjusted hazard ratio of 1.19
[95% confidence interval (CI): 1.06–1.32] for myocardial infarction when compared
with individuals with lipoprotein(a) in the 1st–22nd percentile (<5 mg/dL or
<7 nmol/L). Thus, lipoprotein(a) is a risk factor at a comparatively low level with
regard to myocardial infarction. However, the highest risk of myocardial infarction
Fig. 15.2 Age- and sex-adjusted Cox proportional hazard ratios for the lipoprotein(a)-associated
risk of myocardial infarction and heart failure. Based on ⁓69,000 individuals from the Copenhagen
General Population Study. CI confidence interval. (Data by Børge G. Nordestgaard, Signe Vedel-
Krogh, and Peter E. Thomas)
254 P. E. Thomas et al.
date (Patel et al. 2021). As seen in Fig. 15.4, data from the UK Biobank illustrate the
markedly heterogeneous distribution of lipoprotein(a) levels in individuals of White,
South Asian, Black, and Chinese descent and a clear log-linear association between
higher lipoprotein(a) levels and increased risk of atherosclerotic cardiovascular dis-
ease in the multiethnic population. In the overall study population, a hazard ratio of
1.11 (95% CI: 1.10–1.12) per 50 nmol/L higher lipoprotein(a) levels was observed.
Crucially, despite the clear difference in the distribution of lipoprotein(a) levels
between ethnic groups, the estimated hazard ratio for atherosclerotic cardiovascular
disease per 50 nmol/L higher lipoprotein(a) was similar for Whites [hazard ratio 1.11
(1.10–1.12)], South Asian [hazard ratio 1.10 (1.04–1.16)], and Black individuals
[hazard ratio 1.07 (1.00–1.15)]; there were too few Chinese for meaningful risk esti-
mates in this group. Thus, the risk conferred by a given higher lipoprotein(a) level
was broadly similar across racial groups. This is especially important for Black indi-
viduals where the median lipoprotein(a) concentration is four times higher than the
median concentration in White individuals. These findings are consistent with previ-
ous cohort studies of multiple ethnicities such as the ARIC (Atherosclerosis Risk in
Communities) study, the MESA (Multi-Ethnic Study of Atherosclerosis) study, and
the INTERHEART study (Virani et al. 2012; Paré et al. 2019; Guan et al. 2015).
Data from the Copenhagen cohorts and the UK Biobank clearly illustrate
lipoprotein(a) as a risk factor for atherosclerotic cardiovascular disease including
myocardial infarction and heart failure in primary prevention studies; however,
many other studies have provided crucial contributions to the understanding of
lipoprotein(a) in cardiovascular disease, as reviewed previously (Nordestgaard and
Langsted 2016; Kamstrup 2021). Importantly, generally observational cohort stud-
ies cannot be used to establish causality, as they may be prone to confounding and
reverse causation. However, for lipoprotein(a) levels that are 80–90% genetically
Fig. 15.4 Lipoprotein(a) concentrations according to ethnicity (left panel) and multivariable-
adjusted hazard ratio for atherosclerotic cardiovascular disease according to lipoprotein(a) concen-
tration using cubic natural splines (right panel). Based on 460,506 individuals from the UK
Biobank. Left panel: Median lipoprotein(a) values, dimensions of the box capture the 25th and
75th percentiles, and whiskers capture an additional one interquartile range. (Adapted with permis-
sion from Patel et al. Arterioscler Thromb Vasc Biol. 2021;41:465–474)
256 P. E. Thomas et al.
Table 15.1 Sources of evidence that elevated lipoprotein(a) causes morbidity and mortality in
adults in a primary prevention setting
Case- Large
control or Meta-analyses Large Large genome- Randomized
cross- of prospective prospective Mendelian wide double-blind
sectional observational observational randomization association lipoprotein(a)
studies studies studies studies studies lowering trials
Angina Yes Not examined Yes Yes Yes Not examined
pectoris
Myocardial Yes Yes Yes Yes Yes Trial ongoing
infarction
Heart failure Yes Not examined Yes Yes Yes Not examined
Cardio Not Not examined Yes Yes Yes Trial ongoing
vascular examined
mortality
Table by Børge G. Nordestgaard
Fig. 15.5 Age- and sex-adjusted comparative predictive value of on-statin versus on-placebo
lipoprotein(a) concentrations for incident cardiovascular disease. (Adapted with permission from
Willeit et al. Lancet 2018;392:1311–20)
Fig. 15.6 Cumulative incidence of recurrent major adverse cardiovascular events (MACE)
according to concentrations of lipoprotein(a). CI confidence interval. (Adapted with permission
from Madsen et al. Arterioscler Thromb Vasc Biol. 2020;40:255–266)
levels lower than 50 mg/dL. The data from the seven trials included shows that
higher lipoprotein(a) is positively associated with increased risk of cardiovascular
events in a linear relationship independent of other cardiovascular risk factors in
both patients on statin treatment and on placebo. The association of on-statin higher
lipoprotein(a) with cardiovascular disease risk was stronger than for on-placebo
higher lipoprotein(a), indicating that when low-density lipoprotein (LDL) choles-
terol is reduced by statins, the risk conferred by elevated lipoprotein(a) becomes
more important. Thus, individuals with previous cardiovascular disease and ele-
vated lipoprotein(a) are at substantial residual risk of cardiovascular disease even
while on statin treatment. Nevertheless, randomized trials can be affected by index
event bias, that is, bias that may occur when the occurrence of a particular event is
required for inclusion in a study (Dahabreh and Kent 2011); however, a study of
2527 individuals from the general Danish population with a history of cardiovascu-
lar disease supports the findings from the statin-trial meta-analysis (Madsen et al.
2020). Figure 15.6 shows how higher levels of lipoprotein(a) are associated with
higher risk of recurrent major adverse cardiovascular events. Compared with indi-
viduals with lipoprotein(a) <10 mg/dL (18 nmol/L), the multivariable-adjusted sub-
hazard ratios for major adverse cardiovascular events were 1.29 (95% confidence
258 P. E. Thomas et al.
interval: 1.04–1.59) for individuals with 10–49 mg/dL (18–104 nmol/L), 1.46
(1.14–1.89) for individuals with 50–99 mg/dL (105–213 nmol/L), and 2.17
(1.59–2.98) for individuals with ≥100 mg/dL (≥214 nmol/L). These findings were
independently confirmed in two other cohorts of the Danish general population.
In conclusion, evidence from both post-hoc analyses of clinical statin trials and
from general population studies of individuals with a history of cardiovascular dis-
ease suggest that elevated lipoprotein(a) is associated with an increased risk of
recurrent cardiovascular disease.
Future Perspectives
References
Berg K. A new serum type system in man—the LP system. Acta Pathol Microbiol Scand.
1963;59:369–82.
Boffa MB, Stranges S, Klar N, Moriarty PM, Watts GF, Koschinsky ML. Lipoprotein(a)
and secondary prevention of atherothrombotic events: a critical appraisal. J Clin Lipidol.
2018;12(6):1358–66.
Clarke R, Peden JF, Hopewell JC, Kyriakou T, Goel A, Heath SC, et al. Genetic variants associ-
ated with Lp(a) lipoprotein level and coronary disease. N Engl J Med. 2009;361(26):2518–28.
Dahabreh IJ, Kent DM. Index event bias as an explanation for the paradoxes of recurrence risk
research. JAMA. 2011;305(8):822–3.
Danesh J, Collins R, Peto R. Lipoprotein(a) and coronary heart disease. Meta-analysis of prospec-
tive studies. Circulation. 2000;102(10):1082–5.
Erqou S, Kaptoge S, Perry PL, Di Angelantonio E, Thompson A, White IR, et al. Lipoprotein(a)
concentration and the risk of coronary heart disease, stroke, and nonvascular mortality.
JAMA. 2009;302(4):412–23.
Guan W, Cao J, Steffen BT, Post WS, Stein JH, Tattersall MC, et al. Race is a key variable in
assigning lipoprotein(a) cutoff values for coronary heart disease risk assessment: the multi-
ethnic study of atherosclerosis. Arterioscler Thromb Vasc Biol. 2015;35(4):996–1001.
Kamstrup PR. Lipoprotein(a) and cardiovascular disease. Clin Chem. 2021;67(1):154–66.
Kamstrup PR, Nordestgaard BG. Elevated lipoprotein(a) levels, LPA risk genotypes, and increased
risk of heart failure in the general population. JACC Heart Fail. 2016;4(1):78–87.
15 Lipoprotein(a) in Cardiovascular Disease: Evidence from Large Epidemiological… 259
Kamstrup PR, Benn M, Tybjaerg-Hansen A, Nordestgaard BG. Extreme lipoprotein(a) levels and
risk of myocardial infarction in the general population: the Copenhagen City Heart Study.
Circulation. 2008;117(2):176–84.
Kamstrup PR, Tybjaerg-Hansen A, Steffensen R, Nordestgaard BG. Genetically elevated
lipoprotein(a) and increased risk of myocardial infarction. JAMA. 2009;301(22):2331–9.
Koren MJ, Moriarty PM, Baum SJ, Neutel J, Hernandez-Illas M, Weintraub HS, et al. Preclinical
development and phase 1 trial of a novel siRNA targeting lipoprotein(a). Nat Med.
2022;28(1):96–103.
Langsted A, Varbo A, Kamstrup PR, Nordestgaard BG. Elevated lipoprotein(a) does not cause low-
grade inflammation despite causal association with aortic valve stenosis and myocardial infarc-
tion: a study of 100,578 individuals from the general population. J Clin Endocrinol Metab.
2015;100(7):2690–9.
Madsen CM, Kamstrup PR, Langsted A, Varbo A, Nordestgaard BG. Lipoprotein(a)-lowering by
50 mg/dL (105 nmol/L) may be needed to reduce cardiovascular disease 20% in secondary
prevention: a population-based study. Arterioscler Thromb Vasc Biol. 2020;40(1):255–66.
McLean JW, Tomlinson JE, Kuang WJ, Eaton DL, Chen EY, Fless GM, et al. cDNA sequence of
human apolipoprotein(a) is homologous to plasminogen. Nature. 1987;330(6144):132–7.
Nissen SE, Wolski K, Balog C, Swerdlow DI, Scrimgeour AC, Rambaran C, et al. Single ascend-
ing dose study of a short interfering RNA targeting lipoprotein(a) production in individuals
with elevated plasma lipoprotein(a) levels. JAMA. 2022;327(17):1679–87.
Nordestgaard BG, Langsted A. Lipoprotein(a) as a cause of cardiovascular disease: insights from
epidemiology, genetics, and biology. J Lipid Res. 2016;57(11):1953–75.
Paré G, Çaku A, McQueen M, Anand SS, Enas E, Clarke R, et al. Lipoprotein(a) levels and the risk
of myocardial infarction among 7 ethnic groups. Circulation. 2019;139(12):1472–82.
Patel AP, Wang M, Pirruccello JP, Ellinor PT, Ng K, Kathiresan S, et al. Lp(a) (Lipoprotein[a])
concentrations and incident atherosclerotic cardiovascular disease: New Insights from a large
National Biobank. Arterioscler Thromb Vasc Biol. 2021;41(1):465–74.
Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, Tardif JC, Baum SJ, Steinhagen-
Thiessen E, et al. Lipoprotein(a) reduction in persons with cardiovascular disease. N Engl J
Med. 2020;382(3):244–55.
Virani SS, Brautbar A, Davis BC, Nambi V, Hoogeveen RC, Sharrett AR, et al. Associations
between lipoprotein(a) levels and cardiovascular outcomes in black and white subjects: the
atherosclerosis risk in communities (ARIC) study. Circulation. 2012;125(2):241–9.
Willeit P, Ridker PM, Nestel PJ, Simes J, Tonkin AM, Pedersen TR, 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(10155):1311–20.
Chapter 16
Lipoprotein(a) and Immunity
Introduction
Monocytes and macrophages play a critical role in innate immunity (Libby et al.
2013) and have been the subject of numerous studies in connection with Lp(a).
Lp(a) was detected in macrophage cell-rich areas of atherosclerotic plaques in
humans according to morphology and immunohistochemistry studies (Sotiriou et al.
2006). On the other hand, individuals with elevated Lp(a) level exhibit enhanced
accumulation of peripheral blood mononuclear cells in the arterial wall compared to
individuals with normal levels of Lp(a) (van der Valk et al. 2016). Apo(a) stimulates
the production of reactive oxygen species and matrix metalloproteinase-9 by colla-
gen-adherent monocytes, and this effect was inversely associated with the molecular
weight of apo(a) (Sabbah et al. 2019). Apo(a) also caused increased secretion of
IL-8 by macrophages of the THP-1 and U-937 cell lines (Scipione et al. 2015).
Monocytes isolated from subjects with elevated Lp(a) demonstrated an enhanced
cell surface expression of chemokine receptors, adhesion molecules, and scavenger
receptors (CCR7, CD62L, CD11b, CD11c, CD29, CD36, SR-A). Apo(a) upregu-
lates the expression of the β2-integrin Mac-1 (CD11b/CD18), thereby facilitating
cell adhesion and migration capacity. Several signaling cascades leading to altered
gene expression profiles were found to contribute to Lp(a)-induced monocyte che-
motactic activity (Scipione et al. 2015; Dzobo et al. 2022).
Besides displaying an activated and proinflammatory phenotype, monocytes iso-
lated from individuals with elevated Lp(a) exhibited an increased secretion of pro-
inflammatory cytokines (IL-1β, IL-6, TNFα) and a decrease in the anti-inflammatory
cytokine IL-10 after stimulation via toll-like receptors. OxPLs associated with
apo(a) as potent danger-associated molecular patterns (DAMPs) could be respon-
sible for these effects (Koschinsky and Boffa 2022).
Apo(a) antisense treatment resulted in downregulation of proinflammatory gene
expression in monocytes, including interferon (IFN)α, IFNγ, and toll-like receptor
(TLR) pathways, and subsequent changes in monocyte phenotype and function, that
is, a reduction in chemokine receptors CCR2 and CX3CR1 and transendothelial
migratory capacity (Stiekema et al. 2020).
264 O. I. Afanasieva et al.
The number of circulating monocytes in apo(a) transgenic mice was four times
higher than in wild-type mice and remained elevated for 3 weeks after Ca2+-induced
vascular damage (Huang et al. 2014). Also, Lp(a) affects the maturation of mono-
cytes in humans (Schnitzler et al. 2020).
Monocytes are divided into three subpopulations, depending on the content of
CD14 and CD16 surface markers, classical CD14++CD16−, intermediate
CD14++CD16+, and nonclassical CD14+CD16++, while the latter two populations
have the most pronounced proinflammatory and profibrotic potential. The participa-
tion of circulating monocytes in atherogenesis has been proven (Vergallo and Crea
2020), but the contribution of various subpopulations of monocytes to chronic
inflammatory states is currently under discussion (Yang et al. 2014; Ożańska
et al. 2020).
The high content of CD16+ monocytes is associated with unstable atheroscle-
rotic plaques in the coronary arteries (Kashiwagi et al. 2010) and predicts the risk of
cardiovascular events (Rogacev et al. 2012). In CHD patients, an increased content
of intermediate monocytes CD14++CD16+ occurs with hyperlipoproteinemia(a)
(Krychtiuk et al. 2015a), atherogenic dyslipidemia (Krychtiuk et al. 2015b), and
dysfunctional high-density lipoproteins (Krychtiuk et al. 2014). The association of
elevated Lp(a) concentration with absolute and relative content of CD14+CD16++
was shown in a retrospective study (Afanasieva et al. 2021). Since the function of
this subpopulation is to remove “cellular debris,” it is assumed that it contributes to
elimination of excess Lp(a).
Neutrophil granulocytes are the largest population of circulating phagocytizing
leukocytes capable of synthesizing a wide range of substances. Neutrophils and
“neutrophil extracellular traps” (NETs) formed by them were found in atheroscle-
rotic plaques of laboratory animals and humans (Afanasieva et al. 2021). NETs
stimulate the production of IL-1 by macrophages and activate IL-17-producing
T-helpers (Th17) in apoE-deficient mice, contributing to inflammation in the vessel
wall (Döring et al. 2017). There are no data on the effect of Lp(a) or apo(a) on the
formation of neutrophil traps. The absolute number of neutrophils and the neutrophil-
lymphocyte index, as well as the concentration of Lp(a), was significantly higher in
patients with stenosing atherosclerosis of various vascular beds (Tmoyan et al.
2021). The evaluation of the effect of Lp(a) on neutrophil activation is a promising
avenue of further research.
A higher lymphocyte count is associated with a higher apoB level; Lp(a) was
inversely associated with basophil count in men but not in women according to a
population study with 417,132 participants (Tucker et al. 2021). Low molecular
weight apo(a) phenotype, reduced lymphocyte count, and increases in neutrophil
granulocytes potentiated the risk of CHD in patients with type 2 diabetes (Suzuki
et al. 2013).
The combination of a higher absolute monocyte count (>0.54 × 109 cells/mL)
with elevated Lp(a) (≥30 mg/dL) is associated with higher risk of major adverse
cardiovascular events (MACE) in patients with premature CHD manifestation
(Afanasieva et al. 2022) (Fig. 16.1). An increase of Lp(a) concentration and the
percentage of CD14++CD16+ monocytes potentiated risk of multivessel coronary
disease (Afanasieva et al. 2021; Filatova et al. 2022) (Fig. 16.2).
A lower level of IgM AAbs against Lp(a) is negatively correlated with the con-
centration of sCD25 [the soluble form of the IL-2 receptor and a surrogate marker
of T-cell activation (Brusko et al. 2009)] and associated with stenosing coronary
atherosclerosis (Afanasievа et al. 2016b). This fact may serve as a confirmation of
participation of both Lp(a) and T-cells in atherogenesis and also the immunomodu-
latory ability of IgM AAbs against Lp(a) (Wang et al. 2016).
Systemic inflammation accompanies age-related changes in lymphocyte sub-
populations (Thomas et al. 2020). In patients with ASCVD, the number of naïve
lymphocytes, including regulatory cells, decreases with age, while the level of
effector populations, that is, Th1 and Th17, remains constant (Filatova et al. 2021).
T-Lymphocytes with predominating Th1 are detected in atherosclerotic plaques
(Saigusa et al. 2020). Th17, a subpopulation of CD4+ lymphocytes producing
IL-17, also has a proatherogenic effect. Th17 cells participate in the immune
response against their own and alien antigens by attracting myeloid cells to a place
of inflammation, activating lymphocytes and secreting proinflammatory cytokines
(Gao et al. 2010; Park et al. 2005). On the contrary, regulatory T-cells have anti-
inflammatory activity and inhibit atherogenesis (Albany et al. 2019). Thus,
Fig. 16.1 The proportion of major adverse cardiovascular events (MACE) in patients with prema-
ture coronary heart disease depending on blood monocyte count and lipoprotein(a) concentration.
Two-hundred adult patients with early coronary heart disease manifestation (before 55 years in
men and 60 years in women) were enrolled, median follow-up 12 years. MACE, nonfatal myocar-
dial infarction, ischemic stroke, coronary artery bypass grafting, and hospitalization for unstable
angina (Afanasieva et al. 2022)
266 O. I. Afanasieva et al.
age-related deficiency of regulatory cells and a shift of the immune balance toward
effector populations may contribute to atherosclerosis progression.
Activation and increased amounts of Th17 are related to the progression of ath-
erosclerosis and risk of coronary events (Liuzzo et al. 2013). The ratio of circulating
Treg/Th17 is reduced in patients with severe coronary atherosclerosis (Potekhina
et al. 2015). The concentration of Lp(a) is not associated with the content of various
T-cell subpopulations (Afanasieva et al. 2016a, b). However, an increased content of
circulating Th17 (% of CD4+ lymphocytes), as well as a reduced content of Treg or
IL-10 CD4+-producing cells along with Lp(a) concentrations above 12 mg/dL, is
associated with severe coronary atherosclerosis (Afanasievа et al. 2016b) and
carotid atherosclerosis progression (Afanasieva et al. 2016a). Thus, the increased
concentration of Lp(a) and proinflammatory status with some shifts in immunity
could potentiate atherosclerosis progression.
Fig. 16.3 Possible mechanisms of lipoprotein(a) contribution to immune cell activation. lysoPC
lysophosphatidylcholines, LPARs LPA receptors or G-protein-coupled receptors, IL-1β interleukin
1β, IL-6 interleukin 6, TNF tumor necrosis factor, CXCL2 chemokine (C–X–C motif) ligand 2,
MCP-1 monocyte chemoattractant protein 1, mRNA messenger RNA, LRP-1 low-density lipopro-
tein receptor-related protein 1, TGFβ transforming growth factor beta, IFNγ interferon γ, PRRs
pattern recognition receptors, oxPL oxidized phospholipids, PLA2 phospholipase A2, α2M
alpha-2-macroglobulin, PCSK9 proprotein convertase subtilisin/kexin type 9, C3 and C4 comple-
ment components 3 and 4, and apo(a) apolipoprotein(a)
LDL, removal by extracorporeal treatment can lead to stabilization and even regres-
sion of atherosclerotic lesions in coronary and carotid arteries (Pokrovsky et al.
2016, 2020). This study was the first direct clinical observation and confirmation of
Lp(a) atherogenicity in humans (Pokrovsky et al. 2017). The elucidation of molecu-
lar and cellular mechanisms of Lp(a) involvement in inflammatory remodeling of
the arterial wall engaging the Lp(a) immunity axis is a promising direction for the
development of new therapeutic approaches.
Lp(a) is an extremely interesting polymolecular complex, and as we learn more
about it, it is clear the less we understand about its enormous functional range and
its capacity to interact with and influence important pathways, such as immunity,
inflammation, thrombosis, and oxidation.
Acknowledgements The authors are grateful to Professor Gilbert Thompson for his help in
proofreading text of the manuscript.
References
Afanas’eva OI, Klesareva EA, Levashov PA, Berestetskaya YV, Ezhov MV, Artem’eva NV,
Pokrovskii SN. Autoantibodies against lipoprotein(a) in patients with coronary heart disease.
Kardiologiya. 2014;54(6):4–8.
Afanasieva OI, Pylaeva EA, Arefieva TI, Klesareva EA, Afanasieva MI, Potekhina AV, Shchinova
AM, Balakhonova TV, Pogorelova OA, Tripoten MI, Pokrovsky SN. Lipoprotein(a) and
T-helper cells as independent predictors of rapid progression of carotid atherosclerosis.
Atherosclerosis. 2016a;252:e126–7.
Afanasieva OI, Pylaeva EA, Klesareva EA, Potekhina AV, Provatorov SI, Afanasieva MI,
Krasnikova TL, Masenko VP, Arefieva TI, Pokrovsky SN. Lipoprotein(a), its autoantibodies,
and circulating T lymphocyte subpopulations as independent risk factors for coronary artery
atherosclerosis. Ter Arkh. 2016b;88(9):31–8.
Afanasieva OI, Filatova AY, Arefieva TI, Klesareva EA, Tyurina AV, Radyukhina NV, Ezhov MV,
Pokrovsky SN. The association of lipoprotein(a) and circulating monocyte subsets with severe
coronary atherosclerosis. J Cardiovasc Dev Dis. 2021;8(6):63.
Afanasieva OI, Tyurina AV, Klesareva EA, Arefieva TI, Ezhov MV, Pokrovsky SN. Lipoprotein(a),
immune cells and cardiovascular outcomes in patients with premature coronary heart disease.
J Pers Med. 2022;12(2):269.
Albany CJ, Trevelin SC, Giganti G, Lombardi G, Scottà C. Getting to the heart of the matter: the
role of regulatory T-cells (Tregs) in cardiovascular disease (CVD) and atherosclerosis. Front
Immunol. 2019;10:2795.
Bouchareb R, Mahmut A, Nsaibia MJ, Boulanger MC, Dahou A, Lépine JL, Laflamme MH, Hadji
F, Couture C, Trahan S, Pagé S, Bossé Y, Pibarot P, Scipione CA, Romagnuolo R, Koschinsky
ML, Arsenault BJ, Marette A, Mathieu P. Autotaxin derived from lipoprotein(a) and valve
interstitial cells promotes inflammation and mineralization of the aortic valve. Circulation.
2015;132(8):677–90.
Bourgeois R, Girard A, Perrot N, Guertin J, Mitchell PL, Couture C, Gotti C, Bourassa S, Poggio
P, Mass E, Capoulade R, Scipione CA, Després AA, Couture P, Droit A, Pibarot P, Boffa
MB, Thériault S, Koschinsky ML, Mathieu P, Arsenault BJ. A comparative analysis of the
lipoprotein(a) and low-density lipoprotein proteomic profiles combining mass spectrometry
and Mendelian randomization. CJC Open. 2020a;3(4):450–9.
270 O. I. Afanasieva et al.
Bourgeois R, Devillers R, Perrot N, Després AA, Boulanger MC, Mitchell PL, Guertin J, Couture
P, Boffa MB, Scipione CA, Pibarot P, Koschinsky ML, Mathieu P, Arsenault BJ. Interaction of
autotaxin with lipoprotein(a) in patients with calcific aortic valve stenosis. JACC Basic Transl
Sci. 2020b;5(9):888–97.
Bourgeois R, Bourgault J, Despres AA, Perrot N, Guertin J, Girard A, Mitchell PL, Gotti C,
Bourassa S, Scipione CA, Gaudreault N, Boffa MB, Koschinsky ML, Pibarot P, Droit
A, Thériault S, Mathieu P, Bossé Y, Arsenault BJ. Lipoprotein proteomics and aortic valve
transcriptomics identify biological pathways linking lipoprotein(a) levels to aortic stenosis.
Metabolites. 2021;11(7):459.
Brusko TM, Wasserfall CH, Hulme MA, Cabrera R, Schatz D, Todd Atkinson MA. Influence of
membrane CD25 stability on T lymphocyte activity: implications for immunoregulation. PLoS
One. 2009;4(11):e7980.
de Vries MR, Ewing MM, de Jong RCM, MacArthur MR, Karper JC, Peters EAB, Nordzell
M, Karabina SAP, Sexton D, Dahlbom I, Bergman A, Mitchell JR, Frostegård J, Kuiper J,
Ninio E, Jukema JW, Pettersson K, Quax PHA. Identification of IgG1 isotype phosphoryl-
choline antibodies for the treatment of inflammatory cardiovascular diseases. J Intern Med.
2021;290(1):141–56.
Döring Y, Soehnlein O, Weber C. Neutrophil extracellular traps in atherosclerosis and athero-
thrombosis. Circ Res. 2017;120(4):736–43.
Dzobo KE, Kraaijenhof JM, Stroes E, Nurmohamed NS, Kroon J. Lipoprotein(a): an underesti-
mated inflammatory mastermind. Atherosclerosis. 2022;349:101–9.
Eming SA, Wynn TA, Martin P. Inflammation and metabolism in tissue repair and regeneration.
Science. 2017;356(6342):1026–30.
Filatova AY, Potekhina AV, Aref'eva TI. Age-associated characteristics of CD4+ T-cell composi-
tion in patients with atherosclerosis. Immuno. 2021;1(3):277–84.
Filatova AY, Potekhina AV, Radyukhina NV, Ruleva NY, Provatorov SI, Aref'eva TI. Circulating
monocyte populations in patients with coronary atherosclerosis. Future Cardiol.
2022;18(6):455–60.
Franceschi C, Garagnani P, Parini P, Giuliani C, Santoro A. Inflammaging: a new immune-
metabolic viewpoint for age-related diseases. Nat Rev Endocrinol. 2018;14(10):576–90.
Gao Q, Jiang Y, Ma T, Zhu F, Gao F, Zhang P, Guo C, Wang Q, Wang X, Ma C, Zhang Y, Chen W,
Zhang L. A critical function of Th17 proinflammatory cells in the development of atheroscle-
rotic plaque in mice. J Immunol. 2010;185(10):5820–7.
Holodick NE, Rodríguez-Zhurbenko N, Hernández AM. Defining natural antibodies. Front
Immunol. 2017;8:872.
Huang M, Gong Y, Grondolsky J, Hoover-Plow J. Lp(a)/apo(a) modulate MMP-9 activation and
neutrophil cytokines in vivo in inflammation to regulate leukocyte recruitment. Am J Pathol.
2014;184(5):1503–17.
Kashiwagi M, Imanishi T, Tsujioka H, Ikejima H, Kuroi A, Ozaki Y, Ishibashi K, Komukai K,
Tanimoto T, Ino Y, Kitabata H, Hirata K, Akasaka T. Association of monocyte subsets with
vulnerability characteristics of coronary plaques as assessed by 64-slice multidetector com-
puted tomography in patients with stable angina pectoris. Atherosclerosis. 2010;212(1):171–6.
Kim YU, Kee P, Danila D, Teng B-B. A critical role of PCSK9 in mediating IL-17-producing
T cell responses in hyperlipidemia. Immune Netw. 2019;19(6):e41.
Klesareva EA, Afanas’eva OI, Donskikh VV, Adamova IY, Pokrovskii SN. Characteristics of
lipoprotein(a)-containing circulating immune complexes as markers of coronary heart disease.
Bull Exp Biol Med. 2016;162(2):231–6.
Klesareva EA, Afanasieva OI, Kononova EV, Utkina EA, Ezhov MV, Balakhonova TV, Afanasieva
MI, Pokrovsky SN. Raised IgM autoantibody titer to lipoprotein(a) as antiatherogenic factor in
severe hypercholesterolemia patients. Russ J Cardiol. 2018;8:13–20.
Klimov AN. Autoimmune theory of atherogenesis and the concept of modified lipoproteins. Vestn
Akad Med Nauk SSSR. 1990;11:30–6.
Knowlden S, Georas SN. The autotaxin-LPA axis emerges as a novel regulator of lymphocyte
homing and inflammation. J Immunol. 2014;192(3):851–7.
16 Lipoprotein(a) and Immunity 271
Pokrovsky SN, Afanasieva OI, Safarova MS, Balakhonova TV, Matchin YG, Adamova IY,
Konovalov GA, Ezhov MV. Specific Lp(a) apheresis: a tool to prove lipoprotein(a) atheroge-
nicity. Atheroscler Suppl. 2017;30:166–73.
Pokrovsky SN, Afanasieva OI, Ezhov MV. Therapeutic apheresis for management of Lp(a) hyper-
lipoproteinemia. Curr Atheroscler Rep. 2020;22(11):68.
Potekhina AV, Pylaeva EA, Provatorov SI, Ruleva N, Masenko V, Noeva E, Krasnikova T, Arefieva
T. Treg/Th17 balance in stable CAD patients with different stages of coronary atherosclerosis.
Atherosclerosis. 2015;238(1):17–21.
Prasad A, Clopton P, Ayers C, Khera A, de Lemos JA, Witztum JL, Tsimikas S. Relationship
of autoantibodies to MDA-LDL and ApoB-immune complexes to sex, ethnicity, subclini-
cal atherosclerosis, and cardiovascular events. Arterioscler Thromb Vasc Biol. 2017;37(6):
1213–21.
Puri R, Nissen SE, Arsenault BJ, St John J, Riesmeyer JS, Ruotolo G, McErlean E, Menon V, Cho
L, Wolski K, Lincoff AM, Nicholls SJ. 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(10):1136–43.
Qi Z, Hu L, Zhang J, Yang W, Liu X, Jia D, Yao Z, Chang L, Pan G, Zhong H, Luo X, Yao K,
Sun A, Qian J, Ding Z, Ge J. PCSK9 (Proprotein Convertase Subtilisin/Kexin 9) enhances
platelet activation, thrombosis, and myocardial infarct expansion by binding to platelet CD36.
Circulation. 2021;143(1):45–61.
Ravandi A, Boekholdt SM, Mallat Z, Talmud PJ, Kastelein JJ, Wareham NJ, Miller ER, Benessiano
J, Tedgui A, Witztum JL, Khaw KT, Tsimikas S. Relationship of IgG and IgM autoantibod-
ies and immune complexes to oxidized LDL with markers of oxidation and inflammation
and cardiovascular events: results from the EPIC-Norfolk Study. J Lipid Res. 2011;52(10):
1829–36.
Reyneveld GI, Savelkoul H, Parmentier HK. Current understanding of natural antibodies and
exploring the possibilities of modulation using veterinary models. A review. Front Immunol.
2020;11:2139.
Rogacev KS, Cremers B, Zawada AM, Seiler S, Binder N, Ege P, Große-Dunker G, Heisel I, Hornof
F, Jeken J, Rebling NM, Ulrich C, Scheller B, Böhm M, Fliser D, Heine GH. CD14++CD16+
monocytes independently predict cardiovascular events: a cohort study of 951 patients referred
for elective coronary angiography. J Am Coll Cardiol. 2012;60(16):1512–20.
Sabarinath PS, Appukuttan PS. Immunopathology of desialylation: human plasma lipoprotein(a)
and circulating anti-carbohydrate antibodies form immune complexes that recognize host cells.
Mol Cell Biochem. 2015;403(1–2):13–23.
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):166.
Saigusa R, Winkels H, Ley K. T cell subsets and functions in atherosclerosis. Nat Rev Cardiol.
2020;17(7):387–401.
Salonen JT, Ylä-Herttuala S, Yamamoto R, Butler S, Korpela H, Salonen R, Nyyssönen K, Palinski
W, Witztum JL. Autoantibody against oxidized LDL and progression of carotid atherosclero-
sis. Lancet. 1992;339(8798):883–7.
Schnitzler JG, Poels K, Stiekema LCA, Yeang C, Tsimikas S, Kroon J, Stroes ESG, Lutgens E,
Seijkens TTP. Short-term regulation of hematopoiesis by lipoprotein(a) results in the produc-
tion of pro-inflammatory monocytes. Int J Cardiol. 2020;315:81–5.
Scipione CA, Sayegh SE, Romagnuolo R, Tsimikas S, Marcovina SM, Boffa MB, Koschinsky
ML. Mechanistic insights into Lp(a)-induced IL-8 expression: a role for oxidized phospholipid
modification of apo(a). J Lipid Res. 2015;56(12):2273–85.
Shapiro MD, Tavori H, Fazio S. PCSK9: from basic science discoveries to clinical trials. Circ Res.
2018;122(10):1420–38.
Silverstein RL. PCSK9 (Proprotein Convertase Subtilisin/Kexin 9) goes “DAMP”. Circulation.
2021;143(1):62–4.
16 Lipoprotein(a) and Immunity 273
Wang H, Coligan JE, Morse HC 3rd. Emerging functions of natural IgM and its Fc receptor FCMR
in immune homeostasis. Front Immunol. 2016;7:99.
Wiesner P, Tafelmeier M, Chittka D, Choi SH, Zhang L, Byun YS, Almazan F, Yang X, Iqbal N,
Chowdhury P, Maisel A, Witztum JL, Handel TM, Tsimikas S, Miller YI. MCP-1 binds to oxi-
dized LDL and is carried by lipoprotein(a) in human plasma. J Lipid Res. 2013;54(7):1877–83.
Yang J, Zhang L, Yu C, Yang XF, Wang H. Monocyte and macrophage differentiation: circulation
inflammatory monocyte as biomarker for inflammatory diseases. Biomark Res. 2014;2(1):1.
Zawadzki Z, Tercé F, Seman LJ, Theolis RT, Breckenridge WC, Milne RW, Marcel YL. The link-
age with apolipoprotein(a) in lipoprotein(a) modifies the immunochemical and functional
properties of apolipoprotein B. Biochemistry. 1988;27(22):8474–81.
Zhang Y, Chen YC, Krummel MF, Rosen SD. Autotaxin through lysophosphatidic acid stimu-
lates polarization, motility, and transendothelial migration of naive T cells. J Immunol.
2012;189(8):3914–24.
Chapter 17
When Should We Measure Lipoprotein(a)?
Karam Kostner
Introduction
K. Kostner (*)
Department of Cardiology, Mater Hospital and University of Queensland, Brisbane, Australia
e-mail: [email protected]
Lp(a) levels are relatively stable over a lifespan as they are mainly genetically deter-
mined which is why a single measurement of serum Lp(a) is sufficient for most
patients unless a secondary cause is suspected or a specific treatment is started to
reduce its concentration. Availability and reimbursement of cost-effective methods
to measure Lp(a) as well as standardisation of assays are important and are dis-
cussed in different chapters of this book. It is generally more practical and cost-
effective to measure Lp(a) concentrations instead of its genetic determinants. Lp(a)
measurement may be considered in both primary and secondary preventions. In
children with familial hypercholesterolemia (FH), for example, Lp(a) is a better
predictor of CV disease in family members than LDL (Zawacki et al. 2018).
In primary prevention, focus should be directed towards absolute CV risk assess-
ment, where patients with elevated Lp(a) are treated more aggressively for traditional
risk factors such as LDL, especially if they are in an intermediate-risk group (Verbeek
et al. 2018). The availability of imaging methods such as calcium scoring by CT and
plaque assessment by CT, MRI and ultrasound has improved CV risk assessment and
is often used in conjunction with lipid risk factors such as Lp(a). In secondary pre-
vention, elevated Lp(a) is a driver of residual CV risk. The Justification for the Use
of Statins in the JUPITER study (Khera et al. 2014) supported the premise that Lp(a)
is a significant independent contributor to residual risk. This is further supported by
data from the Atherothrombosis Intervention in Metabolic syndrome with low HDL/
high triglycerides: Impact on Global Health outcomes (AIM-HIGH) study (Albers
et al. 2013) and a recent meta-analysis (Willeit et al. 2018).
Recent outcome studies with proprotein convertase subtilisin/kexin type 9
(PCSK9) inhibitors have underlined the importance of Lp(a) measurement. In the
Further Cardiovascular Outcomes Research With PCSK9 Inhibition in Subjects
With Elevated Risk (FOURIER) trial, reduction in risk of major acute coronary
events (MACE) with evolocumab was associated with the baseline and change in
Lp(a) levels (O’Donoghue et al. 2019). In the Evaluation of Cardiovascular
Outcomes After an Acute Coronary Syndrome During Treatment With Alirocumab
(ODYSSEY OUTCOMES) trial, reduction in risk of total cardiovascular events
with alirocumab was also associated with baseline and change in Lp(a) levels
(Szarek et al. 2020). Reduction in risk of major adverse limb events (MALE) with
alirocumab was also associated with baseline and change in Lp(a) levels (Schwartz
et al. 2020). These trials support the conclusion that elevated Lp(a) is a major driver
of residual risk. Although large cardiovascular outcome trial data does not currently
exist to guide Lp(a) therapeutic intervention, indicators of significant increased risk,
including multivessel disease, PAD (peripheral artery disease), premature disease
onset, familial hypercholesterolaemia (FH), diabetes, renal disease and recurrent
presentations with acute coronary syndrome (ACS), will likely be considered as
clinical indicators for consideration of agents specifically targeting Lp(a) and
17 When Should We Measure Lipoprotein(a)? 277
already lead many clinicians to try to achieve very low LDL targets with statins,
ezetimibe and PCSK9 inhibitors. Measurement of Lp(a) is discussed as follows and
is summarised in Table 17.1:
1. Measurement of Lp(a) should be considered in adults to assess or stratify
ASCVD risk in those with the following clinical features: a personal history of
premature ASCVD (<60 years), family history of premature ASCVD, family
history of high Lp(a) (>200 nmol/L), familial hypercholesterolaemia, significant
renal impairment and early-onset calcific aortic stenosis (<60 years).
2. Measurement of Lp(a) should be considered in those with an intermediate
10-year ASCVD risk (5–15%) when classical risk algorithms are used such as
the Framingham risk score, the PROCAM risk score, the ESC Heart Score or the
Australian and New Zealand risk calculator, if it allows patients to be re-stratified
into a higher-risk category or if Lp(a) is elevated above >200 nmol/L, which in
turn should ultimately lead to more intensive management of treatable risk fac-
tors, especially low-density lipoprotein cholesterol (LDL-C).
3. Measurement of Lp(a) should be considered in those with suboptimal lowering
of LDL-C despite adherence to guideline-recommended therapy.
4. Measurement of Lp(a) should be considered in those with recurrent or progres-
sive ASCVD despite optimally treated plasma LDL-C concentrations.
5. Measurement of Lp(a) should be considered in children and adolescents with
familial hypercholesterolaemia, premature ASCVD, a first-degree relative with
significantly elevated Lp(a) (>200 nmol/L) or a family history of prema-
ture ASCVD.
278 K. Kostner
Even though most major international guidelines recognise that Lp(a) is a risk enhanc-
ing factor, there is still no unanimous agreement as to when to measure Lp(a) and how
to deal with increased Lp(a) values. The reasons for this are that few commonly
accepted assays and reference standards exist, there is a lack of effective medications
available to lower Lp(a) and apart from LDL apheresis, therapeutic interventions to
lower Lp(a) have not yet shown a reduction in MACE. Traditionally, levels >30 mg/
dL were considered elevated, with thresholds for inclusion into outcome trials gener-
ally higher (>50 mg/dL). Table 17.1 shows risk thresholds for different Lp(a) levels.
The European Atherosclerosis Society and European Cardiology Societies, how-
ever, likely underestimate the importance of elevated Lp(a) as they focus only on
people with extremely elevated levels (>180 mg/dL or >430 nmol/L) who they sug-
gest may have a lifetime risk of ASCVD equivalent to that of heterozygous FH (Mach
et al. 2019). They do recommend that Lp(a) measurement be considered at least once
in each adult person’s lifetime to assist with risk stratification, particularly in those
considered at moderate or higher risk (Mach et al. 2019). The HEART-UK consensus
statement on Lp(a) also supports the measurement of Lp(a) levels in patients with a
personal or family history of premature ASCVD, those with FH or other genetic dys-
lipidaemias (such as familial combined hypercholesterolaemia) or early-onset
ASCVD and patients with first-degree relatives who have significantly elevated Lp(a)
(>200 nmol/L) levels. The statement suggests that the cardiovascular risk conferred
by Lp(a) is determined by its serum concentration, with 32–90 nmol/L equivalent to
minor risk, 90–200 nmol/L to moderate risk and 200–400 nmol/L to high risk, with
concentrations >400 nmol/L equivalent to very high risk (Kostner et al. 2018),
Table 17.2.
The National Lipid Association (NLA) suggests that the 80th percentile in pre-
dominantly Caucasian US populations is ~100 nmol/L and ~150 nmol/L in African
Americans, although it is unclear whether different risk thresholds should be applied
(Wilson et al. 2019). The American Heart Association (AHA) and American College
of Cardiology (ACC) recognise elevated Lp(a) as a ‘risk-enhancing factor’ in the
development of ASCVD, with levels ≥125 nmol/L (≥50 mg/dL) considered high
risk (Grundy et al. 2018). Other groups, including the Canadian Cardiovascular
Society and the Mighty Medic Group, suggest that Lp(a) might aid risk assessment
in patients at high risk or with premature CVD/CAD, with Lp(a) levels <30 mg/dL
considered normal (Anderson et al. 2016). Two International Classification of
Table 17.2 Risk thresholds for Lp(a) concentration (adapted from Heart UK consensus statement)
ASCVD risk Lp(a) level, (nmol/L) Lp(a) level, (mg/dL) Percentile of population
Moderate 100–200 40–90 80–95th
High 200–400 90–180 95–99th
Very high >400 >180 99th
Source: Cegla et al. (2019)
17 When Should We Measure Lipoprotein(a)? 279
Diseases (ICD)-10 codes have been added to justify Lp(a) testing, E78.41 = ele-
vated Lp(a) and Z83.430 = Family History of elevated Lp(a).
References
Albers JJ, Slee A, O'Brien KD, Robinson JG, Kashyap ML, Kwiterovich PO Jr, Xu P, Marcovina
SM. Relationship of apolipoproteins A-1 and B, and lipoprotein(a) to cardiovascular outcomes:
the AIM-HIGH trial (atherothrombosis intervention in metabolic syndrome with low HDL/
high triglyceride and impact on Global Health outcomes). J Am Coll Cardiol. 2013;62:1575–9.
Anderson TJ, Gregoire J, Pearson GJ, et al. 2016 Canadian Cardiovascular Society Guidelines for
the management of dyslipidemia for the prevention of cardiovascular disease in the adult. Can
J Cardiol. 2016;32:1263–82.
Cegla J, Neely RDG, France M, Ferns G, Byrne CD, Halcox J, Datta D, Capps N, Shoulders C,
Qureshi N, Rees A, Main L, Cramb R, Viljoen A, Payne J, Soran H, HEART UK Medical,
Scientific and Research Committee. HEART UK consensus statement on lipoprotein(a): a call
to action. Atherosclerosis. 2019;291:62–70.
Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti
S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW,
Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith
SC Jr, Sperling L, Virani SS, Yeboah J. AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/
APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: a report of the
280 K. Kostner
Lipoprotein(a) [Lp(a)] represents one of the most promising, causal independent risk
factors for a chronic disease like atherosclerotic cardiovascular disease [ASCVD]
that has emerged this century. Its appropriate use hinges on well-targeted implemen-
tation. This requires understanding of the whole analytical cycle (pre-analytical, ana-
lytical and post-analytical), as well as the involvement of many stakeholders. Patients
who require testing need to be aware of the importance of undertaking the test.
Ordering physicians need to be cognisant of target populations and the ways in
which results should be applied. Laboratory scientists should consider the subtleties
and intricacies of Lp(a) measurement. Implementation requires dialogue with the
diagnostic industry which carries much of the responsibility for the provision of
robust, validated products with traceable standardization processes which fulfil the
governmental approval and monitoring processes, thereby guaranteeing minimum
standards. Preventive health experts can optimize the manner in which Lp(a) results
are applied, whilst healthcare system administrators need to be able to appreciate the
health and economic benefits associated with testing, as well as the limits beyond
which this may become counterproductive. All stakeholders must be involved in the
establishment of these objectives for the optimum widespread clinical testing of
Lp(a). Ongoing attention will need to be directed towards utilization of Lp(a) testing
in the future because it is likely to evolve over time, especially during the introduc-
tion of specific forms of treatment which target Lp(a) directly.
The transition from a research biomarker to a clinically relevant laboratory risk
factor requires appreciation of the expectations that apply to the clinical environ-
ment in which Lp(a) will be measured. Like research laboratories, clinical service
laboratories need to maintain the highest standards of process control throughout
the analytical cycle. This extends from the pre-analytical sample collection and
preparation to the reporting and management of Lp(a) levels. It represents a setting
in which fastidious attention to correct patient identification and curation of the
sample is paramount. One of the advantages of laboratory automation is the reduc-
tion in the opportunity for sources of human error such as transcription errors.
Optimization and monitoring of accuracy and precision, which are components of
total laboratory error, are implicit in the objectives of any analytical laboratory.
Whilst research laboratories appropriately exploit a degree of independence in their
approach to analytical problems, clinical service laboratories need to function
within the context of laboratory networks and the wider healthcare system. This
requires a high level of collaboration and collegiate activity. Such collaboration
forms the basis for national and international standardization programmes, harmo-
nization initiatives, reference range and laboratory report consensus as well as pub-
lic health recommendations. More subtle considerations such as equity, accessibility
and intrinsic value within the healthcare system also require careful consideration.
The clinical setting in which Lp(a) is tested also affects interpretation. Lp(a) is
an acute phase reactant, so sample collection should be postponed until patients
have recovered from acute inflammatory episodes or concurrent illness. One excep-
tion is in the setting of acute coronary syndrome because the opportunity to identify
high-risk patients outweighs the risk of false-positive results. On the other hand,
requirements for urgent laboratory turnaround time are less applicable to Lp(a) for
the time being. In comparison to other analytes, technical aspects of Lp(a) measure-
ment have posed substantially greater challenges to the implementation of Lp(a)
testing than is usually the case. Nevertheless, clinicians can be assured that Lp(a)
testing is fit for purpose. Furthermore, current developments are rapidly overcoming
the remaining challenges, as will be discussed in more detail.
Method Selection
evolution (Utermann 1999), whilst its absence from other species except humans
and higher apes remains difficult to explain (Utermann 1999). The absence of apo(a)
in research models such as mice and rabbits has created logistic limitations because
studies in such models require gene expression that is limited to one or two specific
isoforms in each model. Furthermore, the high degree of polymorphism of the LPA
gene locus creates an unusual degree of inter-individual genotypic and phenotypic
variability. This poses important demands on clinical laboratory measurement
which impact the commercial production and validation of diagnostic reagents such
as monoclonal antibodies.
Another noteworthy aspect of Lp(a) is its highly skewed distribution (Kronenberg
and Utermann 2013), which deviates markedly from normal distribution. Statistical
analysis requires transformation, such as logarithmic transformation, or the use of
non-parametric statistics. Another consequence of the skewed distribution is Lp(a)’s
wide analytical range. Methods need to be able to quantify levels which may be
nearly tenfold higher than the upper limit of ideal methods (Stefanutti et al. 2020).
Lp(a) is a polymorphic particle that consists of a low-density lipoprotein (LDL)
particle covalently linked via a di-sulphide bond to apo(a). There is one apo(a) and
one apolipoprotein B [apoB] molecule per particle (Albers et al. 1996). Apo(a) is
the protein product of the highly polymorphic LPA gene locus which codes for this
large protein in which a variable number of plasminogen-like kringle repeats are
present (Cegla et al. 2021). In plasma, Lp(a) is the major transporter of oxidized
phospholipid. This may contribute towards Lp(a)’s pro-atherogenic, pro-
inflammatory and pro-thrombotic properties (Scipione et al. 2015). In theory, the
separate components of Lp(a) offer alternative options for quantification.
The cholesterol and other lipid components of lipoproteins are strongly impli-
cated in the pathophysiology of ASCVD. The measurement of Lp(a)’s cholesterol
component would provide a consistent frame of reference for the pathogenicity of
different lipoprotein classes, including Lp(a). On the other hand, the confounding
effect of triglyceride via modification by cholesterol ester transfer protein can alter
the size and density of atherogenic lipoproteins. This confuses the relationship
between lipoprotein cholesterol and lipoprotein number (Carr et al. 2019). There is
clear evidence that the atherogenic effect of most Apo B100-containing particles is
proportional to their number rather than their cholesterol content. Efforts have been
made to quantify Lp(a) in terms of cholesterol content, but the methods are not
robust and evidence of specific advantages over other methods is lacking.
Furthermore, the concordance of Lp(a) cholesterol measurement with Lp(a) molar
results has been called into question (Konerman et al. 2012).
The measurement of Lp(a)’s oxidized phospholipid content has been deduced
from immunoassay quantification via antibody E06 (Tsimikas et al. 2009). This
suggests that most oxidized phospholipid is transported by Lp(a). Whilst excellent
correlation between oxidized phospholipid immunoassay results and measured
Lp(a) levels has been demonstrated, other techniques such as lipidomic measure-
ment by mass spectroscopy suggest that the transport of oxidized phospholipid on
Lp(a) is potentially more complex (Leibundgut et al. 2013). The main advantage of
quantification of Lp(a)-associated oxidized phospholipid is the quantification of a
284 D. Sullivan et al.
particular potentially toxic component, but as has already been explained, other
components such as cholesterol are likely to have a modifying effect. The current
clinical laboratory approach to Lp(a) is that it is best measured via its unique apo(a)
component and that measurement of the cholesterol or oxidized phospholipid com-
ponents of Lp(a) is not warranted because they are not consistent with the need to
measure Lp(a) particle number.
Lp(a) levels have been assessed in terms of mass or molar units. LPA genotype
has been assessed mainly in terms of kringle IV type 2 [KIVT2] repeats. The pres-
ence of a greater number of KIVT2 repeats is associated with a relative reduction in
plasma Lp(a) molar concentrations. Metabolic turnover studies suggest that the
effect is mediated via Lp(a) synthesis (Chan et al. 2019). Other genetic variations
further modify the relationship (Coassin et al. 2017, 2019), but overall the relation-
ship between mass and molar assessments of Lp(a) concentration is confounded
because larger molecular weight isoforms are associated with a smaller number of
particles. For example, the protein composition of Lp(a) has been shown to vary
between 30 and 46% by weight (Ruhaak and Cobbaert 2020). Given that mass (mg/
dL) measurements are affected by all components of the Lp(a) particle, they are
inherently more variable than measures of particle concentration. This has estab-
lished the need to quantify Lp(a) in molar units (nmol/L) (Ruhaak and Cobbaert
2020). Unfortunately, many of the historically seminal clinical studies were con-
ducted at a time when this relationship was less evident. As a result, quantification
in terms of mass units (g/L, mg/dl) lingers as a legacy.
Based on these principles, Lp(a) should be measured by a method (e.g. immuno-
assay or mass spectroscopy) that is apo(a) isoform independent. This has required the
introduction of appropriate standards, calibrators and calibration protocols designed
to permit estimation and reporting in molar rather than mass units. The necessary
processes have been pursued throughout the past two to three decades and have
involved phase 1 and phase 2 standardization programmes conducted by IFCC
(International Federation of Clinical Chemistry) (Tate et al. 1998, 1999), leading to a
WHO/IFCC reference reagent for immunoassay (SRM 2B) (Dati et al. 2004). This
has occurred in parallel with the development of mass spectroscopy methods
(Cobbaert et al. 2021) including a proposed candidate reference method (Marcovina
et al. 2021). Sustained efforts by dedicated clinical scientists have put in place the
associated safety and quality measures which are required to maintain laboratory
performance (Marcovina and Albers 2016). This will be discussed in the next section.
Table 18.1 demonstrates that these initiatives continue to penetrate the market for
diagnostic Lp(a) immunoassays. As a result, isoform-specific assays which report in
molar units have started to predominate. Diagnostic companies will continue to
drive this process provided such a prerequisite continues to be demanded by clini-
cians (Wyness and Genzen 2021). The transition from mass units (mg/dL) to molar
units (nmol/L) is necessary because the inverse relationship between genetically
determined apo(a) KIVT2 repeats and Lp(a) particle number confounds the concept
of a single standard conversion factor (Tsimikas et al. 2018). Conversion factors
also vary with the assay, Lp(a) concentration and storage. Although equivalent mass
levels could be identified for the molar levels designated as medical decision-
making cut-offs, mass units should be phased out as soon as possible. Lp(a)-
lowering treatment may require serial measurements in individual patients,
18 Measurement of Lipoprotein(a) in the Clinical Laboratory 285
preferably using the same assay, which makes the sole use of molar unit measure-
ments more logical and more urgent.
Immunoassay is usually the preferred method for high-throughput laboratories
due to logistic requirements. On the other hand, immunoassays may struggle in
comparison to the sensitive and specific results that can be achieved with dedicated
mass spectroscopy methods. Sophisticated and highly informative mass spectros-
copy methods have been described for the dedicated measurement of Lp(a)
(Lassman et al. 2014). Whilst such methods are generally robust, they may be more
difficult to align with external quality assurance programme method groups. This is
an important consideration because inter laboratory bias would lead to inconsis-
tency in the application of the cut-offs for medical decision-making.
Clinical laboratories are also able to analyse or refer samples for LPA genetic
analysis. LPA genetic polymorphisms exert most of their effects via the quantitative
phenotype of the associated Lp(a) level. Currently, LPA genotyping offers little in
the way of additional clinical benefit beyond quantitative plasma Lp(a) levels, so
there is little incentive to study the genotype separately for clinical purposes. On the
other hand, LPA genotype is one of the major contributors to “polygenic” risk scores
for cardiovascular disease [CVD] (Trinder et al. 2021). The separate contribution of
the two LPA gene alleles is usually managed by summation which reinforces the
concept of co-dominant inheritance of the plasma Lp(a) trait. Another potential
application of LPA genotyping is the possibility that pharmacogenomic assessment
of LPA may identify subjects who are likely to benefit from aspirin therapy for the
prevention of ASCVD (Shiffman et al. 2012). If required, routine genotyping meth-
ods such as massively parallel sequencing should suffice, even though this is not
ideal for detection of nucleotide repeats.
are compared to all participants, and they are also grouped according to method,
reagents, equipment, etc. (Fig. 18.1). Target values are based on participant medians
and acceptable variation about the target determined using biological variation.
Duplication of some samples allows assessment of precision as well as accuracy.
Figure 18.1 shows an EQA result to illustrate several considerations that apply to
Lp(a). Firstly, the results were reported in mass units. The programme has responded
to requests to switch to molar units. Secondly, the samples must cater to several
analytes. In the case of the special lipid programme, the samples are created by
spiking with a lipoprotein concentrate to create a linear escalation of concentration
in duplicate samples. In this case, the concentrate lacked sufficient Lp(a) to approach
the lowest medical decision point. It will be logistically difficult to adjust for the
complexities of Lp(a) for several reasons. Firstly, the lipoprotein concentrate may
be derived from pooled samples, in which case the samples will comprise a mixture
of isoforms rather than the homozygous or heterozygous pattern expected in indi-
vidual patient sera. Secondly, whilst it may be possible to increase levels towards
the lower medical decision points, it may be difficult to encompass higher levels.
This may become important if change in Lp(a) levels becomes a treatment target. It
may be commercially difficult for EQA programmes to deal with Lp(a) separately
from other lipoproteins. The fact that some EQAs add TRL and high-density lipo-
protein in parallel rather than in reciprocal amounts illustrates that certain lipopro-
tein EQA results need to be interpreted with caution (Perera et al. 2010). The
standardization and harmonization initiatives which were mentioned earlier have
been particularly active in this area. EQA programmes for Lp(a) have been estab-
lished and analysed (Cegla et al. 2021; Cobbaert et al. 2012), but ongoing efforts
will be required (Scharnagl et al. 2019).
Fig. 18.1 Representative report (RCPAQAP Special Lipids QAP 2021) of EQA results for Lp(a)
prior to transition to molar units
18 Measurement of Lipoprotein(a) in the Clinical Laboratory 289
Clinical Application
Interpretation
and peripheral artery disease [PAD], as well as calcific aortic valve disease [CAVD],
are based on phenotypic and genotypic techniques. Due to its skewed distribution,
Lp(a) is not suitable for traditional definitions of a “normal” reference interval.
Plasma Lp(a) levels are positively skewed, with the median for Caucasian popula-
tions ~20 nmol/L (<10 mg/dL). However, like many other risk factors for chronic
disease, the upper limit of normal is less relevant than the threshold level at which
increased risk necessitates a particular medical decision. Notions of sensitivity and
specificity of Lp(a) testing will depend on the level at which such a cut-point is set.
Conversely, there is the reassurance that low levels of Lp(a) are associated with
reduced CVD risk (Coassin et al. 2017) and do not seem to be associated with any
pathological outcomes (Langsted et al. 2021).
One perplexing aspect of Lp(a) is its variation in association with racial differ-
ences. This seems to reflect multiple genetic variations including some in the region
of kringles KIV T6–T10 (Utermann 1999). Evidence suggests that this confounds
the quantitative relationship between Lp(a) level and CVD risk in some racial
groups (Geethanjali et al. 2003). This implies that medical decision points may need
to be adjusted to take account of the widely reported effects of race on Lp(a) level
(Stefanutti et al. 2020; Reyes-Soffer 2021; Ogorelkova et al. 2001). Studies are
lacking in the many Indigenous groups in whom socio-economic determinants of
health have created an excessive burden of CVD.
The perceived utility of Lp(a) testing depends on its ability to reclassify CVD
risk, particularly amongst those who are deemed to be at “intermediate risk” by
traditional methods. Lp(a) has demonstrated excellent capability in this regard in
the Bruneck Study (Willeit et al. 2014). The added benefit of Lp(a) in CVD risk
assessment may be presented as its contribution to the “C” statistic, but the author
of studies in which this estimate has been modest or gender-specific (Cook et al.
2018; Khera et al. 2014) has cautioned against the exclusion of biomarkers on this
basis (Cook 2007). Lp(a) levels may also modify the management (Burgess et al.
2018) of individuals who are not identified by routine risk factor assessments. This
includes young MI patients (Berman et al. 2021) and the relatives of those with
increased Lp(a). In due course, measurement of Lp(a) concentration could be con-
sidered in adults on at least one occasion to assess risk of ASCVD, but this amounts
to population screening, which will require convincing cost-benefit evidence. Lp(a)
testing and interpretation of results will be governed by policies determined by the
local healthcare system.
Healthcare Systems
The implementation of Lp(a) testing will depend on the policies of the relevant
healthcare systems. Their expectations will reflect the safety, quality and value per-
spectives outlined above. Clinical laboratories may consider the use of alerts and
interpretive comments on laboratory reports. These may emphasize the potential
need for assessment of ASCVD risk and cascade testing. Digital health technologies
18 Measurement of Lipoprotein(a) in the Clinical Laboratory 291
Conclusion
References
Albers JJ, Kennedy H, Marcovina SM. Evidence that Lp[a] contains one molecule of apo[a] and
one molecule of apoB: evaluation of amino acid analysis data. J Lipid Res. 1996;37(1):192–6.
Berman AN, Biery DW, Singh A, Wu WY, Divakaran S, DeFilippis EM, et al. Atherosclerotic
cardiovascular disease risk and elevated lipoprotein(a) among young adults with myocardial
infarction: the Partners YOUNG-MI Registry. Eur J Prev Cardiol. 2021;28(8):e12–e4.
Bittner VA, Szarek M, Aylward PE, Bhatt DL, Diaz R, Edelberg JM, et al. Effect of alirocumab
on lipoprotein(a) and cardiovascular risk after acute coronary syndrome. J Am Coll Cardiol.
2020;75(2):133–44.
292 D. Sullivan et al.
Burgess S, Ference BA, Staley JR, Freitag DF, Mason AM, Nielsen SF, et al. Association of LPA
variants with risk of coronary disease and the implications for lipoprotein(a)-lowering thera-
pies: a Mendelian randomization analysis. JAMA Cardiol. 2018;3(7):619–27.
Carr SS, Hooper AJ, Sullivan DR, Burnett JR. Non-HDL-cholesterol and apolipoprotein B
compared with LDL-cholesterol in atherosclerotic cardiovascular disease risk assessment.
Pathology. 2019;51(2):148–54.
Cauza E, Kletzmaier J, Bodlaj G, Dunky A, Herrmann W, Kostner K. Relationship of non-LDL-
bound apo(a), urinary apo(a) fragments and plasma Lp(a) in patients with impaired renal func-
tion. Nephrol Dial Transplant. 2003;18(8):1568–72.
Cegla J, Neely RDG, France M, Ferns G, Byrne CD, Halcox J, et al. HEART UK consensus state-
ment on lipoprotein(a): a call to action. Atherosclerosis. 2019;291:62–70.
Cegla J, France M, Marcovina SM, Neely RDG. Lp(a): when and how to measure it. Ann Clin
Biochem. 2021;58(1):16–21.
Chan DC, Watts GF, Coll B, Wasserman SM, Marcovina SM, Barrett PHR. Lipoprotein(a) par-
ticle production as a determinant of plasma lipoprotein(a) concentration across varying
apolipoprotein(a) isoform sizes and background cholesterol-lowering therapy. J Am Heart
Assoc. 2019;8(7):e011781.
Chennamsetty I, Claudel T, Kostner KM, Baghdasaryan A, Kratky D, Levak-Frank S, et al.
Farnesoid X receptor represses hepatic human APOA gene expression. J Clin Invest.
2011;121(9):3724–34.
Coassin S, Erhart G, Weissensteiner H, de Eca Guimaraes Araujo M, Lamina C, Schonherr S, et al.
A novel but frequent variant in LPA KIV-2 is associated with a pronounced Lp(a) and cardio-
vascular risk reduction. Eur Heart J. 2017;38(23):1823–31.
Coassin S, Schonherr S, Weissensteiner H, Erhart G, Forer L, Losso JL, et al. A comprehensive
map of single-base polymorphisms in the hypervariable LPA kringle IV type 2 copy number
variation region. J Lipid Res. 2019;60(1):186–99.
Cobbaert C, Weykamp C, Franck P, de Jonge R, Kuypers A, Steigstra H, et al. Systematic moni-
toring of standardization and harmonization status with commutable EQA-samples—5 year
experience from the Netherlands. Clin Chim Acta. 2012;414:234–40.
Cobbaert CM, Althaus H, Begcevic Brkovic I, Ceglarek U, Coassin S, Delatour V, et al. Towards
an SI-traceable reference measurement system for seven serum apolipoproteins using bottom-
up quantitative proteomics: conceptual approach enabled by cross-disciplinary/cross-sector
collaboration. Clin Chem. 2021;67(3):478–89.
Cohn JS, Lam CW, Sullivan DR, Hensley WJ. Plasma lipoprotein distribution of apolipoprotein(a)
in the fed and fasted states. Atherosclerosis. 1991;90(1):59–66.
Cook NR. Use and misuse of the receiver operating characteristic curve in risk prediction.
Circulation. 2007;115(7):928–35.
Cook NR, Mora S, Ridker PM. Lipoprotein(a) and cardiovascular risk prediction among women.
J Am Coll Cardiol. 2018;72(3):287–96.
Dati F, Tate JR, Marcovina SM, Steinmetz A, International Federation of Clinical Chemistry and
Laboratory Medicine, et al. First WHO/IFCC international reference reagent for lipoprotein(a)
for immunoassay-Lp(a) SRM 2B. Clin Chem Lab Med. 2004;42(6):670–6.
de Boer LM, Oorthuys AO, Wiegman A, Langendam MW, Kroon J, Spijker R, et al. Statin ther-
apy and lipoprotein(a) levels: a systematic review and meta-analysis. Eur J Prev Cardiol.
2022;29(5):779–92.
Dieplinger H, Utermann G. The seventh myth of lipoprotein(a): where and how is it assembled?
Curr Opin Lipidol. 1999;10(3):275–83.
Frank S, Hrzenjak A, Blaschitz A, Dohr G, Kostner GM. Role of various tissues in apo(a) frag-
mentation and excretion of fragments by the kidney. Eur J Clin Investig. 2001;31(6):504–12.
Geethanjali FS, Luthra K, Lingenhel A, Kanagasaba-Pathy AS, Jacob J, Srivastava LM, et al.
Analysis of the apo(a) size polymorphism in Asian Indian populations: association with Lp(a)
concentration and coronary heart disease. Atherosclerosis. 2003;169(1):121–30.
Gencer B, Giugliano RP, Tang M, Mach F, Stroes ES, Gouni-berthold I, et al. Clinical safety out-
comes in relation to lipoprotein(a) concentration: insights from the FOURIER trial. Circulation.
2021;144(Suppl_1):A11043-A.
18 Measurement of Lipoprotein(a) in the Clinical Laboratory 293
Gidding SS, Stone NJ, Bookstein LC, Laskarzewski PM, Stein EA. Month-to-month variability
of lipids, lipoproteins, and apolipoproteins and the impact of acute infection in adolescents. J
Pediatr. 1998;133(2):242–6.
Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, et al. 2018 AHA/
ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline
on the management of blood cholesterol: a report of the American College of Cardiology/
American Heart Association task force on clinical practice guidelines. J Am Coll Cardiol.
2019;73(24):e285–350.
Hopewell JC, Clarke R, Watkins H. Lp(a) (lipoprotein[a]), an exemplar for precision medicine.
Arterioscler Thromb Vasc Biol. 2021;41(1):475–7.
Ignatescu M, Kostner K, Zorn G, Kneussl M, Maurer G, Lang IM, et al. Plasma Lp(a) levels are
increased in patients with chronic thromboembolic pulmonary hypertension. Thromb Haemost.
1998;80(2):231–2.
Jawi MM, Frohlich J, Chan SY. Lipoprotein(a) the insurgent: a new insight into the structure, func-
tion, metabolism, pathogenicity, and medications affecting lipoprotein(a) molecule. J Lipids.
2020;2020:3491764.
Kamstrup PR. Lipoprotein(a) and cardiovascular disease. Clin Chem. 2020;67(1):154–66.
Khera AV, Everett BM, Caulfield MP, Hantash FM, Wohlgemuth J, Ridker PM, et al. Lipoprotein(a)
concentrations, rosuvastatin therapy, and residual vascular risk: an analysis from the JUPITER
trial (justification for the use of statins in prevention: an intervention trial evaluating rosuvas-
tatin). Circulation. 2014;129(6):635–42.
Konerman M, Kulkarni K, Toth PP, Jones SR. Lipoprotein(a) particle concentration and
lipoprotein(a) cholesterol assays yield discordant classification of patients into four physiologi-
cally discrete groups. J Clin Lipidol. 2012;6(4):368–73.
Koschinsky ML, Marcovina SM. Structure–function relationships in apolipoprotein(a): insights
into lipoprotein(a) assembly and pathogenicity. Curr Opin Lipidol. 2004;15(2):167–74.
Kostner KM, Kostner GM. Factors affecting plasma lipoprotein(a) levels: role of hormones and
other nongenetic factors. Semin Vasc Med. 2004;4(2):211–4.
Kostner KM, Kostner GM. Lipoprotein(a): a historical appraisal. J Lipid Res. 2017;58(1):1–14.
Kostner GM, Steinmetz A. Standardization of Lp(a) measurements. Clin Genet. 1997;52(5):
393–7.
Kostner GM, Ibovnik A, Holzer H, Grillhofer H. Preparation of a stable fresh frozen primary
lipoprotein[a] (Lp[a]) standard. J Lipid Res. 1999;40(12):2255–63.
Kostner K, Clodi M, Bodlaj G, Hoffmann U, Hermann K, Maurer G, et al. Urinary apo(a) excre-
tion is not altered by changes in glomerular filtration rate and renal plasma flow in healthy
males. Wien Klin Wochenschr. 2000;112(3):121–5.
Kostner K, Spitzauer S, Rumpold H, Maurer G, Knipping G, Hrzenjak A, et al. Urinary excretion
of apolipoprotein(a): relation to other plasma proteins. Clin Chim Acta. 2001;304(1–2):29–37.
Kraft HG, Lingenhel A, Raal FJ, Hohenegger M, Utermann G. Lipoprotein(a) in homozygous
familial hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2000;20(2):522–8.
Kronenberg F, Utermann G. Lipoprotein(a): resurrected by genetics. J Intern Med. 2013;273(1):6–30.
Kronenberg F, Trenkwalder E, Lingenhel A, Friedrich G, Lhotta K, Schober M, et al. Renovascular
arteriovenous differences in Lp[a] plasma concentrations suggest removal of Lp[a] from the
renal circulation. J Lipid Res. 1997;38(9):1755–63.
Langlois MR, Nordestgaard BG, Langsted A, Chapman MJ, Aakre KM, Baum H, et al. Quantifying
atherogenic lipoproteins for lipid-lowering strategies: consensus-based recommendations from
EAS and EFLM. Clin Chem Lab Med. 2020;58(4):496–517.
Langsted A, Nordestgaard BG, Kamstrup PR. Low lipoprotein(a) levels and risk of disease in a
large, contemporary, general population study. Eur Heart J. 2021;42(12):1147–56.
Lassman ME, McLaughlin TM, Zhou H, Pan Y, Marcovina SM, Laterza O, et al. Simultaneous
quantitation and size characterization of apolipoprotein(a) by ultra-performance liquid chro-
matography/mass spectrometry. Rapid Commun Mass Spectrom. 2014;28(10):1101–6.
Leibundgut G, Scipione C, Yin H, Schneider M, Boffa MB, Green S, et al. Determinants of
binding of oxidized phospholipids on apolipoprotein(a) and lipoprotein(a)1. J Lipid Res.
2013;54(10):2815–30.
294 D. Sullivan et al.
Ma L, Chan DC, Ooi EMM, Marcovina SM, Barrett PHR, Watts GF. Apolipoprotein(a) kinetics
in statin-treated patients with elevated plasma lipoprotein(a) concentration. J Clin Endocrinol
Metab. 2019;104(12):6247–55.
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, et al. 2019 ESC/EAS
Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular
risk: the task force for the management of dyslipidaemias of the European Society of Cardiology
(ESC) and European Atherosclerosis Society (EAS). Eur Heart J. 2020;41(1):111–88.
Marcovina SM, Albers JJ. Lipoprotein(a) measurements for clinical application. J Lipid Res.
2016;57(4):526–37.
Marcovina SM, Koschinsky ML, Albers JJ, Skarlatos S. Report of the National Heart, Lung, and
Blood Institute workshop on lipoprotein(a) and cardiovascular disease: recent advances and
future directions. Clin Chem. 2003;49(11):1785–96.
Marcovina SM, Viney NJ, Hughes SG, Xia S, Witztum JL, Tsimikas S. Temporal variability in
lipoprotein(a) levels in patients enrolled in the placebo arms of IONIS-APO(a)Rx and IONIS-
APO(a)-LRx antisense oligonucleotide clinical trials. J Clin Lipidol. 2018a;12(1):122–9.e2.
Marcovina SM, Moriarty PM, Koschinsky ML, Guyton JR. JCL roundtable-lipoprotein(a): the
emerging risk factor. J Clin Lipidol. 2018b;12(6):1335–45.
Marcovina SM, Clouet-Foraison N, Koschinsky ML, Lowenthal MS, Orquillas A, Boffa MB, et al.
Development of an LC-MS/MS proposed candidate reference method for the standardization
of analytical methods to measure lipoprotein(a). Clin Chem. 2021;67(3):490–9.
McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors.
Pathology. 2019;51(2):155–64.
Nordestgaard BG, Langsted A. Lipoprotein(a) as a cause of cardiovascular disease: insights from
epidemiology, genetics, and biology. J Lipid Res. 2016;57(11):1953–75.
O’Donoghue ML, Fazio S, Giugliano RP, Stroes ES, Kanevsky E, Gouni-Berthold I, et al.
Lipoprotein(a), PCSK9 inhibition, and cardiovascular risk: insights from the FOURIER trial.
Circulation. 2019;139(12):1483–92.
Ogorelkova M, Kraft HG, Ehnholm C, Utermann G. Single nucleotide polymorphisms in exons of
the apo(a) kringles IV types 6–10 domain affect Lp(a) plasma concentrations and have differ-
ent patterns in Africans and Caucasians. Hum Mol Genet. 2001;10(8):815–24.
Pearson GJ, Thanassoulis G, Anderson TJ, Barry AR, Couture P, Dayan N, et al. 2021 Canadian
Cardiovascular Society guidelines for the management of dyslipidemia for the prevention of
cardiovascular disease in adults. Can J Cardiol. 2021;37(8):1129–50.
Perera NJ, Burns JC, Perera RS, Lewis B, Sullivan DR. Adjustment of direct high-density lipopro-
tein cholesterol measurements according to intercurrent triglyceride corrects for interference
by triglyceride-rich lipoproteins. J Clin Lipidol. 2010;4(4):305–9.
RCPAQAP Special Lipids QAP. 2021. https://secureweb.cisco.com/1dzuXRbR1KAX_7ggbjpvP
Gt5IJV2Pnrvs9vznt7Cghk2YiZoRRIUUf_HHRYRYrQHfkMf7KNXjsIc14cP3qzfdJfyQGbW
GsW4u0iOLVseg9TYLy9Q7vdzs7-T888Dn8REx1G7bRxTDaZpLkeguIaw-jN7zonxvy-bI9H
RgZcd4apLAeQ3uMnqck1csbo3UMp8o_W4HZjKRuoEY2XWRmdLtJ5oyh0IqmTb_XzW
QrwaLp6ej8HrZ7AS7NoZ9ccXCqbOzc8zMrYzqlB9DmHwCZpS3IKPtBn5zOnp29HqmHG
vkwQksCUVPurBF9DHiMKhR9knvyh9ZXKTIMlJfm-2t576iB5ETJ6nRF1rZmp2lkDmNtlb
w4AkGtqoZXEXcQoIpDl9JtZ1Vu4-NNSXguZ6xKtC0nL5_shfxbb8qv9bNw7eSKga4a3Cfy-
WivLk4IXIJExIB1TFbjVg7U2M1tKV5x0av57ynHpcT-2sbh0qgGR0RFgSTymLJu_SRDIf_
V0Marsxzt/https%3A%2F%2Fround-lake.dustinice.workers.dev%3A443%2Fhttps%2Frcpaqap.com.au%2Fproducts%2F
Reyes-Soffer G. The impact of race and ethnicity on lipoprotein(a) levels and cardiovascular risk.
Curr Opin Lipidol. 2021;32(3):163–6.
Reyes-Soffer G, Ginsberg HN, Berglund L, Duell PB, Heffron SP, Kamstrup PR, et al.
Lipoprotein(a): a genetically determined, causal, and prevalent risk factor for atherosclerotic
cardiovascular disease: a scientific statement from the American Heart Association. Arterioscler
Thromb Vasc Biol. 2022;42(1):e48–60.
Ruhaak LR, Cobbaert CM. Quantifying apolipoprotein(a) in the era of proteoforms and precision
medicine. Clin Chim Acta. 2020;511:260–8.
18 Measurement of Lipoprotein(a) in the Clinical Laboratory 295
Abbreviations
Apo(a) Apolipoprotein(a)
ApoA-I Apolipoprotein A-I
ApoB-100 Apolipoprotein B100
CV Coefficient of variation
CVD Cardiovascular diseases
ID Isotope dilution
IFCC International Federation of Clinical Chemistry
ISO International Organization for Standardization
JCGM Joint Committee for Guides in Metrology
KIV Kringle IV
KIV2 Kringle IV type 2
KIV9 Kringle IV type 9
LC-MS/MS Liquid chromatography-tandem mass spectrometry
LDL Low-density lipoproteins
Lp(a) Lipoprotein(a)
NIST National Institute of Standards and Technology
NWRL Northwest Lipid Research Laboratories
QC Quality controls
SD Standard deviation
N. Clouet-Foraison · T. Vaisar
Division of Metabolism, Endocrinology, and Nutrition, University of Washington,
Seattle, WA, USA
e-mail: [email protected]; [email protected]
S. M. Marcovina (*)
Medpace Reference Laboratories, Cincinnati, OH, USA
e-mail: [email protected]
conservative model and 7.6 billion dollars per year considering medians (Hoerger
et al. 2011).
Finally, in a globalized society, the availability of internationally recognized
clinical decision thresholds and reference intervals for treatment is of major impor-
tance. However, this requires that clinical test performed worldwide provides com-
parable results, which can only be achieved through the implementation of the
concept of metrological traceability.
issues (Stoppacher et al. 2015; Josephs et al. 2019). Then, instead of proceeding
with standardization, the alternative strategy is to establish harmonization of the
methods by producing a matrix-based secondary reference material with value
assigned by an arbitrarily designated reference method. The use of this material as
common calibrator for all the other procedures usually improves comparability of
the methods to a certain degree. However, since it is not anchored to the SI, harmo-
nization of the methods does not ensure accuracy of the measurements nor stability
of the values over time.
Furthermore, it is important to highlight that even though standardization and
harmonization may both improve between-method comparability, they do not guar-
antee it (Miller et al. 2014b). Indeed, there are multiple additional pre-analytical,
analytical, or post-analytical factors that can negatively impact method comparabil-
ity. In particular, the use of different methodologies to isolate, target, and measure
the analyte such as different antigen epitopes, different isolation techniques or
detection systems and methodologies, varying interferences, and different measure-
ment units can drastically impact method comparability. In this situation, even
though standardization is achieved, method comparability will remain poor until the
assays are improved, properly validated and common measurement units used.
Therefore, a prerequisite for a successful standardization is that the methods dem-
onstrate the necessary analytical performances to be deemed “standardizable.”
Lp(a) is a highly complex lipoprotein formed by a particle very similar in lipid and
protein composition to low-density lipoproteins (LDL) but characterized by the
presence of a single molecule of a unique protein, apolipoprotein(a) [apo(a)], bound
to the ApoB100 of LDL by a single disulfide bond (Schmidt et al. 2016). Circulating
serum levels of Lp(a) are predominantly genetically determined by the LPA gene
and do not substantially vary over time (Kronenberg 2016), although physiological,
dietary, hormonal, and environmental factors do contribute to its biological varia-
tion (Enkhmaa et al. 2016; Garnotel et al. 1998).
Apo(a) is a heavily glycosylated protein and its presence imparts distinct proper-
ties to Lp(a) distinguishing it from LDL (Nordestgaard et al. 2010; Tsimikas 2017;
Van Der Valk et al. 2016). Apo(a) shares a high amino acid sequence homology with
several regions of plasminogen, including the protease domain, and the kringles IV
(KIV) and V domains (Koschinsky and Marcovina 2004) and exhibits a high degree
of size polymorphism. The KIV domain of apo(a) is formed by ten distinct KIV
types numbered from 1 to 10. All KIV types, except KIV type 2 (KIV2), are present
in apo(a) as a single copy, while the KIV2 varies from <3 to >40 identical repeats,
resulting in the large number of apo(a) isoform sizes circulating in human plasma
(Marcovina et al. 1993). Being mostly determined by its hepatic production rate, the
concentration of apo(a) is largely inversely correlated to its size, smaller isoforms
being produced faster (Karwatowska-Prokopczuk et al. 2021). The distribution of
apo(a) serum levels and isoforms varies widely between individuals and popula-
tions of different ancestry, and because most individuals express two different
alleles of the LPA gene, a majority of individuals presents two different size iso-
forms circulating in plasma (Marcovina et al. 1993; Karwatowska-Prokopczuk et al.
2021; Stefanutti et al. 2020; Kamstrup 2021; Marcovina and Albers 2016). In addi-
tion, apo(a) is heterogeneous in its glycosylation pattern, which occurs both within
the core of the KIV motifs and within the linker sequences connecting the different
kringles, resulting in an extremely heterogeneous population of Lp(a) particles in
circulation (Marcovina and Albers 2016).
Because there is one molecule of apo(a) in Lp(a), the measurement of apo(a) is
used as a surrogate measure of Lp(a) in plasma. At present, Lp(a) concentrations are
reported either in nmol/L of Lp(a) protein or in mg/dL of total Lp(a) mass including
302 N. Clouet-Foraison et al.
the protein, lipid, and carbohydrate components. However, because mass units rely
on doubtful hypotheses regarding Lp(a) particle composition, and because the mass
of apo(a) is highly variable (Marcovina and Albers 2016), guidelines now recom-
mend the use of molar units for Lp(a) reporting in clinical laboratories (Marcovina
and Albers 2016; Wilson et al. 2019; Mach et al. 2019; McCormick 2004). A variety
of immunochemical methods is available to measure Lp(a) in plasma or serum such
as ELISA (enzyme-linked immunosorbent assay), nephelometry, immunoturbidim-
etry, and fluorescent immunoassays. All of them are based on the measurement of
the signal generated by the formation of a complex between apo(a) and specific
monoclonal or polyclonal antibodies. By measuring the signal generated by the
antigen-antibody complex in a calibration material containing a known amount of
the analyte, the signal in the sample can be calculated back to a concentration of the
analyte in the sample. For an assay to be accurate, (1) the antibody needs to be spe-
cific to the analyte measured, (2) the analyte should have the same structural char-
acteristics in the sample and in the assay calibrator to ensure a similar degree of
immunoreactivity per particle, (3) an appropriate reference material should be used
to value-assign the assay calibrator to guarantee reproducibility and comparability
of the results, and (4) harmonized protocols should be available to accurately trans-
fer the value from the reference material to the assay calibrator and to verify that
results obtained on test samples are accurate (Marcovina and Albers 2016).
Although specificity of antibodies to apo(a) is not a major issue because possible
immunoreactivity with apoB-100 or plasminogen can be easily eliminated, the high
degree of intra- and interindividual variation in apo(a) size, due to the variable num-
ber of KIV2 repeats, makes it practically impossible to select assay calibrators with
identical structural characteristics as individual samples. So far, only two monoclo-
nal antibodies have been reported that bind to epitopes that are not present in KIV2
(Marcovina and Albers 2016; Gonen et al. 2020), while all polyclonal antibodies
used in various immunoassays also recognize epitopes located in the variably
repeated KIV2 domain (Marcovina and Albers 2016; Kronenberg and Tsimikas
2019). In this situation, the number of antigen-antibody complexes formed during
analyses reflects the number of KIV2 repeats and thus the isoform distribution of
apo(a) in the individual rather than the apo(a) concentration. If the isoforms are
smaller in the sample than in the calibrator, less immunocomplexes will be formed
in the sample, resulting in underestimation of the concentration of Lp(a), and vice
versa will occur in samples with larger isoforms. It has been estimated that the effect
of apo(a) size variability may result in over- or underestimation of Lp(a) concentra-
tion of up to 25–30% with consequent possible misclassification of the individual’s
cardiovascular risk (Kamstrup 2021; Marcovina and Albers 2016; Marcovina
et al. 1995).
In addition, because the concentration of apo(a) is directly correlated to its pro-
duction rate which in turn is correlated to the size of apo(a), concentrations span
more than 1000-fold range from <1 nmol/L in individuals with large isoforms to
>1000 nmol/L in individuals with small isoforms (Kamstrup 2021; Marcovina and
Albers 2016). The immunoassays must therefore have appropriate calibration
dynamic ranges, meaning that most calibrators will be formed by sample pools with
19 Standardization of Analytical Methods for the Measurement of Lipoprotein(a… 303
isoforms. The QCs and the 80 donor samples were value-assigned by the ELISA-
designated reference method (Marcovina et al. 1995) performing repeated measure-
ments over several weeks and the apo(a) isoforms determined by agarose gel
electrophoresis (Marcovina et al. 1993). Criteria were established to determine the
acceptability of the bias obtained between the observed and the target values for
these samples and the contribution of apo(a) isoform variability on the results. All
requesting manufacturers received the multistep validation protocol, the WHO/
IFCC reference material SRM-2B, the six QC materials, and the set of 80 samples.
Among the analytical systems evaluated during the IFCC standardization pro-
gram, only one latex-enhanced turbidimetric method produced by Denka Seiken,
Japan, demonstrated good agreement with Lp(a) values measured by the ELISA-
designated comparison method, most of the inaccuracy being due to overestimation
of Lp(a) levels in samples with large apo(a) isoform size. An extensive evaluation
of the Denka Seiken method was thus performed at the NWRL and showed that
careful optimization of the assay, coupled with value assignment of the five-point
calibrators with the WHO/IFCC SRM-2B reference material, resulted in improved
agreement of Lp(a) values with those obtained by the ELISA-designated compari-
son method (Marcovina and Albers 2016). Following this work, a fairly large num-
ber of manufacturers implemented the use of the Denka Seiken method on their
instruments or distributed the Denka kits to be used by clinical chemistry laborato-
ries on different analytical systems. Between 2012 and 2015, calibration and per-
formance of 42 analytical systems based on the Denka Seiken kit using different
lots of calibrators and reagents, and 6 methods from different manufacturers based
on single diluted calibrators, were evaluated. The 42 analytical systems based on
the use of the Denka kits meet the established performance criteria while the 6
methods using single calibrators did not, due to high apo(a) isoform-size-depen-
dent biases. After uniform calibration with the WHO/IFCC SRM-2B reference
material, the among-method CV across the 42 measurement systems on the 80
samples was 5.5% and ranged from 2.1% in samples with high Lp(a) concentra-
tions to 10.5% in samples with low Lp(a) concentrations. These findings demon-
strate that harmonization of results obtained by a variety of different instruments
and different calibrator lots can be achieved in optimized test systems (Marcovina
and Albers 2016).
In the period 2015–2020, the analytical performances of 29 test systems using
the Denka Seiken reagents were evaluated by the NWRL. All systems were trace-
able to the WHO/IFCC SRM-2B through the NWRL harmonization process. As
presented in Fig. 19.1a, results show that the average CV between the 29 systems
was 5.0% and ranged from 2.3 to 10.5% with only one sample slightly exceeding
the 10.4% desirable allowed imprecision recommended for clinical measurements
by the Westgard biological variation database (Fraser 2022). Comparison of the
average Lp(a) concentration calculated across the 29 systems versus the concentra-
tion measured by the Lp(a) designated comparison method shows a near perfect
correlation with a 1.01 slope (Fig. 19.1b). As evidenced in Fig. 19.1c, the average
relative difference from the Lp(a) assigned values was consistently below the 6.9%
recommended allowable bias from the Westgard biological variation database
19 Standardization of Analytical Methods for the Measurement of Lipoprotein(a… 307
b c d
Fig. 19.1 Performance assessment of 29 systems measuring Lp(a) concentration using Denka
Seiken kits harmonized to the WHO/IFCC SRM-2B. Performance was evaluated on a set of 80
individual patient samples. (a) Average coefficient of variation (CV) of Lp(a) values calculated
across the 29 systems for the 80 samples sorted by increasing Lp(a) concentrations (nmol/L). (b)
Average Lp(a) concentration measured by the 29 systems as a function of the Lp(a) concentrations
determined by the Lp(a) designated comparison ELISA method. Black line is the unity line; slope
and 95% confidence interval are indicated on the graph. (c) Average relative difference to the des-
ignated comparison ELISA across the 29 systems for each sample as a function of the assigned
Lp(a) value measured by the designated comparison ELISA. Black dotted line is the 6.9% desir-
able bias recommended by the Westgard biological variation database. (d) Average relative differ-
ence to the designated comparison ELISA as a function of the predominant Lp(a) isoform size.
Slope is not statistically different from zero (green full line)
(Fraser 2022), with the exception of 8 samples with low Lp(a) concentration.
Finally, evaluation of the mean relative bias as a function of apo(a) isoforms con-
firmed that the Denka Seiken-based assays were minimally affected by systematic
accuracy errors caused by different Lp(a) isoform sizes present in the test samples
(Fig. 19.1d).
The availability of optimized assays for measuring Lp(a) with calibrators trace-
able to the WHO/IFCC SRM-2B reference material and with analytical perfor-
mances monitored by the NWRL had a very beneficial effect on Lp(a) research:
wide use and acceptance of common expression of Lp(a) values in nmol/L, compa-
rability of data obtained in different laboratories and studies, and establishment of
reliable risk thresholds for Lp(a) as a clinical biomarker of increased CVD risk
(Cegla et al. 2019; Patel et al. 2021). Establishing traceability of Lp(a) measure-
ments to the WHO/IFCC SRM-2B through the NWRL validation process provided
consistent harmonization of a good number of Lp(a) assays, thus ensuring a suitable
degree of comparability across traceable methods. Unfortunately, since the closure
in 2020 of the NWRL by the University of Washington, the ELISA-designated com-
parison method and the Lp(a) harmonization protocol are no longer available, effec-
tively ending decades of harmonization efforts.
308 N. Clouet-Foraison et al.
However, in 2018, the IFCC formed a new working group for the standardization
of Lp(a) assays with the intent to develop a higher-order reference measurement
procedure to establish full standardization of clinical methods for measuring seven
apolipoproteins including apo(a) (Ruhaak and Cobbaert 2020; Cobbaert et al. 2020).
In parallel, a new method, based on the quantification of apo(a)-specific peptides by
liquid chromatography-tandem mass spectrometry, has been developed and pro-
posed as a candidate reference method for Lp(a) standardization (Marcovina
et al. 2021).
multiple approaches have been investigated that entail various forms of recombinant
protein fragments that include proteolytic digestion sites which produce the SIL
internal peptide standards such as protein fragments or cleavable labelled peptides
(Kulyyassov et al. 2021; Villanueva et al. 2014; Oeckl et al. 2018). While these are
generally short and easy to characterize to the required level, these strategies are
based on the assumption that digestion is comparable to that of the endogenous
protein, a prerequisite for accurate absolute quantification. However, the local envi-
ronment of each proteolytic cleavage site, the higher-order structure of the con-
structs, and their post-translational modifications differ from that of the endogenous
protein which can significantly affect the proteolytic kinetics.
To achieve accurate and reproducible absolute quantification, the strategy called
double isotope dilution (ID) is considered as the gold standard (Hoofnagle et al.
2020). In this approach, the SIL peptides (or protein) are spiked into the analyzed
samples and into an external calibration curve constructed of the pure recombinant
protein or pure proteotypic peptide (Villanueva et al. 2014; Shuford et al. 2017;
Bunk and Lowenthal 2012). It was used in several higher-order reference measure-
ment procedures for small molecules or large peptides like hemoglobin A1c and
C-peptide (Hoofnagle et al. 2020) and has also been used for the absolute quantifi-
cation of larger proteins (Cobbaert et al. 2020; Huynh et al. 2021; Neubert et al.
2020; Jin et al. 2019; Dittrich et al. 2018; Sabbagh et al. 2016).
higher-order reference method for amino acid quantification certified by the NIST
(Lowenthal et al. 2010). The size of the recombinant apo(a) (14 kringles) was con-
firmed by agarose gel electrophoresis, and purity of the preparation was verified by
SDS-PAGE electrophoresis (sodium dodecyl-sulfate polyacrylamide gel electro-
phoresis), Electrospray Differential Ion Mobility Analysis, and anion exchange Fast
Protein Liquid Chromatography (Marcovina et al. 2021). This first intention assess-
ment did not indicate the presence of significant impurities in the preparation.
However, for a material to be considered as a primary reference material, its purity
should be thoroughly assessed and certified, which is a significant challenge for a
full-length protein (Stoppacher et al. 2015; Josephs et al. 2019). Indeed, the sheer
complexity of a full-length protein in terms of post-translational modifications and
glycosylation patterns, particularly for apo(a), and the considerable size of the pro-
tein (200–>800 kDa) present major obstacle to the use of most reference methods to
assess purity (Stoppacher et al. 2015; Josephs et al. 2019). Consequently, even
though the purity of the material reported by Marcovina and colleagues appears
satisfactory, assigning a certified purity, and therefore a certified concentration, is
still needed to propose this material as candidate primary reference material for
apo(a) standardization.
In this published method, the authors used a double isotope dilution strategy
involving both SIL peptides and the pure recombinant apo(a) for calibration
(Marcovina et al. 2021) with an external six-point calibration curve constructed in a
blank human serum. Each calibrator level and all samples were spiked with a mix-
ture of the pure SIL peptides corresponding to the proteotypic peptides of interest.
No sample clean-up or pre-concentration were included in this protocol, allowing to
limit losses that could impact assay accuracy. The authors investigated six candidate
quantification peptides of which three were validated for quantification of apo(a).
Because the method was proposed as a candidate reference method, it underwent
a thorough validation. Linearity, limits of quantification, intermediate precision,
reproducibility, and digestion kinetics were assessed for all measured peptides
(Marcovina et al. 2021). To confirm that the recombinant apo(a) calibrator behaved
similarly to endogenous apo(a), parallelism was verified, and no significant differ-
ences were found between the endogenous protein and the recombinant apo(a) cali-
brator. As first intent, the method was also transferred to a high-throughput LC-MS/
MS in a clinical laboratory, and high degree of agreement was achieved (Marcovina
et al. 2021). The authors additionally evaluated accuracy of the method on a set of
64 individual samples with a wide Lp(a) concentration range and varying isoform
sizes. Comparison of Lp(a) values with those obtained by the designated ELISA
method comparison (Marcovina et al. 1995) showed a Pearson correlation r2 = 0.958.
The Bland–Altman difference plot indicated minimal differences of LC-MS/MS
values compared to ELISA with a 1.7 nmol/L mean difference (2.5%) {1.96 × SD
limits of agreement −29.8 to 33.2 nmol/L} (Marcovina et al. 2021). The measure-
ment of the WHO/IFCC SRM-2B secondary reference material produced a value of
104.7 ± 8.4 nmol/L, in close agreement with its assigned reference value of
107.1 ± 8.6 nmol/L (Dati et al. 2004). Even though this does not represent a metro-
logically sound assessment of the method’s accuracy, it is a first indication that the
19 Standardization of Analytical Methods for the Measurement of Lipoprotein(a… 311
Conclusions
In this chapter, we first presented and discussed the rationale for method standard-
ization and its necessity for clinical biomarkers. We then described the challenges
associated with the measurement of Lp(a), the past standardization efforts, and the
subsequent implementation of a calibration protocol spearheaded by the NWRL to
verify that a common calibration traceable to the WHO/IFCC reference material
SRM-2B results in harmonized Lp(a) results in patient samples in selected methods.
We then presented and discussed the implementation of a new higher-order refer-
ence measurement procedure for Lp(a) using LC-MS/MS and the different strate-
gies envisioned for this new approach. Finally, we provided a critical discussion on
the feasibility of Lp(a) standardization and its practical implementation in routine
clinical laboratories in contrast to what we believe is more urgently needed, which
is among-method comparability and reliability of Lp(a) measurements.
There are still many obstacles to overcome and several technical challenges to
solve such as developing and implementing an accurate reference measurement pro-
cedure, producing in-matrix commutable serum reference materials, and imple-
menting the whole scheme in clinical practice. The experience of the IFCC working
group on the initial standardization efforts of Lp(a) highlighted the complexity of
this process for Lp(a) and clearly defined the limitations of the immunochemical
methods available for its measurement indicating the need to compromise between
19 Standardization of Analytical Methods for the Measurement of Lipoprotein(a… 319
what are the goals of standardization and what is possible to achieve with the exist-
ing methods.
Implementing a new traceability chain is a long and arduous process that will
require significant international collaboration and efforts from metrology institutes
and regulatory institutions, scientific and clinical communities, assay manufactur-
ers, and routine clinical laboratories.
Meanwhile, while the new standardization procedure is being developed, a paral-
lel activity should be implemented to continue the verification of the manufacturers’
calibration process and the comparability of Lp(a) results previously performed by
the NWRL, with the ultimate goal to transition from harmonization to
standardization.
References
Albers JJ, Marcovina SM. Standardization of Lp(a) measurements. Chem Phys Lipids.
1994;67(68):257–63.
Armbruster D, Miller RR. The Joint Committee for Traceability in Laboratory Medicine
(JCTLM): a global approach to promote the standardisation of clinical laboratory test results.
Clin Biochem Rev. 2007;28:105–13.
Blanchard V, Garcon D, Jaunet C, Chemello K, Billon-Crossouard S, Aguesse A, et al. A high-
throughput mass spectrometry—based assay for large-scale profiling of circulating human apo-
lipoproteins. J Lipid Res. 2020;33:1–36.
Bunk DM, Lowenthal MS. Isotope dilution liquid chromatography-tandem mass spectrometry for
quantitative amino acid analysis. Methods Mol Biol. 2012;828:29–38.
Cegla J, Neely RDG, France M, Ferns G, Byrne CD, Halcox J, et al. HEART UK consensus state-
ment on lipoprotein(a): a call to action. Atherosclerosis. 2019;291:62–70.
Clouet-Foraison N, Gillery P, Delatour V. Letter to the Editor regarding “Achieving comparability
with IFCC reference method for the measurement of hemoglobin A1c by use of an improved
isotope-dilution mass spectrometry method”. Anal Bioanal Chem. 2017;409:5789–90.
Clouet-Foraison N, Marcovina SM, Guerra E, Aarsand AK, Coşkun A, Díaz-Garzón J, et al.
Analytical performance specifications for lipoprotein(a), apolipoprotein B-100, and apoli-
poprotein A-I using the biological variation model in the EuBIVAS population. Clin Chem.
2020;66:727–36.
Cobbaert CM, Althaus H, Begcevic Brkovic I, Ceglarek U, Coassin S, Delatour V, et al. Towards
an SI-traceable reference measurement system for seven serum apolipoproteins using bottom-
up quantitative proteomics: conceptual approach enabled by cross-disciplinary/cross-sector
collaboration. Clin Chem. 2020;12:1–12.
Dati F, Tate JR, Marcovina SM, Steinmetz A, International Federation of Clinical Chemistry and
Laboratory Medicine; IFCC Working Group for Lipoprotein(a) Assay Standardization, et al.
First WHO/IFCC international reference reagent for lipoprotein(a) for immunoassay—Lp(a)
SRM 2B. Clin Chem Lab Med. 2004;42:670–6.
Deconinck A, Morra S, Glassée N, van de Borne P. Value of repeated measurements of
lipoprotein(a) to assess cardiovascular risk: a retrospective study. Acta Cardiol. 2022;0:1–7.
Dittrich J, Adam M, Maas H, Hecht M, Reinicke M, Ruhaak LR, et al. Targeted on-line SPE-
LC-MS/MS assay for the quantitation of 12 apolipoproteins from human blood. Proteomics.
2018;18:1–10.
Enkhmaa B, Anuurad E, Berglund L. Lipoprotein(a): impact by ethnicity and environmental and
medical conditions. J Lipid Res. 2016;57:1111–25.
320 N. Clouet-Foraison et al.
Fraser CG. Biological variation data for setting quality specifications—Westgard QC [Internet].
2022. https://www.westgard.com/guest12.htm. Accessed 13 Apr 2022.
Gabel BR, Koschinsky ML. Analysis of the proteolytic activity of a recombinant form of
apolipoprotein(a). Biochemistry. 1995;34:15777–84.
Garnotel R, Monier F, Lefèvre F, Gillery P. Long-term variability of serum lipoprotein(a) concen-
trations in healthy fertile women. Clin Chem Lab Med. 1998;36:317–21.
Gonen A, Yang X, Yeang C, Alekseeva E, Koschinsky M, Witztum JL, et al. Generation and
characterization of LPA-KIV9, a murine monoclonal antibody binding a single site on
apolipoprotein(a). J Lipid Res. 2020;61:1263–70.
Hoerger TJ, Wittenborn JS, Young W. A cost-benefit analysis of lipid standardization in the United
States. Prev Chronic Dis. 2011;8:A136.
Hoofnagle AN, Wener MH. The fundamental flaws of immunoassays and potential solutions using
tandem mass spectrometry. J Immunol Methods. 2009;347:3–11.
Hoofnagle AN, Whiteaker JR, Carr SA, Kuhn E, Liu T, Massoni SA, et al. Recommendations for
the generation, quantification, storage, and handling of peptides used for mass spectrometry-
based assays. Clin Chem. 2016;62:48–69.
Hoofnagle AN, Cobbaert CM, Delatour V, Kelleher NL, Lowenthal MS, Shuford CM. Should
LC-MS/MS be the reference measurement procedure to determine protein concentrations in
human samples? Clin Chem. 2020;6:1–6.
Huynh HH, Boeuf A, Derbez-Morin M, Dupuy A-M, Lalere B, Delatour V, et al. Development
of an antibody-free ID-LC MS methods for the quantification of procalcitonin in human
serum at sub-microgram per liter level using a peptide-based calibration. Anal Bioanal Chem.
2021;413:4917–9.
International Organization for Standardization. ISO 17511:2020—In vitro diagnostic medical
devices—Requirements for establishing metrological traceability of values assigned to cali-
brators, trueness control materials and human samples [Internet]. 2020. https://www.iso.org/
standard/69984.html. Accessed 13 Apr 2022.
Jin Z, Collier TS, Dai DL, Chen V, Hollander Z, Ng RT, et al. Development and validation of apo-
lipoprotein AI-associated lipoprotein proteome panel for the prediction of cholesterol efflux
capacity and coronary artery disease. Clin Chem. 2019;65:282–90.
Joint Committee for Guides in Metrology (JCGM). JCGM 200: 2012 International vocabu-
lary of metrology (VIM). Basic and general concepts and associated terms. 3rd ed. Geneva:
International Organization Standards; 2012.
Josephs RD, Martos G, Li M, Wu L, Melanson JE, Quaglia M, et al. Establishment of measure-
ment traceability for peptide and protein quantification through rigorous purity assessment—a
review. Metrologia. 2019;56:044006.
Kamstrup PR. Lipoprotein(a) and cardiovascular disease. Clin Chem. 2021;166:154–66.
Karwatowska-Prokopczuk E, Clouet-Foraison N, Xia S, Viney NJ, Witztum JL, Marcovina SM,
et al. Prevalence and influence of LPA gene variants and isoform size on the Lp(a)-lowering
effect of pelacarsen. Atherosclerosis. 2021;324:102–8.
Koschinsky ML, Marcovina SM. Structure-function relationships in apolipoprotein(a): insights
into lipoprotein(a) assembly and pathogenicity. Curr Opin Lipidol. 2004;15:167–74.
Koschinsky ML, Tomlinson JE, Zioncheck TF, Schwartz K, Eaton DL, Lawn RM.
Apolipoprotein(a): expression and characterization of a recombinant form of the protein in
mammalian cells. Biochemistry. 1991;30:5044–51.
Kostner KM, Kostner GM, Wierzbicki AS. Is Lp(a) ready for prime time use in the clinic? A pros-
and-cons debate. Atherosclerosis. 2018;274:16–22.
Kronenberg F. Human genetics and the causal role of lipoprotein(a) for various diseases. Cardiovasc
Drugs Ther. 2016;30:87–100.
Kronenberg F. Prediction of cardiovascular risk by Lp(a) concentrations or genetic variants within
the LPA gene region. Clin Res Cardiol Suppl. 2019;14:5–12.
Kronenberg F, Tsimikas S. The challenges of measuring Lp(a): a fight against hydra?
Atherosclerosis. 2019;289:181–3.
19 Standardization of Analytical Methods for the Measurement of Lipoprotein(a… 321
Nordestgaard BG, Chapman MJ, Ray K, Borén J, Andreotti F, Watts GF, et al. Lipoprotein(a) as a
cardiovascular risk factor: current status. Eur Heart J. 2010;31:2844–53.
Oeckl P, Steinacker P, Otto M. Comparison of internal standard approaches for SRM analysis of
alpha-synuclein in cerebrospinal fluid. J Proteome Res. 2018;17:516–23.
Patel AP, Wang M, Pirruccello JP, Ellinor PT, Ng K, Kathiresan S, et al. Lp(a) (Lipoprotein[a])
concentrations and incident atherosclerotic cardiovascular disease: new insights from a large
national biobank. Arterioscler Thromb Vasc Biol. 2021;41:465–74.
Plebani M. Errors in clinical laboratories or errors in laboratory medicine? Clin Chem Lab Med.
2006;44:750–9.
Reyes-Soffer G, Ginsberg HN, Berglund L, Duell PB, Heffron SP, Kamstrup PR, et al.
Lipoprotein(a): a genetically determined, causal, and prevalent risk factor for atheroscle-
rotic cardiovascular disease. A scientific statement from the American Heart Association.
Arterioscler Thromb Vasc Biol. 2021;41:48–60.
Ruhaak LR, Cobbaert CM. Quantifying apolipoprotein(a) in the era of proteoforms and precision
medicine. Clin Chim Acta. 2020;511:260–8.
Ruhaak LR, Smit NP, Romijn FP, Pieterse MM, Van Der Laarse A, Van Der Burgt YE, et al. Robust
and accurate 2-year performance of a quantitative mass spectrometry-based apolipoprotein test
in a clinical chemistry laboratory. Clin Chem. 2018;64:747–9.
Sabbagh B, Mindt S, Neumaier M, Findeisen P. Clinical applications of MS-based protein quanti-
fication. Proteom Clin Appl Appl. 2016;10:323–45.
Scharnagl H, Stojakovic T, Dieplinger B, Dieplinger H, Erhart G, Kostner GM, et al. Comparison
of lipoprotein(a) serum concentrations measured by six commercially available immunoas-
says. Atherosclerosis. 2019;289:206–13.
Schmidt K, Noureen A, Kronenberg FG. Structure, function, and genetics of lipoprotein(a). J Lipid
Res. 2016;57:1339–59.
Shuford CM, Walters JJ, Holland PM, Sreenivasan U, Askari N, Ray K, et al. Absolute protein quan-
tification by mass spectrometry: not as simple as advertised. Anal Chem. 2017;89:7406–15.
Stefanutti C, Pisciotta L, Favari E, Di Giacomo S, Vacondio F, Zenti MG, et al. Lipoprotein(a) con-
centration, genetic variants, apo(a) isoform size, and cellular cholesterol efflux in patients with
elevated Lp(a) and coronary heart disease submitted or not to lipoprotein apheresis: an Italian
case-control multicenter study on Lp(a). J Clin Lipidol. 2020;14:487–497.e1.
Stoppacher N, Josephs RD, Daireaux A, Choteau T, Westwood S, Wielgosz RI. Accurate quan-
tification of impurities in pure peptide material—angiotensin I: comparison of calibration
requirements and method performance characteristics of liquid chromatography coupled to
hybrid tandem mass spectrometry and linear ion trap high-resolution mass spectrometry. Rapid
Commun Mass Spectrom. 2015;29:1651–60.
Tate JR, Rifai N, Berg K, Couderc R, Dati F, Kostner GM, et al. International Federation of Clinical
Chemistry standardization project for the measurement of lipoprotein(a). Phase I. Evaluation
of the analytical performance of lipoprotein(a) assay systems and commercial calibrators. Clin
Chem. 1998;44:1629–40.
Tate JR, Berg K, Couderc R, Dati F, Kostner GM, Marcovina SM, et al. International Federation of
Clinical Chemistry and Laboratory Medicine (IFCC) standardization project for the measure-
ment of lipoprotein(a). Phase 2: selection and properties of a proposed secondary reference
material for lipoprotein(a). Clin Chem Lab Med. 1999;37:949–58.
Trinder M, Paruchuri K, Haidermota S, Bernardo R, Zekavat SM, Gilliland T, et al. Repeat
measures of lipoprotein(a) molar concentration and cardiovascular risk. J Am Coll Cardiol.
2022;79:617–28.
Tsimikas S. A test in context: lipoprotein(a). Diagnosis, prognosis, controversies, and emerging
therapies. J Am Coll Cardiol. 2017;69:692–711.
Van Den Broek I, Romijn FP, Nouta J, Van Der Laarse A, Drijfhout JW, Smit NPM, et al. Automated
multiplex LC-MS/MS assay for quantifying serum apolipoproteins A-I, B, C-I, C-II, C-III, and
E with qualitative apolipoprotein E phenotyping. Clin Chem. 2016;62:188–97.
van den Broek I, Sobhani K, Van Eyk JE. Advances in quantifying apolipoproteins using LC-MS/
MS technology: implications for the clinic. Expert Rev Proteom. 2017;14:869–80.
19 Standardization of Analytical Methods for the Measurement of Lipoprotein(a… 323
Van Der Valk FM, Bekkering S, Kroon J, Yeang C, Van Den Bossche J, Van Buul JD, et al.
Oxidized phospholipids on lipoprotein(a) elicit arterial wall inflammation and an inflammatory
monocyte response in humans. Circulation. 2016;134:611–24.
Villanueva J, Carrascal M, Abian J. Isotope dilution mass spectrometry for absolute quantification
in proteomics: concepts and strategies. J Proteome. 2014;96:184–99.
Watts GF, Boffa MB. Lipoprotein(a): lodestar for future clinical trials. Lancet. 2018;392:1281–2.
Wilson DP, Jacobson TA, Jones PH, Koschinsky ML, McNeal CJ, Nordestgaard BG, et al. Use
of lipoprotein(a) in clinical practice: a biomarker whose time has come. A scientific statement
from the National Lipid Association. J Clin Lipidol. 2019;13:374–92.
Wyness SP, Genzen JR. Performance evaluation of five lipoprotein(a) immunoassays on the Roche
Cobas c501 chemistry analyzer. Pract Lab Med. 2021;25:e00218.
Chapter 20
On the Way to a Next-Generation Lp(a)
Reference Measurement System Based
on Quantitative Protein Mass
Spectrometry and Molar Units
Abbreviations
Introduction
In order for Lp(a) test results to be comparable in time and space across the globe,
we need traceability of test results. Traceability is the ability to trace, for example,
the origin of a product, the ancestors of an individual, or the absolute value of a test
result. The word “traceability” comes from the Latin verb trahere: to draw.
Traceability can refer to documentation such as sampling procedures, laboratory
methods, lab processes, etc., but as in ISO/IEC 17025, we are dealing with trace-
ability of medical test results. It is key that test results are traceable to endorsed
metrological references. Metrology refers to the science of measurement. In the
case of medical test results, we use the wording metrological traceability. The
current VIM definition of metrological traceability is property of a measurement
result whereby the result can be related to a reference through a documented unbro-
ken chain of calibrations each contributing to the measurement uncertainty (JCGM
2012). Ideally, the references are international standards that mimic the measurand
of interest and have assigned values expressed in the International System of Units
(SI units). For temperature and many other physical quantities, for example, mass
and time traceability is relatively easily established. Also, in forensic toxicology or
chemistry, the working standards are substances with defined purity and solutions of
pure substances. Yet, in laboratory medicine, metrological traceability of protein
test results measured in body fluids within total allowable error (Westgard QC 2014;
Fraser 2001; EFLM 2019) is an enormous challenge due to the huge interindividual
variability of endogenous measurands and the complexity of the human body fluid
matrix in which the measurands are dissolved.
Proteins are the primary effectors in human biology systems. Hence, complete
knowledge of their structure and biological function is fundamental for understand-
ing their potential role as promising biomarkers and/or future medical tests. Proteins
from a single gene can vary widely in their amino acid sequence, and posttransla-
tional modifications also give rise to a variety of proteoforms. As it is now recog-
nized that the variation at the protein level is functionally relevant and much larger
than the variation at the genetic level, the Human Proteoform Project has recently
been launched. This ambitious initiative aims to define the human proteome by
generating a reference set of proteoforms produced from the ~20,000 genes encoded
20 On the Way to a Next-Generation Lp(a) Reference Measurement System Based… 327
in the human genome. The human proteome is the set of all proteoforms expressed
by humans. The underlying rationale is that proteoform-level knowledge is essential
to understand biological function of proteins. Unraveling the human proteome will
lead to a more refined, molecular definition of human health and disease.
Conventional immunoassays that measure human proteins are generally blinded to
the underlying proteoforms of the protein of interest and hence do not recognize or
differentiate dysfunctional from functional proteoforms. This may of course nega-
tively impact patient management and patient outcome. Enabling technology is
needed to detect proteoforms at the molecular level. The Consortium for Top-Down
Proteomics that runs the Human Proteome Initiative uses such technology (Smith
et al. 2021).
Clinicians and laboratory professionals should be aware that improved technol-
ogy is instrumental to advance science and the science of measurement. In order to
enable accurate apolipoprotein quantitation, protein methods should specifically
measure the measurand intended to be measured rather than an ill-defined mixture
of proteoforms with different biological functions. This implies that in this era of
precision medicine, we should introduce higher-order measurement procedures that
can recognize molecular forms, that is, proteoforms, of interest. To that end, triple-
quadrupole mass spectrometry-based technology has built up a reputation as a
higher-order reference measurement procedure for direct, immunoassay-indepen-
dent protein measurement in complex mixtures.
In this chapter, the authors clarify the current state-of-the-science around Lp(a)
measurement, with special attention to the unique structural characteristics of the
highly polymorphic apolipoprotein(a) [apo(a)] in Lp(a). The apo(a) features should
be considered during Lp(a) test standardization, in order to guarantee that commer-
cial Lp(a) tests are fit for clinical purpose and produce accurate results within allow-
able limits of uncertainty. To accomplish this, a reference measurement system that
produces test results traceable to the SI units is needed. The rationale for the devel-
opment of a higher-order reference measurement system and for transitioning from
the former WHO-IFCC immunoassay-based reference measurement system for
Lp(a) into a more robust immunoassay-independent mass spectrometry-based refer-
ence measurement system and molar units is pointed out.
The Lp(a) particle is the most complex and polymorphic of all serum lipoproteins.
Lp(a) was discovered by Kare Berg in 1963 and is only present in humans and the
hedgehog and not in other mammals. Lp(a) is an apoB100-containing LDL-like
particle that is rich in cholesterol and is associated with a unique hydrophilic, highly
328 C. Cobbaert et al.
glycosylated second major protein, that is, the apo(a). Apo(a) is covalently attached
to apoB in Lp(a), shows ~80% amino acid homology with plasminogen, and con-
tains multiple copies (3–> 40) of plasminogen-like kringle IV type 2 (K-IV2). This
peculiarity of apo(a) is known as apo(a)-size polymorphism. Beyond apo(a)-size
polymorphism, other prominent determinants of Lp(a) levels are genetic variants
such as K-IV2 4925G>A and K-IV2 4733G>A (Schachtl-Riess et al. 2021). Although
apo(a) has structural homology to plasminogen, it lacks fibrinolytic activity. As a
consequence of its composite structure, Lp(a) can trigger prothrombotic and antifi-
brinolytic actions favoring clot stability as well as atherosclerosis progression via its
tendency for retention in the arterial intima, with deposition of its cholesterol load
at sites of plaque formation. In addition, Lp(a) can induce inflammation and calcifi-
cation in the aortic leaflet valve interstitium, leading to calcific aortic valve stenosis.
Recent epidemiological and genetic evidence support the proposition that elevated
concentrations of Lp(a) are causally related to atherothrombotic risk and calcific
aortic valve stenosis.
The concentration of Lp(a) in blood is largely determined by genetic factors and
hardly influenced by diet or lifestyle conditions. After reaching adulthood, the con-
centration remains constant over the lifetime of an individual. Remarkably, the
interindividual variation is large as blood Lp(a) levels among individuals vary up to
1000-fold. Its frequency distribution differs across populations, being skewed to the
right in Caucasians and displaying a more Gaussian distribution in Blacks. Its meta-
bolic role is after 60 years of intense research still not understood. As Lp(a) is an
independent genetic risk factor which accelerates cardiovascular disease (CVD)
through pro-atherogenic, pro-thrombogenic, and proinflammatory mechanisms, the
downstream consequences for patients are as devastating as in the case of untreated
patients with heterozygous familial hypercholesterolemia (Reyes-Soffer et al. 2022;
Kronenberg and Tsimikas 2019). Consequently, the pharmaceutical industry has
invested in the development of novel therapies for Lp(a) lowering that target hepatic
synthesis of apo(a). These therapies are in various phases of clinical trials, and the
completion of these studies will provide critical insight into the cardiovascular ben-
efits of Lp(a) lowering.
to be measured. Secondly, the analyte being measured in the patient sample should
have the same structural characteristics as the analyte in the assay calibrator(s) to
achieve the same degree of immunoreactivity per Lp(a) particle. Thirdly, an
accuracy-based target value should be assigned to the assay calibrator(s) using an
internationally recognized reference measurement system to guarantee consistency
and comparability of results. Fourthly, common protocols should be used for trans-
ferring an accurate value from the reference material to the assay calibrators. Most
commercial Lp(a) tests express Lp(a) in mass units [in mg/L or mg/dL Lp(a) mass]
and make assumptions and oversimplifications to that end, which all confound
Lp(a) levels (Ruhaak and Cobbaert 2020). Few routine tests express Lp(a) levels in
molar units (nmol/L).
Most routine Lp(a) tests with mass units use multiple calibrators that are inter-
nally prepared by the IVD-manufacturer and produce results that are not traceable
to a common internationally accepted standard. The few routine Lp(a) tests with
molar units were—until recently—calibrated by the WHO-IFCC Lp(a) Reference
Measurement System developed at the Northwest Lipid Metabolism and Diabetes
Research Laboratories (NLMDRL), Washington DC, USA (Tsimikas et al. 2018).
That reference measurement procedure (RMP) is a reference ELISA that uses
monoclonal K-IV2-independent antibodies both for capturing and detecting intact
Lp(a). The RMP was calibrated with a matrix-based WHO-IFCC Reference Material
named SRM2B. This reference material was value-assigned based on its apo(a)
protein content and expressed in nmol/L, thus reflecting the number of Lp(a)
particles.
The results of a correlation study performed in 2020 (Dikaios et al. 2023) have
shown that expressing Lp(a) concentrations in molar units traceable to the SRM2B
largely improves the intermethod variability (Fig. 20.1). Twenty-one human serum
samples were analyzed with six methods. Three of the six methods provided the
results in both molar and mass concentration units (Roche, Diasys and Sentinel).
For the mass units, the average coefficient of variation (CV) between the methods
was 12.8% (range: 8.4–20.5%) for all six methods and 9.7% (range: 3.4–21.8%) for
the methods from Roche, Diasys, and Sentinel. For the results in molar units, the
intermethod CV of these same three methods was only 3.1% (range: 1.3–5.1%)
(Tables 20.1 and 20.2). These results also clearly indicate that commercial assays
which are based on one serially diluted calibrator will underestimate the Lp(a) con-
centration in serum samples with small apo(a) isoforms.
Notwithstanding the availability of SRM2B since 2003, most commercial
Lp(a) assays still use mass units. Conservatism and ignorance together with the
limitations of the WHO-IFCC Lp(a) Reference Measurement System have pre-
vented worldwide implementation of Lp(a) results in molar units. The important
limitation was the fact that SRM2B could not be used directly by the test manufac-
turer as part of their internal traceability procedures to value-assign their product
calibrators.
330 C. Cobbaert et al.
Fig. 20.1 Intermethod variability among six commercial immunoassay-based assays for the quan-
tification Lp(a) concentration in mass (a) and molar units (b). This study was performed in 2020
(Dikaios et al. 2023). Twenty-one human serum samples were analyzed, and the average results
from three replicate measurements in one run are shown in the graphs. The apo(a) isoforms present
in the serum samples were determined by Western blot at the Institute of Genetic Epidemiology,
Medical university of Innsbruck, Austria. All assays used a multipoint calibration curve, and for
five of the six assays, the curve consisted of independent calibrator solutions. Only for the Siemens
Healthineers N latex method one serially diluted calibrator was used. According to the information
provided by the assay manufacturers, the values expressed in mass units were traceable to internal
standards, while the values in molar units were traceable to the WHO-IFCC Reference Material
SRM2B. The inserts demonstrate that at an Lp(a) mass level of 40–50 mg/dL, the relative differ-
ence between the highest and the lowest method result is 22% for the sample with average molecu-
lar weight apo(a) isoforms (28 and 31 K-IV) rising to 37% in a sample with small apo(a) isoforms
(homozygous for 20 K-IV). In the same specimens, Lp(a) levels expressed in molar units showed
an relative difference between the highest and the lowest method result ranging between 5.7 and
8.7%, that is, three to fourfold lower. The larger intermethod variability between the methods in
mass units can be explained by the fact that these test results are traceable to different internal
manufacturer’s standards and by the fact that different calibration approaches are used, that is, five
independent calibrators with specific apo(a) isoforms per calibrator level versus serial dilutions
from one master calibrator solution. The variable protein/lipid content of Lp(a) particles also
brings along confounded Lp(a) measurements
Table 20.1 The measurement results obtained in 2020 by measuring 21 human serum samples with 6 commercial immunoassay-based assays for the
20
(continued)
Table 20.1 (continued)
332
curve with independent calibrator solutions. According to the information provided by the assay manufacturers, the values in molar units were traceable to the
WHO-IFCC Reference Material SRM2B. The average intermethod CV over the 21 samples was 6.0%
334 C. Cobbaert et al.
In the 1980s and 1990s of the twentieth century, many research groups were inter-
ested in Lp(a) studies and its implementation in patient care. Unfortunately, a turn-
point came after the publication of negative findings from the Physician Health
Study in JAMA (Journal of the American Medical Association) which demonstrated
no association between Lp(a) levels and the risk of future MI (myocardial infarc-
tion), stroke, or peripheral vascular disease (Ridker 1995; Ridker et al. 1993, 2001).
Fortunately, Rifai et al. reanalyzed the specimens from the Physician Health Study
with the gold standard reference ELISA and found that median Lp(a) in cases were
significantly higher than in controls with reference ELISA, whereas no significant
difference was found between cases and controls when using a commercial immu-
nonephelometric assay (Rifai et al. 2004). It was revealed that Lp(a) test inaccuracy
in the immunonephelometric assay had obscured the true relationship between
Lp(a) levels and CVD in the former JAMA papers. It is very essential to have accu-
rate Lp(a) tests that are not confounded by apo(a)-size polymorphism. In Fig. 20.2,
the masking impact of apo(a)-size polymorphism on Lp(a) levels is visualized in
case of isoform-dependent Lp(a) tests using a one-point calibration strategy
(Tsimikas et al. 2018). The attenuating effect of apo(a)-size polymorphism occurs
irrespective of the use of mass or molar units.
In the last years, the situation has improved as current commercially available
Lp(a) tests make use of multiple independent calibrators, well spread across the
Lp(a) concentration range and representing different apo(a) isoforms. This calibra-
tion strategy enables IVD manufacturers to measure Lp(a) with reduced impact of
apo(a)-size polymorphism (Tsimikas et al. 2018).
250
isoform independent assay Higher Lp(a)
values are
200 underestimated
by isoform
dependent
Lower Lp(a) assays
Lp(a) Value
50
0
0 40 35 30 25 20 15
Lp(a) Isoform Size
Fig. 20.2 Theoretical relationship of Lp(a) values according to isoform size in isoform-
independent and isoform-dependent assays in case of a one-point calibration strategy. Lp(a) values
are inversely related to isoform size, with large isoforms being associated with lower levels and
vice versa. Irrespective of the expression of Lp(a) values (nmol/L or mg/dL), isoform-dependent
assays will tend to overestimate low values and underestimate high values. Lp(a) lipoprotein(a).
[Reprinted with permission from (Tsimikas et al. 2018)]
20 On the Way to a Next-Generation Lp(a) Reference Measurement System Based… 335
The determinants of variability in Lp(a) particle composition are multiple and were
theoretically modeled based on existing literature (Ruhaak and Cobbaert 2020)
(Fig. 20.3). To that end and beyond the effect of apo(a)-size polymorphism, post-
translational modifications such as N- and O-glycosylations in apo(a) and apoB and
the lipid/protein ratio were considered. Depending on the number of K-IV2 repeats,
the theoretical protein content of the Lp(a) particle varies between 30 and 46%
(w/w), which inescapably confounds Lp(a) mass measurements. Based on variation
in number of K-IV2 repeats alone, the composition of lipid/protein in Lp(a) ranges
from 31% (w/w) in case of apo(a) with 6 K-IV2repeats to 42% (w/w) in case of
apo(a) with 35 K-IV2 repeats. This brings along a difference in Lp(a) mass of 19%:
2821 kDa Lp(a) mass in case of 6 K-IV2 repeats compared to 3344 kDa Lp(a) mass
in case of 35 K-IV2 repeats. This model also clarifies why using fixed factors for
converting Lp(a) particle mass into molar units that represent Lp(a) particle num-
ber—or vice versa—is metrologically not sound (Reyes-Soffer et al. 2022;
Guadagno et al. 2015). Therefore, it is of utmost importance to measure Lp(a) in
terms of its apo(a) component and no longer in terms of Lp(a) mass because varia-
tion in mass content can occur in each of its constituents, leading to large heteroge-
neity among individuals and confounded Lp(a) mass values. Because each Lp(a)
particle carries one molecule of apo(a), the molar apo(a) concentration reflects
unequivocally the number of Lp(a) particles.
In the Netherlands, the Dutch External Quality Assessment (EQA) organizer, named
SKML, performs national Lp(a) surveys using frozen, native human sera. In
Fig. 20.4, the national EQA data from 2018 are displayed. In total, 17 accredited
laboratories participated with nearly complete data sets, which comprised 11 rounds
of 2 blinded samples each. EQA samples were analyzed with two weekly intervals.
Each of the blinded samples was included twice in the EQA survey, that is, in the
first, respectively, and in the second half of the calendar year, to evaluate indepen-
dent duplicates within a year. A scatterplot of Lp(a) mass results (in mg/L) produced
by individual labs and stratified by IVD manufacturer is presented. Different IVD
manufacturers are marked with specific colored symbols, whereas the all-lab total
mean (ALTM) is presented with a black horizontal stripe. The overall interlabora-
tory variation of Lp(a) mass tests ranges from 16.4 to 32.1% at Lp(a) mass levels of
~150–450 mg/L. Siemens Healthineers demonstrates a negative bias whereas
Abbott and Beckman reveal a positive bias compared to the ALTM. The Roche
Lp(a) test is closest to the ALTM.
336 C. Cobbaert et al.
Particle size
6 KIV-2
repeats
35 KIV-2
repeats
PL FC CE TG apoB apo(a)
Fig. 20.3 Theoretical model of Lp(a) mass and compositional variation depending on apo(a)
K-IV2-size polymorphism based on literature. Lp(a) particle mass is dependent on the lipid/protein
composition and amount, the apo(a)-size polymorphism, and the N- and O-glycosylation of apo(a)
and apoB (upper left). Based on the variation in number of K-IV2 repeats in apo(a) alone, the dis-
tribution of lipid/protein in Lp(a) varies from 31% (w/w) protein with 6 K-IV2 repeats to 42%
(w/w) protein with 35 K-IV2repeats (upper right and bottom), leading to 19% difference in Lp(a)
mass. PL phospholipids, FC free cholesterol, CE cholesteryl esters, and TG triglycerides.
[Reprinted with permission from (Ruhaak and Cobbaert 2020)]
Taking into consideration published clinical thresholds of 150 and 300 mg/L for
Lp(a) particle mass, the EQA findings suggest serious impact of Lp(a) recovery on
CVD risk classification of patients and on patient management depending on the
IVD manufacturers’ reagents used. Yet, the systematic differences between IVD
manufacturers suggest that use of a common calibrator traceable to a generally rec-
ognized point of reference (i.e., SI unit through mass spectrometry) can signifi-
cantly improve the method variability. A more accurate assessment of each
individual’s CVD risk is also anticipated if molar Lp(a) assays rather than mass
assays become the norm, because of improved reflection of low risk in patients with
a small number of very large Lp(a) particles and high molecular weight apo(a) iso-
forms and high risk in patients with a high number of small Lp(a) particles and low
molecular weight apo(a) isoforms.
20 On the Way to a Next-Generation Lp(a) Reference Measurement System Based… 337
450
400
350
mg/L Lp(a) mass
300
250
200
150
100
50
0
1A 3B 2F 4A 2B 4C 2C 3D 1C 4E 1F 3C 2A 3F 1B 4B 1E 4F 2D 3E 2E 4D
Paired EQA-samples (NL, 2018)
Fig. 20.4 Dutch External Quality Assessment data of Lp(a) mass as measured in 11 paired,
blinded human serum samples analyzed at two weekly intervals in 2018 in 17 accredited medical
laboratories. Each of the blinded samples was included twice in the EQA survey to evaluate inde-
pendent duplicates within a year. For example, samples 1A and 3B are identical, but sample 1A
was analyzed in the first half of 2018 and sample 3B in the second half. Absolute Lp(a) levels vary
from half to double, with interlaboratory coefficients of variation ranging between 16 and 32%.
[Reprinted with permission from (Ruhaak and Cobbaert 2020)]
SI units
Measurement uncertainty
Secondary reference material
SRM-2B Secondary reference
measurement procedure
(NWLRL, Seattle, WA, USA)
Manufacturer’s master
calibrators
Manufacturer’s Standing
Measurement procedure
Commercial product
calibrators from different lots
Fig. 20.5 Metrological traceability chain as outlined in ISO 17511:2021. Currently, apo(a) test
results are at best traceable to WHO-IFCC secondary reference material SRM2B (blue), through
an ELISA-based K-IV2-independent method (green). However, the top of the traceability chain is
not in place (grey) and under development (Cobbaert et al. 2021), as described in this chapter. To
ensure SI traceability, primary reference materials (i.e., peptide-based calibrators) and a higher-
order, K-IV2-independent reference measurement procedure (MS-based cRMP) are needed.
[Reprinted with permission from (Ruhaak and Cobbaert 2020)]
Fig. 20.6 The infographic illustrates how a high-order mass spectrometry-based measurement
procedure allows for K-IV2-independent measurement of apo(a) at the molecular level, in contrast
to immunoassays which are by design confounded by the apo(a)-size polymorphism. Heterogeneous
apo(a) with variable sizes (3, 10, 20, and 30 K-IV2 repeats) are measured by immunoassay with
polyclonal antibodies, resulting in a poor definition of the apo(a) measurand and K-IV2-dependent
results, while LC-MS-based quantitation of apo(a) at the peptide level is K-IV2 independent with
high specificity of the measurand and in molar concentration
50 100
Bias (nmol/L)
Percent Bias
400
0 0
–100 –200
0 –150 –300
0 200 400 600 0 200 400 600 0 200 400 600
apo(a) (LFLEPT) (nmol/L) apo(a) (LFLEPT) (nmol/L) apo(a) (LFLEPT) (nmol/L)
Fig. 20.7 Method comparison in molar units between Roche immunoassay and an in-house-
developed immunoassay-independent higher-order MS-based method. Three-hundred sixty-five
serum leftover serum specimens from the Leiden lipid clinic were compared. Deming regression
line: Y = 0.985 X + 0.52 in nmol/L, Xmean = 75.2 nmol/L, and Ymean = 74.6 nmol/L; R = 0.984.
Outliers were confirmed in independent runs. The red arrow points to a clinical specimen that
demonstrates a pronounced discordance between the two assays: 227 nmol/L with the MS-based
test and 87 nmol/L with the Roche immunoassay. It was demonstrated with Western blotting that
apo(a) in this discordant specimen had only nine K-IV repeats (unpublished data)
observations are remarkable: substantial scatter is noted in the absolute bias plot (up
to +/− 50 nmol/L) and marked discordances may occur in rare samples, poten-
tially leading to discrepant classifications with effect on patient management.
Apo(a) isoforms have been determined in all clinical specimens, and apo(a) isoform
data corroborate the average equivalence of Lp(a) test results between MS test and
IA test as well as individual scatter in clinical specimens across all apo(a) isoform
groups (see Fig. 20.1). For personalized medicine, accurate Lp(a) test results are
expected that are not confounded by apo(a)-size polymorphism and/or by molecular
diversity arising from apo(a) variants or truncated proteins (Coassin et al. 2017).
From the above head-to-head comparison, we deduce that as immunoassays are
routinely used in clinical practice, immunoassay test results (Roche) are fit for clini-
cal purpose and equivalent within total allowable error for >99% of the specimens
but can be inaccurate in rare individual cases due to genetic variants/mutants who
go undetected.
The Centers for Disease Control and Prevention (CDC) in Atlanta, Georgia, USA,
is the custodian laboratory for the WHO Biological Reference Material for
lipoprotein(a) (i.e., IFCC SRM2B). As part of this function, it holds and distributes
342 C. Cobbaert et al.
Interim Solutions
The IFCC WG APO-MS members and CDC as envisioned future network coordi-
nator of Lp(a) calibration laboratories can help IVD manufacturers, clinical trial
laboratories, and EQA providers transitioning to the new IFCC reference system
by making available individual donor and pooled serum materials with Lp(a) val-
ues assigned by the IFCC’s LC-MS/MS method (molar units, not K-IV2 depen-
dent). Although this cRMP is still under development, data obtained with these
materials can provide information about the agreement of a laboratory method
with the MS-based method. Also, the CDC Clinical Standardization Program
(CSP) will prepare a formal standardization program for guiding manufacturers
on their path to molar (first step), respectively, SI-traceable (second step) Lp(a)
test results and certification. In preparation of this activity, CDC-CSP will be
conducting an interlaboratory comparison study for Lp(a) with routine clinical
laboratories as participants. An infographic on the transition is shown at the IFCC
website: https://www.ifcc.org/media/479001/210514_ifcc-apo-traceability_info-
graphic_def.pdf.
20 On the Way to a Next-Generation Lp(a) Reference Measurement System Based… 343
Accurate medical test results are key for safe and effective management of patients.
Moreover, exchangeability of test results, reference intervals, and decision limits
among healthcare institutions is essential for efficiency reasons along the patients’
journey in primary, secondary, and tertiary care settings. In case of inaccuracy
exceeding the total allowable error, Lp(a) test results can be misleading and harm
patients due to misclassification and undetected or untreated cardiovascular risk.
The (in)accuracy and analytical performance of a test affects its clinical perfor-
mance as both key elements of test evaluation are interdependent. On top, flawed
test results may mask the clinical utility and medical value of a test, preventing its
adoption and implementation in clinical practice. These general principles also
apply for Lp(a) testing. Nevertheless, a multitude of Lp(a) test design flaws are
causally related to about 15 years of unjustified silence around Lp(a) testing.
Currently, a revival of Lp(a) testing is taking place and its clinical relevance has
been reinvented (Ellis et al. 2017).
Firstly, the definition of the measurand intended to be measured should have been
unequivocal, but that was not the case. In the past decades, Lp(a) particle mass,
Lp(a) particle concentration, and Lp(a) cholesterol have all been considered and
used mixed. As most routine Lp(a) tests were based on an immunoassay-based read-
out, the apo(a)-size polymorphism confounded the Lp(a) recovery, especially in
case of polyclonal antibodies. But also the type and number of calibrators are rele-
vant: multiple calibrator levels, composed of well-selected apo(a) isoforms across
the Lp(a) concentration range that mimic clinical specimens, are preferred. However,
if IVD manufacturers make serial dilutions from a single calibrator, the isoform
composition of the diluted calibrators is not aligned with that in the clinical speci-
mens, and Lp(a) test results become inaccurate (Fig. 20.1). Finally, also the post-
analytical phase and unit choice (mass or molar) have further aggravated the
inaccuracy of Lp(a) test results due to assumptions and oversimplified models for
estimating the Lp(a) particle mass (Ruhaak and Cobbaert 2020) and Figs. 20.1 and
20.4). The cumulative errors of former Lp(a) kit designs often exceeded the allow-
able total error, making those Lp(a) tests not fit for clinical purpose.
344 C. Cobbaert et al.
Currently, immunoassay-based Lp(a) tests are still the methods of choice in medical
labs. Test design and analytical performance of immunoassay-based tests have been
substantially improved, especially those that are standardized against the former
IFCC-WHO reference measurement system and express Lp(a) test results in molar
units. In these tests, on average, equivalent results and correlation coefficients
>0.975 are found between immunoassays and higher-order MS-based measurement
procedures (Dikaios et al. 2023). A representative method comparison is presented
in Fig. 20.7. IVD manufacturers took lessons from the past and currently use inde-
pendent calibrators with well-selected apo(a) isoforms which are good mimics of
clinical specimens. Notable is the fact that unexplained Lp(a) scatter—within the
total allowable error zone—can be observed in the absolute/relative bias plots, also
within all predominant apo(a) isoform classes when comparing routine and higher-
order MS-based measurement procedures (data not shown). In addition, less than
1% discordances—exceeding the total allowable error—were found between immu-
noassay and higher-order MS-based measurement procedure results, pointing to
further molecular variation/truncation on top of apo(a)-size polymorphism and
affecting immunoassay-based measurements. As >99% of the results from current
immunoassays are concordant with higher-order mass spectrometry-derived results,
we can state that contemporary Lp(a) immunoassay tests are in general fit for clini-
cal purpose with a manageable risk for patient harm.
the molar Lp(a) immunoassay kits already better fit for clinical purpose while still
being standardized to the former WHO-IFCC RMS. A second step can be made by
IVD manufacturers once the SI-traceable MS-based RMS is in place and interna-
tionally recognized. To guide IVD manufacturers toward sustainable standardiza-
tion of Lp(a) tests with SI traceability of Lp(a) test results, commutable,
value-assigned secondary RMs will be made available by JRC which IVD manufac-
turers can purchase for internal standardization.
The calibration laboratories running the SI-traceable cRMP together with JRC,
LNE, CDC, and the IFCC WG APO-MS will prepare the transition from old to new
Lp(a) RMS along these lines, in a two-phased process.
References
Ridker PM, Hennekens CH, Stampfer MJ. A prospective study of lipoprotein(a) and the risk of
myocardial infarction. JAMA. 1993;270(18):2195–9.
Ridker PM, Stampfer MJ, Rifai N. Novel risk factors for systemic atherosclerosis: a comparison of
C-reactive protein, fibrinogen, homocysteine, lipoprotein(a), and standard cholesterol screen-
ing as predictors of peripheral arterial disease. JAMA. 2001;285(19):2481–5.
Rifai N, Ma J, Sacks FM, Ridker PM, Hernandez WJ, Stampfer MJ, et al. Apolipoprotein(a) size
and lipoprotein(a) concentration and future risk of angina pectoris with evidence of severe cor-
onary atherosclerosis in men: the Physicians’ Health Study. Clin Chem. 2004;50(8):1364–71.
Ruhaak LR, Cobbaert CM. Quantifying apolipoprotein(a) in the era of proteoforms and precision
medicine. Clin Chim Acta. 2020;511:260–8.
Ruhaak LR, Romijn FPHTM, Begcevic Brkovic I, Kuklenyik Z, Dittrich J, Ceglarek U, Hoofnagle
AN, Althaus H, Angles-Cano E, Coassin S, Delatour V, Deprez L, Dikaios I, Kostner GM,
Kronenberg F, Lyle A, Prinzing U, Vesper HW, Cobbaert CM. IFCC working group for stan-
dardization of apolipoproteins by mass spectrometry. Towards an SI-traceable LC-MRM-MS
based candidate reference measurement procedure for serum apolipoprotein (a). Clin Chem.
2023;69.
Schachtl-Riess JF, Kheirkhah A, Gruneis R, Di Maio S, Schoenherr S, Streiter G, et al. Frequent
LPA KIV-2 variants lower lipoprotein(a) concentrations and protect against coronary artery
disease. J Am Coll Cardiol. 2021;78(5):437–49.
Smith LM, Agar JN, Chamot-Rooke J, Danis PO, Ge Y, Loo JA, et al. The Human Proteoform
Project: defining the human proteome. Sci Adv. 2021;7(46):eabk0734.
Tate JR, Berg K, Couderc R, Dati F, Kostner GM, Marcovina SM, et al. International Federation
of Clinical Chemistry and Laboratory Medicine (IFCC) Standardization Project for the
Measurement of Lipoprotein(a). Phase 2: selection and properties of a proposed secondary
reference material for lipoprotein(a). Clin Chem Lab Med. 1999;37(10):949–58.
Tsimikas S, Fazio S, Viney NJ, Xia S, Witztum JL, Marcovina SM. Relationship of lipoprotein(a)
molar concentrations and mass according to lipoprotein(a) thresholds and apolipoprotein(a)
isoform size. J Clin Lipidol. 2018;12(5):1313–23.
Westgard QC. 2014. https://www.westgard.com/biodatabase1.htm.
Chapter 21
Therapy of Elevated Lipoprotein(a)
implications for clinical practice guidelines on the use of future Lp(a) lowering
therapies. These results also imply that randomized controlled trials evaluating
Lp(a) lowering therapies should enroll individuals with very high baseline Lp(a)
levels of 70–100 mg/dL or more, to demonstrate clinically meaningful reductions in
the risk of cardiovascular events.
Since the largest ASCVD risk reduction can be achieved in patients with the
highest baseline cardiovascular risk with concomitant highest Lp(a) levels, second-
ary prevention patients with Lp(a) elevation are the first to qualify for therapies
specifically and potently lowering Lp(a). In anticipation of the new drugs, this
patient group could benefit from more intensive LDL-C lowering [using either
statins or proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors].
Lifestyle and blood pressure lowering are also effective in partly attenuating the
Lp(a)-induced CV risk increase. Importantly, not only secondary prevention patients
are likely to benefit from Lp(a) lowering. An analysis from the Justification for the
Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin
(JUPITER) trial demonstrated that Lp(a) was also a significant determinant of
residual CVD risk in a cohort of primary prevention participants with low LDL-C
(Khera et al. 2014). In this group of primary prevention patients with very high
Lp(a), earlier and more intensive health behavior modification counselling and
management of other ASCVD risk factors are therefore often recommended
(Pearson et al. 2021).
As specific Lp(a) lowering therapies are still in development; for now, Lp(a) can
be incorporated into established ASCVD risk algorithms (Nurmohamed et al.
2021a). This holds true for both primary and secondary preventions. Nurmohamed
and colleagues demonstrated that in individuals with very high Lp(a) (>99th percen-
tile), the addition of Lp(a) into ASCVD risk algorithms resulted in 31% reclassifica-
tion in primary prevention and 63% reclassification in secondary prevention
(Nurmohamed et al. 2021a).
As circulating Lp(a) levels are, for more than 85%, mediated by genetic variation at
the LPA gene locus with only modest influence of environmental factors, pharmaco-
logical strategies to lower Lp(a) levels hold a major promise (Table 21.1) (Tsimikas
and Hall 2012). New therapies potently reducing apolipoprotein(a) (apo[a]) produc-
tion by the liver have shown the potential to lower Lp(a) levels by 80–98%.
Nonetheless, the clinical benefit of such substantial reductions in Lp(a) remains to
be established in phase 3 cardiovascular outcome trials. This leaves clinicians with
existing approaches to reduce ASCVD risk, which have varying effects on
Lp(a) levels.
21 Therapy of Elevated Lipoprotein(a) 349
Table 21.1 Therapeutic approaches in cardiovascular risk management that lower lipoprotein(a)
concentration
FDA/
Lp(a) lowering Lp(a) Cardiovascular EMA
strategies reduction risk reduction approval Reference(s)
Monoclonal PCSK9 23.0– Yes Yes Sabatine et al. (2017),
antibodies 27.0% Schwartz et al. (2018a),
O’Donoghue et al. (2019) and
Szarek et al. (2020)
PCSK9 siRNA 18.6– Under Yes Ray et al. (2020)
25.6% investigation
Niacin 23.0% No Yes Sahebkar et al. (2016)
CETP inhibitors 10.0– No or minimal Not Arsenault et al. (2018),
56.5% currently Schwartz et al. (2018b),
Bowman et al. (2017),
Thomas et al. (2017) and
Nicholls et al. (2022)
MTP inhibitors 17.0% Not investigated Yes Samaha et al. (2008)
Apheresis 30.0– Not investigated Yes Moriarty et al. (2019)
45.0%
IL-6 inhibitors 30.0– Under Yes Schultz et al. (2010), McInnes
40.0% investigation et al. (2015), García-Gómez
et al. (2017), Gabay et al.
(2016) and Ridker et al.
(2021)
Antisense 80.0% Under Not Viney et al. (2016)
oligonucleotides investigation currently
targeting Apo(a)
mrna
Small interfering >90.0% Under Not Koren et al. (2020),
RNA targeting investigation currently NCT04270760,
Apo(a) mrna NCT04606602
Statins
As compelling evidence has shown that statins are highly effective in reducing both
LDL-C levels and cardiovascular events, this drug class now represents the corner-
stone for treating patients with dyslipidemia and for attenuating cardiovascular risk
in both primary and secondary preventions (Baigent et al. 2010). Clinical trials
exploring the effect of statin therapy on Lp(a) levels, however, have shown mixed
results. The JUPITER trial, for example, showed no median change in Lp(a) with
rosuvastatin and placebo (Khera et al. 2014). Nonetheless, rosuvastatin did result in
a small but statistically significant positive shift in the overall Lp(a) distribution
(Khera et al. 2014). Furthermore, cell culture studies have found a time- and dose-
dependent, statin-mediated increase in the expression of LPA mRNA and
apolipoprotein(a) protein production when incubating HepG2 hepatocytes with
atorvastatin (Tsimikas et al. 2020). A meta-analysis, conducted by Tsimikas et al.
including 5256 patients randomized to receive rosuvastatin, atorvastatin,
350 S. Ibrahim and E. S. G. Stroes
Ezetimibe
Based on the available data that has been published, ezetimibe has a neutral
effect on circulating Lp(a) levels. A meta-analysis including 5188 individuals
from 10 randomized placebo-controlled trials demonstrated that ezetimibe ther-
apy had no effect on plasma Lp(a) concentrations (Sahebkar et al. 2018). In addi-
tion, a subgroup analysis indicated no significant alteration in plasma Lp(a) in
trials assessing the impact of ezetimibe monotherapy versus placebo nor in trials
evaluating the impact of adding ezetimibe to a statin versus statin therapy alone
(Sahebkar et al. 2018). Another meta-analysis, including 2337 patients with pri-
mary hypercholesterolemia from 7 randomized controlled trials, demonstrated
that ezetimibe 10 mg/day significantly reduced Lp(a) by 7.1% (Awad et al.
2018). However, investigators concluded that this small reduction was not clini-
cally significant.
PCSK9 Inhibitors
To date, PCSK9 inhibitors are the only class of LDL-C lowering drugs that has been
shown to lower Lp(a) as well as reducing CVD risk. The monoclonal PCSK9 anti-
bodies, alirocumab and evolocumab, have been evaluated in two large placebo-
controlled outcome trials, together comprising more than 100,000 patient years of
observation (Sabatine et al. 2017; Schwartz et al. 2018a). Both outcome trials
reported a significant reduction in major adverse cardiovascular events (MACE)
during treatment with PCSK9 antibodies for a median 2.3–2.7 years. Interestingly,
in both trials, the absolute cardiovascular risk reduction with a PCSK9 inhibitor was
higher in patients with higher baseline Lp(a) levels. In the FOURIER (Further
Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with
Elevated Risk) study, the absolute reduction in MACE with evolocumab was 2.41%
for Lp(a) >50 mg/dL versus 1.41% at lower levels (O’Donoghue et al. 2019). In the
21 Therapy of Elevated Lipoprotein(a) 351
There are several lipid-modifying agents that have been shown to reduce Lp(a) lev-
els in studies, however, without evidence substantiating that the Lp(a) lowering abil-
ity leads to improved clinical outcomes. Niacin, for example, which is an essential
micronutrient that raises the concentration of high-density lipoprotein cholesterol
(HDL-C) and reduces triglycerides (TG) as well as LDL-C concentrations, has also
been shown to reduce Lp(a) levels (Sahebkar et al. 2016). A meta-analysis of 14
randomized placebo-controlled trials demonstrated a mean Lp(a) reduction of 23%
in patients treated with extended-release niacin (Sahebkar et al. 2016). However,
two placebo-controlled trials evaluating the cardiovascular efficacy of extended-
release niacin demonstrated no cardiovascular efficacy of the drug and even showed
an increased risk of serious adverse events (Boden et al. 2011; Landray et al. 2014).
However, it should be noted that the median Lp(a) levels in these trials were low and
the number of patients with very high Lp(a) levels was very limited, which may
reduce the validity of these findings in the group with markedly elevated Lp(a) lev-
els. Nevertheless, due to adverse effects and the lack of evidence that Lp(a) reduc-
tion with niacin is associated with a cardiovascular benefit, niacin is not recommended
352 S. Ibrahim and E. S. G. Stroes
Apheresis
Non-apoB-Directed Therapies
Antisense Oligonucleotides
There are currently two small interfering RNA (siRNA) agents targeting LPA that
are being tested in different phases in clinical trials. SiRNAs are mostly 21–25
nucleotides long, double-stranded RNA that have sequence-homology-driven gene-
knockdown capability (Tromp et al. 2020). Following incorporation into the RNA-
induced silencing complex (RISC) in the plasma, the modified RISC allows for
gene blocking by binding and then degrading the mRNA produced by the gene by
the protein Argonaut-1. Two major differences with the antisense mechanism is it’s
mode of action in the cytoplasm rather than the nucleus; and second the long half-
life of the inhibitory effect of the siRNA due to the stability of the siRNA in the
RISC complex. Olpasiran (formerly known as AMG-890 and ARO-LPA) is one
example of an siRNA targeting apo(a) mRNA, which has shown reductions in Lp(a)
levels of more than 90% with no safety concerns identified in healthy volunteers in
a phase 1 study (Koren et al. 2020). A phase 2 dose finding study was recently com-
pleted in 290 patients with established ASCVD and Lp(a) levels ≥200 nmol/L,
showing a 97.4% reduction in placebo-adjusted changes of Lp(a) with the 75-mg
dose administered once every 3 months sucutaneously, (O’Donoghue et al. 2022). A
cardiovascular outcomes trial using Olpasiran has recently started (Ocean(a);
NCT05581303). Another GalNAc-conjugated siRNA targeting the mRNA tran-
script of LPA is SLN360 has reported a >95% Lp(a) reduction in the APOLLO trial
(Nissen et al. 2022). A larger phase II trial using SLN360 in patient with atheroscle-
rotic cardiovascular disease and Lp(a) elevation is expected to start in the beginning
of 2023 (NCT05537571).
Conclusion
In summary, current available therapeutic agents have only mild to moderate Lp(a)
lowering effects with PCSK9 inhibitors and apheresis being the only existing thera-
peutic approaches that have been shown to lower Lp(a) levels and reduce CVD risk.
The magnitude of treatment benefit for PCSK9 inhibitors is associated with baseline
Lp(a) levels and seems to be associated with the degree of Lp(a) reduction. Specific
and potent RNA-targeted interventions have the potential to greatly reduce Lp(a) con-
centrations. Cardiovascular outcomes trials will have to show whether such substan-
tial Lp(a) reductions are associated with meaningful clinical benefit, the outcomes of
which are expected in 2025 (HORIZON) and 2026/7 (Ocean(a)), respectively.
References
Arsenault BJ, Petrides F, Tabet F, Bao W, Hovingh GK, Boekholdt SM, et al. Effect of atorvastatin,
cholesterol ester transfer protein inhibition, and diabetes mellitus on circulating proprotein
subtilisin kexin type 9 and lipoprotein(a) levels in patients at high cardiovascular risk. J Clin
Lipidol. 2018;12(1):130–6.
21 Therapy of Elevated Lipoprotein(a) 355
Nissen SE, Wolski K, Balog C, Swerdlow DI, Scrimgeour AC, Rambaran C, et al. Single ascend-
ing dose study of a short interfering RNA targeting lipoprotein(a) production in individuals
with elevated plasma lipoprotein(a) levels. JAMA. 2022;327(17):1679–87.
Nurmohamed NS, Kaiser Y, Schuitema PCE, Ibrahim S, Nierman M, Fischer JC, et al. Finding very
high lipoprotein(a): the need for routine assessment. Eur J Prev Cardiol. 2021a;29(5):769–76.
Nurmohamed NS, Ditmarsch M, Kastelein JJP. CETP-inhibitors: from HDL-C to LDL-C lowering
agents? Cardiovasc Res. 2021b;118(14):2919–31.
O’Donoghue ML, Fazio S, Giugliano RP, Stroes ESG, Kanevsky E, Gouni-Berthold I, et al.
Lipoprotein(a), PCSK9 inhibition, and cardiovascular risk. Circulation. 2019;139(12):1483–92.
https://doi.org/10.1161/CIRCULATIONAHA.118.037184.
O’Donoghue ML, Rosenson RS, Gencer B, López JA, Lepor NE, Baum SJ, et al. Small
interfering RNA to reduce lipoprotein(a) in cardiovascular disease. N Engla J Med.
2022;387(20):1855–64.
Pearson GJ, Thanassoulis G, Anderson TJ, Barry AR, Couture P, Dayan N, et al. 2021 Canadian
Cardiovascular Society Guidelines for the management of dyslipidemia for the prevention of
cardiovascular disease in adults. Can J Cardiol. 2021;37(8):1129–50.
Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, et al. Two phase 3 trials of incli-
siran in patients with elevated LDL cholesterol. N Engl J Med. 2020;382(16):1507–19.
Ridker PM, Devalaraja M, Baeres FMM, Engelmann MDM, Hovingh GK, Ivkovic M, et al.
IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): a
double-blind, randomised, placebo-controlled, phase 2 trial. Lancet (London, England).
2021;397(10289):2060–9.
Rubino J, MacDougall DE, Sterling LR, Kelly SE, McKenney JM, Lalwani ND. Lipid lowering
with bempedoic acid added to a proprotein convertase subtilisin/kexin type 9 inhibitor therapy:
a randomized, controlled trial. J Clin Lipidol. 2021;15(4):593–601.
Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, et al.
Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med.
2017;376(18):1713–22.
Sahebkar A, Reiner Ž, Simental-Mendía LE, Ferretti G, Cicero AFG. Effect of extended-release
niacin on plasma lipoprotein(a) levels: a systematic review and meta-analysis of randomized
placebo-controlled trials. Metabolism. 2016;65(11):1664–78.
Sahebkar A, Simental-Mendía LE, Pirro M, Banach M, Watts GF, Sirotri C, et al. Impact of ezeti-
mibe on plasma lipoprotein(a) concentrations as monotherapy or in combination with statins: a
systematic review and meta-analysis of randomized controlled trials. Sci Rep. 2018;8(1):1–9.
Samaha FF, McKenney J, Bloedon LT, Sasiela WJ, Rader DJ. Inhibition of microsomal triglycer-
ide transfer protein alone or with ezetimibe in patients with moderate hypercholesterolemia.
Nat Clin Pract Cardiovasc Med. 2008;5(8):497–505.
Schultz O, Oberhauser F, Saech J, Rubbert-Roth A, Hahn M, Krone W, et al. Effects of inhibition
of interleukin-6 signalling on insulin sensitivity and lipoprotein(a) levels in human subjects
with rheumatoid diseases. PLoS One. 2010;5(12):e14328.
Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, et al. Alirocumab and cardio-
vascular outcomes after acute coronary syndrome. N Engl J Med. 2018a;379(22):2097–107.
Schwartz GG, Ballantyne CM, Barter PJ, Kallend D, Leiter LA, Leitersdorf E, et al. Association
of lipoprotein(a) with risk of recurrent ischemic events following acute coronary syndrome:
analysis of the dal-outcomes randomized clinical trial. JAMA Cardiol. 2018b;3(2):164–8.
Szarek M, Bittner VA, Aylward P, Baccara-Dinet M, Bhatt DL, Diaz R, et al. Lipoprotein(a)
lowering by alirocumab reduces the total burden of cardiovascular events independent of
low-density lipoprotein cholesterol lowering: ODYSSEY OUTCOMES trial. Eur Heart
J. 2020;41(44):4245–55. https://doi.org/10.1093/eurheartj/ehaa649.
Thomas T, Zhou H, Karmally W, Ramakrishnan R, Holleran S, Liu Y, et al. CETP (cholesteryl
ester transfer protein) inhibition with anacetrapib decreases production of lipoprotein(a) in
mildly hypercholesterolemic subjects. Arterioscler Thromb Vasc Biol. 2017;37(9):1770–5.
Tromp TR, Stroes ESG, Hovingh GK. Gene-based therapy in lipid management: the winding road
from promise to practice. Expert Opin Investig Drugs. 2020;29(5):483–93. https://doi.org/1
0.1080/13543784.2020.1757070.
21 Therapy of Elevated Lipoprotein(a) 357
Tsimikas S, Hall JL. Lipoprotein(a) as a potential causal genetic risk factor of cardiovascular dis-
ease: a rationale for increased efforts to understand its pathophysiology and develop targeted
therapies. J Am Coll Cardiol. 2012;60(8):716–21.
Tsimikas S, Gordts PLSM, Nora C, Yeang C, Witztum JL. Statin therapy increases lipoprotein(a)
levels. Eur Heart J. 2020;41(24):2275–84.
Viney NJ, van Capelleveen JC, Geary RS, Xia S, Tami JA, Yu RZ, et al. Antisense oligonucleotides
targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind,
placebo-controlled, dose-ranging trials. Lancet. 2016;388(10057):2239–53.
Chapter 22
Antisense Oligonucleotide Therapy
to Treat Elevated Lipoprotein(a)
Sotirios Tsimikas
S. Tsimikas (*)
Division of Cardiology, Sulpizio Cardiovascular Center, University of California San Diego,
La Jolla, CA, USA
e-mail: [email protected]
One of the limitations in developing modalities to lower Lp(a) in plasma has been
the narrow species distribution of the LPA gene, limited to hedgehogs, Old World
monkeys, apes, and humans (McLean et al. 1987; Tomlinson et al. 1989). Thus,
appropriate animal models that can encompass the entire Lp(a) pathophysiology,
including regulatory elements, have generally been lacking (Yeang et al. 2016).
One of the earliest reported attempts to interfere with apolipoprotein(a) biosyn-
thesis was by Frank et al. (2001). They doubly transfected mice with an adenovirus-
mediated N-terminal truncated apolipoprotein(a) construct comprising the
5′-untranslated region, the signal sequence, and the first three kringles of native
apolipoprotein(a) along with a concomitant antisense molecule directed to
apolipoprotein(a) mRNA. Evidence of transient but efficient reductions of
apolipoprotein(a) synthesis was shown. However, this approach could not be easily
translated to human applications due to the limitations of the animal models and the
rapid degradation and presumed inefficiency of the antisense constructs used.
Several approaches have been shown to nonselectively and modestly lower
Lp(a), including lipid apheresis, niacin, CETP inhibitors, and PCSK9 inhibitors
(Tsimikas et al. 2021). The first proof-of-concept description of effective and long-
lasting in vivo lowering of Lp(a) was reported by Merki et al. (2008) in 2008 in a
collaborative effort of the Ionis Pharmaceuticals (then named Isis Pharmaceuticals)
led by Mark Graham and Rosanne Crooke and our laboratory at UCSD (University
of California, San Diego). In this study, Lp(a) lowering was achieved by the anti-
sense oligonucleotide mipomersen, directed to human apoB-100, which signifi-
cantly reduced human apoB-100 levels in Lp(a) transgenic mice expressing both
human apoB-100 and apolipoprotein(a) needed to generate authentic Lp(a) parti-
cles. Over the 11-week treatment period, compared with baseline, mipomersen
reduced Lp(a) levels by up to 75% (p < 0.0001) in a time-dependent fashion
(Fig. 22.1a). This was primarily due to limiting the availability of apoB-100 to bind
to apolipoprotein(a), as LPA mRNA expression and plasma apolipoprotein(a) levels
were not affected by mipomersen. Furthermore, mipomersen significantly reduced
plasma levels of oxidized phospholipids on apoB (OxPL-apoB) and apolipoprotein(a)
[OxPL-apo(a)] particles (Fig. 22.1b). This study provided proof of concept that
reducing the availability of apoB-100 is a limiting factor in Lp(a) particle assembly
in this Lp(a) transgenic mouse model. These preclinical findings were later con-
firmed in clinical studies of mipomersen in a variety of settings, including in patients
with homozygous and heterozygous familial and multifactorial hypercholesterol-
emia, showing approximately 25% reduction in Lp(a) that was independent of
LDL-C lowering (Santos et al. 2015). The study also taught us that targeting apoB
is not an ideal mechanism to lower Lp(a), as it does not affect the pathognomonic
protein of Lp(a), apolipoprotein(a). Furthermore, the pathophysiological effect of
free apolipoprotein(a), and whether it is more or less atherothrombotic and proin-
flammatory than Lp(a), is not known.
22 Antisense Oligonucleotide Therapy to Treat Elevated Lipoprotein(a) 361
Fig. 22.1 Temporal changes in Lp(a) levels (A) and in OxPL-apoB levels in Lp(a)-transgenic
mice treated with control ASO or mipomersen directed to human apoB. Temporal changes in
apo(a) levels (A) and in OxPL-apo(a) levels in apo(a)-transgenic mice treated with saline, control
ASO or ASO 144367. *P<001, **P<0.01 vs baseline values. Merki et al. 2008, 2011)
in 8K-apo(a) mice (Fig. 22.1d). These studies provided proof that targeting liver
expression of apolipoprotein(a) with ASOs directed to KIV2 repeats may provide an
effective approach to lower elevated Lp(a) levels in humans.
Natural DNA and RNA are not suitable as effective drugs due to insufficient stabil-
ity mediated by their rapid plasma degradation by nucleases and limited tissue dis-
tribution in animals. Antisense oligonucleotides (ASOs) are single-stranded
modified DNA molecules comprised of 15–20 nucleic acids that display a comple-
mentary sequence to a target messenger ribonucleic acid (mRNA). To overcome the
limitations of naked DNA/RNA as pharmaceutical agents, ASOs contain certain
modifications of the phosphate backbone and the 2′ ribose position. ASOs have a
specific sequence to the target of interest that is not repeated throughout genome,
reducing the potential for off-target binding.
Using medicinal chemistry approaches, first-generation ASOs led to the substitu-
tion of phosphodiester (PO) bonds with phosphorothioate (PS) at one of the two
available PO bonds at each phosphate group in the backbone to improve stability of
the DNA:RNA complex and improve distribution to tissues. The PS bonds provide
stability and protection against nucleases. Second-generation ASOs are called
MOE-gapmers, in that the middle ten nucleic acids are unmodified DNA, which is
required for RNAse H1-mediated cleavage, whereas the five nucleic acids on each
wing are modified at the 2′ position by MOE. The 2′-MOE moiety increases stabil-
ity in biological systems, increases potency due to improved binding affinity to its
target mRNA, and improves the safety profile by decreasing proinflammatory
effects and class toxicities. The nucleic acid bases are in their native chemical con-
figuration and generally not modified in ASOs. Examples of 2′-MOEs are inotersen
(Benson et al. 2018) that is approved clinically for hereditary amyloid polyneuropa-
thy and volanesorsen (Witztum et al. 2019) that is approved clinically for familial
chylomicronemia syndrome in the European Union.
A further advance was made in generation 2+ molecules by improving the
screening process for proinflammatory and other undesirable side effects, as well as
by removing some of the PS groups and replacing them with their native PO groups
in the backbone. This class of drugs are represented by IONIS-APO(a)LRx, ole-
zarsen (Tardif et al. 2022), targeting ApoCIII to treat hypertriglyceridemia and
AGT-LRx (Morgan et al. 2021) targeting angiotensinogen for the proposed treatment
of resistant hypertension and heart failure.
Additional improvements in antisense oligonucleotides include generation 2.5
molecules, where the 2′-MOE has been replaced by a constrained ethyl moiety, as
exemplified by ION409/AZD8233 targeting PCSK9 (Gennemark et al. 2021). The
changes in chemistry, from first generation to second generation and to generation
2.5, each improved potency by approximately tenfold and cumulatively by approxi-
mately 1000-fold, along with additional improvements in safety and tolerability
(Crooke et al. 2018).
22 Antisense Oligonucleotide Therapy to Treat Elevated Lipoprotein(a) 363
Fig. 22.2 Screening process to identify human antisense oligonucleotides to lower plasma Lp(a)
364 S. Tsimikas
ASOs that span broad regions of the gene of interest. Top leads are then evaluated
in dose-response comparisons with leads demonstrating the greatest potency
selected for in vivo evaluation. Additionally, lead targeting sites are interrogated
with “microwalks” to identify the most potent ASOs with an optimal safety and
tolerability profile. In the case of identifying pelacarsen, this process required the
synthesis of over 2800 unique ASOs prior to the identification of the clinical candi-
date. The most promising candidates, in this case 38 ASOs, were further interro-
gated for efficacy in Lp(a) or apo(a) transgenic mice and in rodent toxicology and
pharmacokinetic studies. This process narrowed the choice to six potential candi-
dates that were then evaluated in cynomolgus monkey tolerability studies, and the
best candidate was identified to enter IND (Investigational New Drug) enabling
toxicology and pharmacokinetic studies.
ISIS-APO(a)Rx (later named IONIS-APO(a)Rx) is a second-generation
2′-O-(2-methoxyethyl) (2′-MOE)-modified ASO drug with the sequence
5′-TGCTCCGTTGGTGCTTGTTC-3′ (Fig. 22.3) (Graham et al. 2016). ISIS-
APO(a)Rx/IONIS-APO(a)Rx contains five 2′-MOE-modified ribonucleosides at the 5′
and 3′ ends and ten 2-O-deoxyribonucleosides within the central portion of the mol-
ecule. A modified version of IONIS-APO(a)Rx containing GalNAc3 was generated,
initially named IONIS-APO(a)-LRx, and then AKCEA-APO(a)LRx and ultimately
received the generic name pelacarsen. Pelacarsen contains the same 20-nucleotide
sequence as IONIS-APO(a)Rx and five 2′-MOE-modified ribonucleosides at the 5′
Fig. 22.3 ISIS-APO(a)Rx complementary binding sites within the human LPA transcript (GenBank
accession NM_005577.2) at position 3901–3920 bp. ISIS-APO(a)Rx was designed to perfectly
match only the exon 24–25 splice sites (indicated with bold type) but may also bind at 11 other
apolipoprotein(a) exon splice sites containing 1–4 mismatched nucleotides (indicated by under-
lined letters) (Graham et al. 2016)
22 Antisense Oligonucleotide Therapy to Treat Elevated Lipoprotein(a) 365
and 3′ ends while retaining ten 2-odeoxyribonucleosides within the central portion
of the molecule. However, 6 of the 19 PS linkages were replaced with PO linkages
at positions 2, 3, 4, 5, 16, and 17. The GalNAc3 complex was covalently attached
with a proprietary linker to the 5′ end (Viney et al. 2016) (Fig. 22.4).
specific sequence of the ASO or the target, and cleaves the target mRNA, thereby
disrupting protein translation (Fig. 22.5). The ASO is relatively resistant to RNAse
H1-mediated cleavage and becomes available to bind to additional mRNA LPA mol-
ecules. This is part of reason the intra-hepatocyte half-life is relatively long (2–4 weeks).
A total of four clinical phase 1 or phase 2 trials have been performed with ISIS-
APO(a)Rx/IONIS-APO(a)Rx/and pelacarsen (Table 22.1). For historical purposes
and to be consistent with the literature, the names of the drugs will be given
according to those used when the trials were published.
The first clinical demonstration that Lp(a) levels can be potently reduced in
patients was documented in a randomized, double-blind, placebo-controlled, phase
1 study of healthy adults with Lp(a) concentration of ≥25 nmol/L assigned to
receive ISIS-APO(a)Rx or placebo. Multiple doses of ISIS-APO(a)Rx (100–300 mg)
resulted in dose-dependent, mean percentage decreases in plasma Lp(a) concentra-
tion of 39.6% from baseline in the 100 mg group (p = 0.005), 59.0% in the 200 mg
group (p = 0.001), and 77.8% in the 300 mg group (p = 0.001) (Fig. 22.6). Similar
reductions were observed in OxPL-apoB and OxPL-apo(a) (Fig. 22.6). Mild injec-
tion site reactions were the most common adverse events. No serious or severe
adverse events were recorded. Two of the 37 participants treated with ISIS-APO(a)Rx
discontinued the study drug for tolerability reasons, which was an improved experi-
ence compared to previous earlier drugs in this class of chemicals.
ISIS-APO(a)Rx was renamed IONIS-APO(a)Rx in concert with the change in the
company’s name. The phase 2 study that followed was performed in participants
with elevated Lp(a) concentrations (125–437 nmol/L in cohort A with 51 partici-
pants, ≥438 nmol/L in cohort B with 13 participants) who were randomly assigned
to escalating-dose subcutaneous IONIS-APO(a)Rx (100 mg, 200 mg, and then
300 mg, once a week for 4 weeks each) or saline placebo, once a week, for 12 weeks.
At day 85/99, participants assigned to IONIS-APO(a)Rx had mean Lp(a) reductions
of 66.8% in cohort A and 71.6% in cohort B (both p < 0.0001 vs pooled placebo)
Fig. 22.6 Plasma trough concentrations of ISIS-APO(a)Rx and mean percent change in Lp(a),
OxPL-apoB, and OxPL-apo(a) with time by treatment group in the multidose cohorts measured 7
days after the last dose in the 300 mg dose cohort in relation to change in concentration of plasma
Lp(a), OxPL-apoB, and OxPL-apo(a). The shaded area represents the dosing window, and arrows
indicate dosing at days 1, 3, 5, 8, 15, and 22 (Tsimikas et al. 2015)
22 Antisense Oligonucleotide Therapy to Treat Elevated Lipoprotein(a) 369
(Fig. 22.7). Mean concentrations were also reduced in OxPL-apoB (35.2% for
cohort A and 42.5% for cohort B), OxPL-apo(a) (26.6% for cohort A and 36.7% for
cohort B), LDL-C (13.0% for cohort A and 23.9% for cohort B), and apoB-100
(11.3% for cohort A and 18.5% for cohort B) (Fig. 22.7).
Baseline hsCRP concentrations were 2.39 mg/L for the placebo group, 1.78 mg/L
for cohort A, and 3.46 mg/L for cohort B. At day 85/99, mean absolute change in
hsCRP was −0.64 mg/L (SD 4.38, p = 0.44 vs baseline) for the pooled placebo
group, −0.23 mg/L for cohort A (SD 1.54, p = 0.92 vs baseline and p = 0.63 vs
change for placebo), and − 1.5 mg/L for cohort B (SD 3.27, p = 0.37 vs baseline
and p = 0.20 vs change in placebo). Overall, IONIS-APO(a)Rx was generally well
Lp(a) OxPL-apoB
Placebo
Mean change in fasting ApoB from baseline (%) Mean change in fasting OxPL-apo(a) from baseline (%) Mean change in fasting Lp(a) from baseline (%)
0 0
–25 –25
–50 –50
–75
–75
OxPL-apo(a) LDL-C
Mean change in fasting Lp(a) from baseline (%)
25 25
0 0
–25 –25
–50 –50
–75 –75
–25
–50
–75
Fig. 22.7 Mean percent changes in plasma concentrations of Lp(a), OxPL-apoB, OxPL-apo(a),
LDL-C, and apoB in the IONIS-APO(a)Rx trial. Error bars are SEM (standard error of the mean).
The shaded area represents the dosing window and arrows indicate dosing every week. p values
show differences between treatment and pooled placebo at day 85/99. *p ≤ 0.0001, †p = 0.0002,
‡
p = 0.0005, §p = 0.0007, ¶p = 0.0003 (Viney et al. 2016)
370 S. Tsimikas
(Fig. 22.9b). The percent of patients with an Lp(a) level of ≤50 mg/dL (125 nmol/L)
or lower at 6 months of exposure was 23% in the group who received 20 mg of pelac-
arsen every 4 weeks, 63% in the group who received 40 mg every 4 weeks, 65% in
the group who received 20 mg every 2 weeks, 81% in the group who received 60 mg
every 4 weeks, and 98% in the group who received 20 mg every week (Fig. 22.9c).
The mean percent reductions in OxPL-apoB were 37% at a dose of 20 mg every
4 weeks, 57% at 40 mg every 4 weeks, 64% at 20 mg every 2 weeks, 79% at 60 mg
every 4 weeks, and 88% at 20 mg every week, as compared with a 14% increase in
the placebo group. The corresponding mean percent reductions in OxPL-apo(a)
were 28%, 49%, 45%, 63%, and 70%, respectively, compared with a 20% decrease
in the placebo group. Corresponding absolute reductions in LDL-C were −5.6,
−13.5, −13.2, −8.2, and −16.4 mg/dL, respectively, compared to −1.2 mg/dL in
placebo. Corresponding absolute reductions in apoB were −2.2, −8.3, −6.3, −3.9,
and −10.9 mg/dL, respectively, compared to 0.6 mg/dL increase in placebo.
Corresponding absolute changes (nonsignificant) in hsCRP were −0.9, −0.7, −0.3,
−0.5, and −0.1 mg/L, respectively, compared to −0.8 mg/L in placebo.
Individual responses revealed that all patients in the pelacarsen 20 mg weekly
dose had declines in Lp(a) (−42.6 to −99.5%), OxPL-apoB (−37.0 to 99.7%), and
OxPL-apo(a) (−12.6 to −99.5%), compared to +16.1 to −40.6%, −28.7 to +150.0%,
and −66.6 to +18.1 for the three variables in the placebo groups, respectively.
There were no significant differences between any pelacarsen dose and placebo
with respect to platelet counts, liver and renal measures, or influenza-like symp-
toms. The most common adverse events were injection site reactions.
Based on the totality of epidemiologic, genetic, and clinical evidence, as well as the
proof of concept in potently lowering Lp(a) these four trials provided, further devel-
opment of pelacarsen was undertaken. The Lp(a) HORIZON trial (NCT04023552,
Assessing the Impact of Lipoprotein(a) Lowering With Pelacarsen on Major
Cardiovascular Events in Patients With CVD [Lp(a) HORIZON] is a pivotal phase
3 study designed to support an indication for the reduction of cardiovascular risk in
patients with established CVD and elevated Lp(a). It is a global, international mul-
ticenter, randomized, double-blind, placebo-controlled study in >8000 patients with
elevated Lp(a) levels (≥70 mg/dL, ≥175 nmol/L) and history of CVD (myocardial
infarction, ischemic stroke, peripheral artery disease). Key inclusion criteria include
1-Lp(a) ≥70 mg/dL, 2-myocardial infarction ≥3 months from screening and ran-
domization to ≤10 years prior to the screening visit, 3-ischemic stroke ≥3 months
from screening and randomization to ≤10 years prior to the screening visit, and
clinically significant symptomatic peripheral artery disease.
Subjects are required to have risk factors, particularly LDL-C, optimized accord-
ing to local guidelines. They are then randomized to pelacarsen 80 mg
374 S. Tsimikas
Acknowledgements We thank Rosanne Crooke, Mark Graham, Richard Geary, Nick Viney, and
other members of the Ionis Pharmaceuticals and Joseph L. Witztum for their contributions in dis-
covering and developing pelacarsen and Trace Reigle of the Ionis Pharmaceuticals for the prepara-
tion of some of the artwork.
References
Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treat-
ment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22–31.
Berg K. A new serum type system in man—the LP system. Acta Pathol Microbiol Scand.
1963;59:369–82.
22 Antisense Oligonucleotide Therapy to Treat Elevated Lipoprotein(a) 375
Berg K, Dahlen G, Frick MH. Lp(a) lipoprotein and pre-beta1-lipoprotein in patients with coro-
nary heart disease. Clin Genet. 1974;6(3):230–5.
Crooke ST, Witztum JL, Bennett CF, Baker BF. RNA-targeted therapeutics. Cell Metab.
2018;27(4):714–39.
Frank S, Gauster M, Strauss J, Hrzenjak A, Kostner GM. Adenovirus-mediated apo(a)-antisense-
RNA expression efficiently inhibits apo(a) synthesis in vitro and in vivo. Gene Ther.
2001;8(6):425–30.
Gennemark P, Walter K, Clemmensen N, Rekić D, Nilsson CAM, Knöchel J, et al. An oral anti-
sense oligonucleotide for PCSK9 inhibition. Sci Transl Med. 2021;13(593):eabe9117.
Graham MJ, Viney N, Crooke RM, Tsimikas S. Antisense inhibition of apolipoprotein(a) to lower
plasma lipoprotein(a) levels in humans. J Lipid Res. 2016;57(3):340–51.
McLean JW, Tomlinson JE, Kuang WJ, Eaton DL, Chen EY, Fless GM, et al. cDNA sequence of
human apolipoprotein(a) is homologous to plasminogen. Nature. 1987;330(6144):132–7.
Merki E, Graham MJ, Mullick AE, Miller ER, Crooke RM, Pitas RE, et al. Antisense oligonu-
cleotide directed to human apolipoprotein B-100 reduces lipoprotein(a) levels and oxidized
phospholipids on human apolipoprotein B-100 particles in lipoprotein(a) transgenic mice.
Circulation. 2008;118(7):743–53.
Merki E, Graham M, Taleb A, Leibundgut G, Yang X, Miller ER, et al. Antisense oligonucleotide
lowers plasma levels of apolipoprotein(a) and lipoprotein(a) in transgenic mice. J Am Coll
Cardiol. 2011;57(15):1611–21.
Morgan ES, Tami Y, Hu K, Brambatti M, Mullick AE, Geary RS, et al. Antisense inhibition of
angiotensinogen with IONIS-AGT-LRx: results of phase 1 and phase 2 studies. JACC Basic
Transl Sci. 2021;6(6):485–96.
Santos RD, Raal FJ, Catapano AL, Witztum JL, Steinhagen-Thiessen E, Tsimikas S. Mipomersen,
an antisense oligonucleotide to apolipoprotein B-100, reduces lipoprotein(a) in various popula-
tions with hypercholesterolemia: results of 4 phase III trials. Arterioscler Thromb Vasc Biol.
2015;35(3):689–99.
Tardif JC, Karwatowska-Prokopczuk E, Amour ES, Ballantyne CM, Shapiro MD, Moriarty PM,
et al. Apolipoprotein C-III reduction in subjects with moderate hypertriglyceridaemia and at
high cardiovascular risk. Eur Heart J. 2022;43(14):1401–12.
Tomlinson JE, McLean JW, Lawn RM. Rhesus monkey apolipoprotein(a). Sequence, evolution,
and sites of synthesis. J Biol Chem. 1989;264(10):5957–65.
Tsimikas S, Viney NJ, Hughes SG, Singleton W, Graham MJ, Baker BF, et al. Antisense therapy
targeting apolipoprotein(a): a randomised, double-blind, placebo-controlled phase 1 study.
Lancet. 2015;386(10002):1472–83.
Tsimikas S, Fazio S, Viney NJ, Xia S, Witztum JL, Marcovina SM. Relationship of lipoprotein(a)
molar concentrations and mass according to lipoprotein(a) thresholds and apolipoprotein(a)
isoform size. J Clin Lipidol. 2018;12(5):1313–23.
Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, Tardif JC, Baum SJ, Steinhagen-
Thiessen E, et al. Lipoprotein(a) reduction in persons with cardiovascular disease. N Engl J
Med. 2020;382(3):244–55.
Tsimikas S, Moriarty PM, Stroes ES. Emerging RNA therapeutics to lower blood levels of Lp(a):
JACC focus seminar 2/4. J Am Coll Cardiol. 2021;77(12):1576–89.
Viney NJ, van Capelleveen JC, Geary RS, Xia S, Tami JA, Yu RZ, et al. Antisense oligonucleotides
targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind,
placebo-controlled, dose-ranging trials. Lancet. 2016;388(10057):2239–53.
Vuorio A, Watts GF, Schneider WJ, Tsimikas S, Kovanen PT. Familial hypercholesterolemia and
elevated lipoprotein(a): double heritable risk and new therapeutic opportunities. J Intern Med.
2020;287(1):2–18.
Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, et al.
Volanesorsen and triglyceride levels in familial chylomicronemia syndrome. N Engl J Med.
2019;381(6):531–42.
Yeang C, Cotter B, Tsimikas S. Experimental animal models evaluating the causal role of
lipoprotein(a) in atherosclerosis and aortic stenosis. Cardiovasc Drugs Ther. 2016;30(1):75–85.
Chapter 23
Lipoprotein Apheresis for Reduction
of Lipoprotein(a)
Abbreviations
A Atorvastatin
AIC Arteria iliaca communis
Aorta-mes bypass Mesenteric artery bypass
A. mes sup Arteria mesenterica superior
ApoE Apolipoprotein E-polymorphism
BMS Bare metal stent
CAD Coronary artery disease
CX Left circumflex coronary artery
DEB Drug-eluting balloon
DES Drug-eluting stent
LAD Left anterior descending artery
LCA Left coronary artery
LMS Left main stem
nk Not known
MI Myocardial infarction
PLA Posterolateral artery
PTA Percutaneous transluminal angioplasty
PTCA Percutaneous transluminal coronary angioplasty
RCA Right coronary artery
RDI Ramus diagonalis, diagonal branch 1
R Rosuvastatin
S Simvastatin
TEA Thromboendarteriectomy
Tr. coel Truncus coeliacus
Introduction
Between 1975 and 2004, several LA systems were developed to treat patients with
familial hypercholesterolemia (Julius 2017; Thompson 2022). Lp(a) appeared as a
target molecule only after 2000.
The largest number of patients treated with LA worldwide exists in Germany. In
2008, the Joint Federal Committee (Gemeinsamer Bundesausschuss), a paramount
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 379
Table 23.1 Numbers of patients treated with LA at the end of the given years in Germany
Data are officially published by the National Association of Statutory Health Insurance Physicians
(Kassenärztliche Bundesvereinigung) (Kassenärztliche Bundesvereinigung 2014, 2015, 2016,
2017, 2018, 2019, 2020, 2021)
hoFH homozygous familial hypercholesterolemia, HCH hypercholesterolemia
The percentage of patients who were included into LA treatment with the indica-
tion of an isolated Lp(a) elevation was increasing from 35% in 2013 to 59% in 2019.
At the Dresden Center for Extracorporeal Therapy, in the last years, the vast major-
ity of new patients had elevations of Lp(a). Unfortunately, our 160 patients are not
included into the data shown in Table 23.1 because working in a university hospital,
we do not belong to the National Association of Statutory Health Insurance
Physicians.
An LA session acutely reduces lipid concentrations which rise again in the follow-
ing days. Thus a sawtooth picture is seen. In our experience, after 7 days, the Lp(a)
level is again at its maximum.
In order to describe the efficiency of LA on Lp(a) levels, the following
criteria are used
1. Acute reductions—comparing the Lp(a) level after LA sessions with those
before the sessions.
2. Interval mean values—taking into account that Lp(a) increases following an LA
session. This parameter defines the burden imposed by Lp(a) on arteries. We
have published a formula to calculate interval mean values (Tselmin et al. 2017).
3. Comparison of the presession and post-session and interval mean values with the
initial Lp(a) concentrations measured before the first LA session (reflecting the
steady-state condition till the start of the extracorporeal therapy).
The following data have been measured at our center in 2019; the first LA ses-
sion usually took place years ago (Julius et al. 2020). The vast majority of our
patients are treated with LA weekly.
In our patients, the acute reductions amounted to 128 nmol/L (−77%) (see
Table 23.2). When comparing with the initial Lp(a) level before the first LA session,
the interval mean values were decreased by 134 nmol/L (−55%). Waldmann and
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 381
Table 23.2 Lp(a) before the first LA session, before and after an LA session, and interval mean
values after years of extracorporeal therapy—percent reductions comparing with the initial levels
are marked in red (n = 97; data have been recalculated on the basis of a publication in 2020) (Julius
et al. 2020)
Fig. 23.1 Interval mean values of Lp(a) (n = 97; x-axis: upper bounds of intervals, nmol/L)
Parhofer reported a decrease of this parameter between 25 and 40% (Waldmann and
Parhofer 2016). Evidently, the results obtained in Dresden are superior to these data.
But it has to be taken into account that an additional Lp(a)-lowering effect may be
due to the application of PCSK9 antibodies in some patients at our center.
The following histogram (Fig. 23.1) shows the interval mean values of Lp(a).
Only in 20 (21%) patients the interval mean value was below 75 nmol/L.
382 U. Julius and S. Tselmin
Almost half of the patients showed interval mean values above 120 nmol/L.
The corresponding LDL-C interval mean value [calculated according to Kroon
et al. (2000)] was 1.75 mmol/L (IQR 1.32/2.22 mmol/L), which is −15% lower than
the initial level [2.07 mmol/L (IQR 1.77/3.04 mmol/L)].
There are three lipoprotein classes carrying apolipoprotein B (ApoB): VLDL,
LDL, and Lp(a). All these lipoproteins are removed by LA.
We measured ApoB in our patients in January 2021 and obtained the results
shown in Fig. 23.2.
The median of ApoB is effectively acutely reduced by −70%. The mean ApoB
concentration (between pre- and post-values) is 55 mg/dL.
In a recently published American statement on Lp(a), it was recommended to
calculate the percentage of ApoB transported with the Lp(a) particles (Reyes-Soffer
et al. 2022). Interestingly, in the HEART UK consensus statement, an expression of
Lp(a) in molar units in order to appreciate its concentration relative to ApoB
expressed in molar units is discussed (Cegla et al. 2019).
Our data before and after an LA session are depicted in Fig. 23.3.
The median is a little bit decreased. In Fig. 23.3b, it is shown that the percentage
of ApoB contained in Lp(a) exceeding 20% is increased after the LA session com-
paring with the initial data.
The rebound of LDL and Lp(a) particle concentrations following LA allows the
determination of fractional catabolic rate (FCR) and hence production rate (PR)
during nonsteady-state conditions (Ma et al. 2019). The FCR of Lp(a) was signifi-
cantly lower than that of LDL-ApoB, implying that different metabolic pathways
are involved in the catabolism of these lipoproteins, with no significant differences
in the corresponding PR.
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 383
40
30
Frequency
20
10
0
0 50 100 150 200 250
40
30
Frequency
20
10
0
20 40 60 80
Fig. 23.2 ApoB concentrations (mg/dL) before (a) and after (b) one LA session. (a) Median
80 mg/dL (IQR 68/98 mg/dL). (b) Median 24 mg/dL (IQR 10/30 mg/dL)
384 U. Julius and S. Tselmin
25
20
15
Frequency
10
0
.00 10.00 20.00 30.00 40.00 50.00
30
20
Frequency
10
0
.00 10.00 20.00 30.00 40.00 50.00
Fig. 23.3 Percentage of ApoB transported with Lp(a) particles before (a) and after (b) an LA ses-
sion. (a) Median 11.3% (IQR 6.9/14.6%). (b) Median 9.8% (IQR 5.0/18.0%)
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 385
Oxidized Phospholipids
Oxidized phospholipids (OxPL) are mainly transported by Lp(a) particles and play
an important role in atherogenesis (Yeang et al. 2016; Tsimikas 2019).
A small study in 18 patients with familial hypercholesterolemia and with low
(≈10 mg/dL; range 10–11 mg/dL), intermediate (≈50 mg/dL; range 30–61 mg/dL),
or high (>100 mg/dL; range 78–128 mg/dL) Lp(a) levels was performed to check
the effect of LA on OxPL-ApoB and OxPL-apolipoprotein(a) [Apo(a)] concentra-
tions (Arai et al. 2012). Plasma OxPL-ApoB was not reduced in the low Lp(a)
group, but the levels were very low and near the level of detection of this assay.
There was a strong trend for acute reduction (48%) in OxPL-ApoB in the intermedi-
ate Lp(a) group, and there was a significant decline (62%) in the high Lp(a) group.
OxPL-Apo(a) was significantly reduced in all groups.
Observational Studies
Observational Studies
The Dresden Center for Extracorporeal Therapy was involved in both studies.
Moreover, we were the first to report that LA is more efficient with respect to the
reduction of CVEs in patients whose Lp(a) levels are elevated when comparing with
patients with normal or non-detectable Lp(a) (von Dryander et al. 2013; Schatz
et al. 2017). This difference had been confirmed by another group (Heigl et al. 2015)
and also in the German Lipoprotein Apheresis Registry (see below).
Moreover, we compared patients who developed CVEs while being treated with
LA (n = 48) to those who did not suffer from CVEs (n = 60) (Julius et al. 2020).
Both groups were on extracorporeal therapy for years already, for a mean period of
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 387
Twenty-three patients with Lp(a) levels above 60 mg/dL and a pre-apheresis LDL-C
<100 mg/dL on maximally tolerated lipid-lowering therapy were included (Bigazzi
et al. 2018). They were treated with LA for several years (median 7, interquartile
range 3–9 years) by heparin-induced LDL precipitation apheresis (16/23), dextran-
sulfate (4/23), cascade filtration (2/23), and immunoadsorption (1/23). The time lapse
between first cardiovascular event and beginning of apheresis was 6 years (interquar-
tile range 1–12 years). The rates of adverse cardiovascular events were reduced by
74% when comparing the situation before and after the LA treatment inception.
Ten patients with severe PAD and isolated Lp(a)-HLP who recently underwent revas-
cularization (index procedure) were included (Poller et al. 2017). When comparing
the situation before LA with the results after 12 months, the pain level, ankle-brachial
index (ABI), transcutaneous oxygen pressure (tcpO2), and walking distance all
improved. Importantly, the frequency of revascularization procedures was strongly
decreased under LA. All patients combined underwent 35 revascularizations within
the 12 months prior to the index procedure (mean interval between two revascular-
izations: 104.3 days). Since the index procedure, only one revascularization was nec-
essary within 79 patient-months under LA (mean interval: 2404.5 days, p < 0.001).
Fourteen patients with cardiovascular disease with elevated Lp(a) and near-normal
LDL-C were treated with LA over a mean treatment period of 48 months (range
8–105 months) (Moriarty et al. 2019). The authors describe a 94% reduction in
major adverse cardiovascular events.
388 U. Julius and S. Tselmin
This registry has existed since 2011. In the annual report for 2020, data on 1111
patients (from 44 LA centers, 6791 LA sessions) have been documented (Schettler
et al. 2020). Following the suggestion from Dresden, three hyperlipoproteinemia
(HLP) groups have been defined (based on the initial lipid values): (a) with isolated
elevation of LDL-C [Lp(a) level <60 mg/dL or <120 nmol/L or not detectable,
n = 180], (b) with isolated elevation of Lp(a) [Lp(a) ≥60 mg/dL or ≥120 nmol/L
and LDL-C <2.6 mmol/L, n = 500), and (c) with combined elevation of both LDL-C
and Lp(a) (using the abovementioned criteria, n = 228). The latter group is totally
neglected in the officially published data.
The following mean acute reduction rates were reported for the LA sessions:
LDL-C—69%
Lp(a)—73%
The mean Lp(a) concentrations (the mean levels are a surrogate parameter for the
interval mean values; formula: mean = ½ × (pre value + post value); levels have
been separately reported according to the dimension provided by the lab): Group
B—54.20 mg/dL and 98.60 nmol/L, respectively; Group C—61.50 mg/dL and
104.75 nmol/L, respectively.
Mean LDL-C levels ranged between 2.00 mmol/L (Group A) and 1.59 mmol/L
(Group C). LDL-C levels for Group B are not given in detail.
CVEs have been subdivided into MACE [major adverse cardiac events: acute
coronary syndrome (unstable angina pectoris, NSTEMI, STEMI), coronary inter-
vention/surgery (PTCA, stent, CABG)] and MANCE [major adverse noncardiac
events: arterial occlusive disease (AOD) at noncoronary arteries with occlusion or
necessity for intervention/operation (PTA, stent, bypass, amputation), AOD of brain
arteries with TIA/stroke (CAOD), AOD of aorta thoracalis or aorta abdominalis
including visceral vessels and renal arteries, peripheral arterial occlusive disease
(PAOD)] (Table 23.4).
Follow-up data are contained in the annual report of the registry up to 7 years
under LA; low CVE rates were constantly seen throughout these years.
As an example, the graphs for HLP Group B are depicted (Fig. 23.4; with per-
mission of the Lipid-League).
Table 23.4 Percent reductions of MACE (major adverse cardiac events) and of MANCE (major
adverse noncardiac events) in the HLP Groups A, B, and C comparing the 2 years before the start
of LA with those during the first 2 years under LA (Schettler et al. 2020)
Fig. 23.4 MACE (a) and MANCE rates (b) in HLP Group B (with permission of Lipid-League).
(a) MACE rate (per patient and year) in HLP Group B. (b) MANCE rate (per patient and year) in
HLP Group B
390 U. Julius and S. Tselmin
Many patients had suffered from CVEs in the years anteceding the years included
in these graphs.
These data underline the already previously reported higher efficiency of LA
with respect to the reduction rate of CVEs in patients with elevated Lp(a) when
compared to patients without this feature.
The commonly available LA systems decrease both LDL-C and Lp(a) concentra-
tions. The Russian POCARD Ltd. company offers antibody-coated columns which
specifically decrease Lp(a) only (Pokrovsky et al. 2017; Safarova et al. 2013). A
slight decrease in LDL-C does not represent the removal of LDL but only the
removal of cholesterol in Lp(a) particles. In a prospectively carried out angiographic
study over 18 months, it could be shown that a weekly Lp(a) reduction was associ-
ated with decrease in the mean percent diameter stenosis by 5.05% and increase in
minimal lumen diameter by 14%; mean total atheroma volume was reduced by
4.60 mm3 (p < 0.05 for all). These data were compared to those seen in the control
group which was treated with atorvastatin only. This small study points to the effec-
tiveness of a specific elimination of Lp(a) as a tool to combat atherosclerotic lesions.
Unfortunately, these specific columns are not used in a large scale anywhere.
MultiSELECt Study
group with placebo “sham” sessions weekly for 3 months. Treatments were per-
formed using the DX21 DHP (Direct Hemo Perfusion) Lipoprotein Apheresis
machine (Kaneka Corporation, Osaka, Japan) with the Liposorber DL-75 column,
which uses dextran sulfate to covalently bind ApoB-containing lipoproteins.
Baseline tests were repeated after treatment periods for both groups. After a 1-month
washout period, patients crossed over to the opposite treatment arm.
Patients underwent cardiovascular magnetic resonance imaging at baseline
assessing quantitative first-pass stress/rest perfusion and assessment of carotid ath-
erosclerosis with measurement of total carotid wall volume. Patients had exercise
capacity tested using the Six Minute Walk test (6MWT) and assessment of their
angina symptoms using the Seattle Angina Questionnaire (SAQ) and quality of life
(QoL) with the SF-36 questionnaire.
The results indicate that an improvement in myocardial perfusion rate was pri-
marily driven by improvements in stress perfusion, with insignificant change in rest
perfusion. In terms of secondary endpoints, improvements with apheresis compared
with sham also occurred in carotid atherosclerotic burden as assessed by total
carotid wall volume (p < 0.001), exercise capacity measured by the 6MWT
(p = 0.001), four of five domains of the SAQ (all p < 0.02), and quality of life physi-
cal component summary assessed by the SF-36 survey (p = 0.001).
Some patients with elevated Lp(a) levels develop CVEs in several arterial regions.
We report two patients who were treated with LA at our center. Both suffered from
new CVEs despite being treated extracorporeally and were switched to two LA ses-
sions per week (Figs. 23.4 and 23.5).
Despite the intensive therapeutic regimen starting in 2018, the patient under-
went new interventions of her carotids, leg, and visceral arteries. Before being
referred to our center, she was smoking and it was not easy to persuade her to stop
this habit.
In Fig. 23.4, the lipid data are also shown. Lp(a) was effectively reduced, but in
all these years, LDL-C remained above the requested level (1.0 mmol/L). That is
why we started inclisiran in 04/2021. Since then, no new CVEs were observed. Her
actual lipid concentrations are as follows (before/after an LA session in 2022/02):
LDL-C 0.66/0.14 mmol/L, Lp(a) 153/26 nmol/L, HDL cholesterol
1.43/1.12 mmol/L, and triglycerides 1.68/0.56 mmol/L. Now LDL-C is optimal;
Lp(a) level is still high.
Another patient suffered from severe atherosclerosis of his coronaries
(Fig. 23.6).
At the age of 39 years, he had an acute myocardial infarction. His Lp(a) concen-
tration was found to be extremely high (≈593 nmol/L) only in 2006 (23 years after
his MI). He was also suffering from an increasing statin intolerance. From 1994 to
392 U. Julius and S. Tselmin
Fig. 23.5 A 56-year-old female patient with atherosclerotic affections of all vessel territories; LA
was started in 2017, switched to two sessions per week in 10/2018 (patient agreed that her data
could be included into this manuscript)
Fig. 23.6 A 77-year-old male patient with multiple events at his coronary arteries; LA was started
in 2012 and switched to two LA sessions per week in 2016 (patient agreed that his data could be
included into this manuscript)
(Tmoyan et al. 2020). In the group with severe carotid atherosclerosis, 16 patients
(24%) had ischemic stroke. Lp(a) concentration in these patients was higher 36 [20;
59] mg/dL than in the patients with isolated carotid atherosclerosis without stroke
15 [7; 54] mg/dL (p = 0.04).
All patients listed in Table 23.5 had an elevation of Lp(a) and a (mostly) mod-
est increase of LDL-C. Only in two patients atherosclerotic lesions of their carot-
ids were documented. In one patient, coronary atherosclerosis was observed.
One patient does not take a statin (statin intolerance, he started evolocumab
in 2019).
Our patients did not develop any further strokes after they started to be treated
with LA, though Lp(a) interval mean values were not optimal.
In the literature, a genetically lowered Lp(a) concentration predicted a decreased
risk of stroke (Kamstrup 2021). An elevated Lp(a) level was associated with unfa-
vorable functional outcomes in patients with ischemic stroke (Jiang et al. 2021).
Thus we are convinced that the lowering of Lp(a) levels by LA is a beneficial con-
tribution to the further follow-up situation of stroke patients.
394
Table 23.5 Male patients who are treated with LA because of an elevation of Lp(a) and following cerebrovascular events (patients agreed that their data could
be included into this manuscript)
LDL-C Lipoprotein(a)
Stroke events (mmol/L) (nmol/L)
Actual/
Before Before interval Statins: name/
ID Born Year Localization Atherosclerosis ApoE LA start LA Actual LA mean daily dose (mg)
RB 1967 2019 Left A. Cerebri Mild in Aa. carotis 3/3 08/04/20 2.0 1.7 363 321/236 A/80
nk Media Subcortical arteriosclerotic
Old lacunar encephalopathy
infarctions in white One-vessel-CAD, no
matter interventions
MG 1980 2017 Caput nuclei caudate, No 3/3 02/27/19 1.56 1.46 125 138/90 S/30
on the left side
HK 1962 2003 Cerebellum TEA in the left A. car int in 4/3 03/23/20 1.41 1.43 137 95/62 No
2007 Left A.cer media left 2007
2008 A.cer ant Actual—severe plaques Aa.
2009 Left A.cer ant parietal carotis without relevant stenosis
2009 operculum
F-PK 1968 2020 Cerebellum No 3/3 08/25/20 4.76 1.83 428 287/214 A/60
HS 1986 2018 Left A.cer media No 4/3 05/16/19 1.94 1.39 120 121/56 A/40
U-OS 1978 nk Cerebellum: small old No Nk 01/07/21 2.5 1.22 346 227/151 A/60
2020 defect
Vertebral-basilar TIA
JW 1980 nk Cerebellum: old No 3/3 01/28/20 3.21 2.3 152 122/87 R/10
2017 lacunar defect
Right basal ganglia
U. Julius and S. Tselmin
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 395
Unresolved Problems
Other differences between LA systems have been described, for example, for
proteins, PCSK9 levels, and coagulation factors (Julius et al. 2002, 2015a, b). The
significance of these differences for the prognosis of the patients remains still to be
clarified.
Moreover, additional pleiotropic effects of LA (removal of C-reactive protein,
complement, of apolipoprotein CIII, TNF-α, interleukin 6, and adhesion molecules
like ICAM-1 or VCAM-1) may have an impact on the course of atherosclerotic
lesions (Waldmann and Parhofer 2016; Makino et al. 2019). LA leads to vascular
tone reduction, reduced thrombogenesis, increased neo-angiogenesis, and impor-
tantly plaque stabilization (Poller et al. 2017). No data comparing different LA
methods with regard to these parameters have been published.
For LDL-C in the last years, target values have been defined with the aim to effec-
tively reduce the cardiovascular risk. The major message is “the lower the better.”
In the absence of randomized, controlled trial data demonstrating reduced car-
diovascular risk with reduction in Lp(a), no such targets have been proclaimed for
this parameter.
Usually, in lab reports, a normal range for Lp(a) is given below 30 mg/dL (about
75 nmol/L). It has been discussed in a paper by Boffa et al. that lowering Lp(a)
below this threshold would ameliorate the atherogenic risk (Boffa et al. 2018).
We think that in order to obtain an optimal effect of LA therapy with respect to
CVEs, interval mean values should be normalized [probably below 30 mg/dL (about
75 nmol/L)]. As shown in Fig. 23.1, the reality is far from this request. In only 21%
of our patients, this goal was reached. About half of them had an interval mean
Lp(a) concentration higher than 120 nmol/L. Though as a matter of fact, we did not
see a relationship between these concentrations and the incidence of CVEs during
LA therapy (Julius et al. 2020). And in the Russian prospective study, using specific
anti-Lp(a) columns, with coronary angiography, a beneficial effect on coronary ath-
erosclerosis was observed, though the mean interval value in the apheresis group
was 73 mg/dL (about 175 nmol/L) (Pokrovsky et al. 2020).
In the literature, two papers suggested, based on data with Mendelian randomiza-
tion, that a decrease of Lp(a) by about 100 mg/dL (about 240 nmol/L) (Burgess
et al. 2018) or 65 mg/dL (about 156 nmol/L) (Lamina et al. 2019) will induce a
similar reduction of CVEs as a decrease of 1 mmol/L of LDL-C, for example, by
about 22%. Populations included into these meta-analyses had much lower median
Lp(a) concentrations (approximately 30 nmol/L, maximally in one study
104 nmol/L) than those who are usually treated extracorporeally. Moreover, both
studies were population based. In contrast, a Danish group looked at patients with a
history of cardiovascular disease who were followed after their initial event (Madsen
et al. 2020). The authors calculated that plasma Lp(a) should be lowered by 50
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 397
(about 120 nmol/L) and 99 mg/dL (about 240 nmol/L) for 5 years to achieve 20%
and 40% MACE risk reduction in secondary prevention. Accordingly, for a 22%
MACE reduction, a reduction of Lp(a) by 55 mg/dL (about 132 nmol/L) would be
required.
From the viewpoint of LA data, these publications are not conclusive at all.
In the HPS2-THRIVE Study, niacin laropiprant reduced mean Lp(a) by
12 nmol/L overall and by 34 nmol/L in the top quintile by baseline Lp(a) level
≥128 nmol/L (Parish et al. 2018). The authors write that estimates from genetic
studies suggest that these Lp(a) reductions during the short term of the trial might
yield proportional reductions in coronary risk of ≈2% overall and 6% in the top
quintile by Lp(a) levels.
In studies using PCSK9 antibodies (evolocumab, alirocumab), a small decrease
of Lp(a) concentrations was seen (Kronenberg 2022; Julius et al. 2019). When
excluding the impact of the reduction of LDL-C on outcome data by mathematical
modeling, the decrease of Lp(a) was effective with respect to a certain lowering of
CVEs when compared with the placebo groups.
A comparison between PCSK9 antibodies and pelacarsen [an antisense oligo-
nucleotide against Apo(a)] showed an interesting difference: Pelacarsen reduced
Lp(a) by 47% and as a consequence the pro-inflammatory gene expression in mono-
cytes of cardiovascular disease patients with elevated Lp(a), which coincided with a
functional reduction in transendothelial migration capacity of monocytes ex vivo
(Stiekema et al. 2020). In contrast, PCSK9 antibody treatment lowered Lp(a) by
16% and did not alter transcriptome nor functional properties of monocytes, despite
an additional reduction of 65% in low-density lipoprotein cholesterol (LDL-C). The
effect of Lp(a) lowering by LA is in the same range as described for pelacarsen in
this manuscript.
When LDL-C is measured, both LDL and Lp(a) particles are included (Yeang et al.
2015). In order to calculate the LDL-C mass transported with LDL, the following
steps are required: (1) The Lp(a) mass should be in mg/dL—we measure Lp(a) in
nmol/L—the conversion into the Lp(a) mass is not correct (and no longer recom-
mended). (2) The estimated percentage of LDL-C in the Lp(a) particles usually is
set to be 30%. Data have shown that this percentage may be variable interindividu-
ally. (3) Due to these problems, in some patients, negative “true” LDL-C levels are
seen. The British colleagues do not recommend to calculate “true” LDL-C because
(1) it is not validated with isoform-independent assays in treated and untreated
patients, (2) it is not validated in large epidemiological studies for cardiovascular
risk prediction or in RCTs of lipid-lowering therapies, and (3) it is not in clinical use
(Cegla et al. 2019). Recently, a novel method for quantification of Lp(a) cholesterol
had been suggested (Yeang et al. 2021). This problem is relevant for patients who
are treated with LA and with PCSK9 inhibitors.
398 U. Julius and S. Tselmin
LA allows the treatment of high-risk patients with elevated Lp(a) levels who have
suffered from CVEs. It is tolerated very well. In patients with familial hypercholes-
terolemia, which is not seldom associated with elevated Lp(a) levels, tendon xan-
thomas usually disappear under a year-long LA therapy. This points to the fact that
the body cholesterol pool is diminished.
The extracorporeal removal of Lp(a) particles significantly decreases their con-
centration in blood, especially in the days immediately after LA sessions. The
increase of Lp(a) thereafter makes it necessary to perform LA sessions weekly. In
some countries, a 2-week interval is the prevailing therapeutic approach, mainly
because of financial problems.
Interval mean values reflect approximately the averaged Lp(a) level in the days
between LA sessions and in this way the atherogenic burden. With the available LA
methods, it is possible to reach optimal Lp(a) concentrations (<75 nmol/L) only in
a small part of the patients. But extremely high Lp(a) levels which confer a very
high atherogenic risk can be reduced a lot.
In LA patients, all other risk factors (hypertension, diabetes mellitus, hyperurice-
mia, smoking habit, hypothyroidism, obesity) should be optimized.
The usual way to describe the effect of LA therapy on outcome data—by com-
paring the incidences before the start of the extracorporeal treatment with those
during this treatment—has been criticized (Waldmann and Parhofer 2016). It should
be noted that the observational studies suffer from potential confounding due to the
selection bias for survivors inherent in their design, as well as the lack of the ability
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 399
to rule out the effect of apheresis on other drivers of events such as fibrinogen (Boffa
et al. 2018). Hopefully, the MultiSELECt study will offer a clarification of this dis-
pute. Clearly, placebo-controlled apheresis studies are not feasible. The British
study which had an individual sham control lasted only a few months.
Under extracorporeal therapy, some patients will develop new CVEs. We did not
detect any difference in lipid concentrations, including Lp(a), before or after LA
sessions or in interval mean values between patients with or without CVEs during
LA therapy (Julius et al. 2020). According to our data, older age at the start of the
LA therapy and a higher number of CVEs before LA started playing a role. Both
these aspects point to the fact that atherosclerosis has progressed in patients who
suffer from CVEs during the extracorporeal therapy.
Thus, in order to be on the safe side, an LA therapy should not be initiated too
late. But on the other hand, LA may be lifesaving in high-risk patients. The number
of patients who die when they are undergoing LA therapy is rather low; no reliable
data on these numbers are available.
In general, an LA therapy should not be started in patients who are (biologically)
older than 70 years. On the other hand, LA is a lifelong treatment and should not be
discontinued even in very old patients who started LA years ago. The diagnosis of a
malignant tumor, a severe cardiac insufficiency, or a poor compliance may be rea-
sons for stopping the extracorporeal therapy.
In the British sham-controlled apheresis study, a regression of atherosclerosis
was seen at the neck vessels. In some angiographic studies, a certain percentage of
regression at the coronary arteries was found in LA-treated patients. In our experi-
ence, we are already happy when in a given patient a nonprogression of the lesions
(no new stenoses, no new CVEs) is observed. Of course, patients should regularly
be checked by a cardiologist and/or angiologist.
LA is expensive and time-consuming (2–3 h) and needs the work of a qualified
staff. But by avoiding new CVEs, money can be saved in the long run. In Germany,
nephrologists may apply for the permission to perform LA. Taking into attention the
fact that LA can be optimally performed only in centers with sufficient experience
in this field, the number of patients at each center should not be too low (probably
not less than ten patients, at least two LA systems should be offered).
If in a given patient with elevated Lp(a) concentration the LDL-C level exceeds
the internationally accepted targets (1.4 mmol/L for high-risk patients, 1.0 mmol/L
for patients with repeated CVEs) (Mach et al. 2019), usually a statin therapy should
be started. More effective statins (atorvastatin, rosuvastatin) are to be preferred.
When the effect is not satisfactory, ezetimibe can be added. Bempedoic acid can be
administered either together with a statin (in order to improve LDL-C) or instead of
a statin when the latter is not tolerated. Ezetimibe can be continued in these cases.
When the result seen after the introduction of these first steps is not optimal, after
several months, the indication to use PCSK9 inhibitors is given.
All these measures are the prerequisite before an LA is allowed to be started, at
least in Germany.
An LA therapy may be commenced in the following LDL-C ranges in patients
with Lp(a) concentrations exceeding 60 mg/dL or 120 nmol/L:
1. The LDL-C target has been reached. That is the purpose of the official
regulations.
2. The LDL-C target was not reached despite the patient regularly took the lipid-
lowering drugs. This indication is not officially covered by the existing rules, but
this situation is not a very rare one.
3. Patients experience a drug intolerance—this may be the case with statins or
PCSK9 inhibitors, very seldom with ezetimibe. LDL-C is still too high.
Of course, a progression of atherosclerosis has to have been documented
(repeated CVEs or shown by imaging techniques). Exceptions from this demand are
made in very young patients (aged under 40 years) with extremely high Lp(a) levels
who survived a life-threatening acute myocardial infarction and who have a positive
family history for CVEs among first-degree relatives in younger ages.
During the LA therapy, the administration of lipid-lowering therapy should
always be continued. Some adjustment of doses may be necessary depending on the
measured LDL-C level.
In our hands, the addition of PCSK9 inhibitors to LA procedures may further
improve LDL-C and—at least in a majority of patients—Lp(a) concentrations. The
combined treatment with PCSK9 monoclonal antibodies and apheresis may be pref-
erable in certain hypercholesterolemic patients with high Lp(a), because of the com-
bined benefits of both approaches in lowering LDL-C, triglyceride-rich lipoproteins,
inflammation, hemorheology, and Lp(a) (Ruscica et al. 2019).
We saw the end of a series of cardiovascular interventions in high-risk patients
after the start of this intensive injection therapy. In some patients, we then switched
to a biweekly LA regimen—provided the cardiovascular situation remains stable.
Evinacumab, a fully human monoclonal antibody against angiopoietin-like 3, is
a new drug which can be applied in therapy-resistant hypercholesterolemia
(Rosenson et al. 2020). But this drug does not decrease Lp(a). In an ApoE*3-Leiden
CETP mouse model, a triple therapy with atorvastatin, alirocumab, and evinacumab
has been successfully performed (Pouwer et al. 2020). The future role of intrave-
nously infused evinacumab in the daily routine has still to be defined.
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 401
Conclusions
LA is at present the only accepted therapy to decrease highly elevated Lp(a) concen-
trations in high-risk patients with the aim to revert a progressive course of Lp(a)-
associated cardiovascular disease to a stable course and to prevent future CVEs.
Most probably, pleiotropic (anti-inflammatory, antithrombotic, rheologic) effects
exert an additional benefit. Up to now only observational studies documented a high
efficiency of LA with respect to reduction of the incidence of CVEs. LA requires a
402 U. Julius and S. Tselmin
References
Arai K, Orsoni A, Mallat Z, Tedgui A, Witztum JL, Bruckert E, et al. Acute impact of apher-
esis on oxidized phospholipids in patients with familial hypercholesterolemia. J Lipid Res.
2012;53(8):1670–8. https://doi.org/10.1194/jlr.P027235.
Arnold M, Schweizer J, Nakas CT, Schutz V, Westphal LP, Inauen C, et al. Lipoprotein(a)
is associated with large artery atherosclerosis stroke aetiology and stroke recurrence
among patients below the age of 60 years: results from the BIOSIGNAL study. Eur Heart
J. 2021;42(22):2186–96. https://doi.org/10.1093/eurheartj/ehab081.
Bigazzi F, Sbrana F, Berretti D, Maria Grazia Z, Zambon S, Fabris A, et al. Reduced incidence of
cardiovascular events in hyper-Lp(a) patients on lipoprotein apheresis. The G.I.L.A. (Gruppo
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 403
Julius U, Tselmin S, Schatz U, Fischer S, Bornstein SR. Lipoprotein(a) and proprotein convertase
subtilisin/kexin type 9 inhibitors. Clin Res Cardiol Suppl. 2019;14(Suppl 1):45–50. https://doi.
org/10.1007/s11789-019-00099-z.
Julius U, Kuss S, Tselmin S, Schatz U, Bornstein SR. Why some patients undergoing lipoprotein
apheresis therapy develop new cardiovascular events? J Cardiovasc Dev Dis. 2020;7(3):30025.
https://doi.org/10.3390/jcdd7030025.
Kamstrup PR. Lipoprotein(a) and cardiovascular disease. Clin Chem. 2021;67(1):154–66. https://
doi.org/10.1093/clinchem/hvaa247.
Kassenärztliche Bundesvereinigung. Qualitätsbericht—Ausgabe 2014—Berichtsjahr 2013.
Special edition. 2014. www.kbv.de.
Kassenärztliche Bundesvereinigung. Qualitätsbericht—Ausgabe 2015—Berichtsjahr 2014.
Special edition. 2015. www.kbv.de.
Kassenärztliche Bundesvereinigung. Qualitätsbericht—Ausgabe 2016—Berichtsjahr 2015.
Special edition. 2016. www.kbv.de.
Kassenärztliche Bundesvereinigung. Qualitätsbericht—Ausgabe 2017—Berichtsjahr 2016.
Special edition. 2017. www.kbv.de.
Kassenärztliche Bundesvereinigung. Qualitätsbericht—Ausgabe 2018—Berichtsjahr 2017.
Special edition. 2018. www.kbv.de.
Kassenärztliche Bundesvereinigung. Qualitätsbericht—Ausgabe 2019—Berichtsjahr 2018.
Special edition. 2019. www.kbv.de.
Kassenärztliche Bundesvereinigung. Qualitätsbericht—Ausgabe 2020—Berichtsjahr 2019.
Special edition. 2020. www.kbv.de.
Kassenärztliche Bundesvereinigung. Qualitätsbericht—Ausgabe 2021—Berichtsjahr 2020.
Special edition. 2021. www.kbv.de.
Khan TZ, Hsu LY, Arai AE, Rhodes S, Pottle A, Wage R, et al. Apheresis as novel treatment
for refractory angina with raised lipoprotein(a): a randomized controlled cross-over trial. Eur
Heart J. 2017;38(20):1561–9. https://doi.org/10.1093/eurheartj/ehx178.
Klingel R, Heigl F, Schettler V, Roeseler E, Grutzmacher P, Hohenstein B, et al. Lipoprotein(a)—
marker for cardiovascular risk and target for lipoprotein apheresis. Atheroscler Suppl.
2019;40:17–22. https://doi.org/10.1016/j.atherosclerosissup.2019.08.037.
Koren MJ, Moriarty PM, Baum SJ, Neutel J, Hernandez-Illas M, Weintraub HS, et al. Preclinical
development and phase 1 trial of a novel siRNA targeting lipoprotein(a). Nat Med.
2022;28(1):96–103. https://doi.org/10.1038/s41591-021-01634-w.
Korneva VA, Kuznetsova TY, Julius U. Modern approaches to lower lipoprotein(a) concentra-
tions and consequences for cardiovascular diseases. Biomedicine. 2021;9(9):1271. https://doi.
org/10.3390/biomedicines9091271.
Kronenberg F. Lipoprotein(a). Handb Exp Pharmacol. 2022;270:201–32. https://doi.
org/10.1007/164_2021_504.
Kroon AA, van’t Hof MA, Demacker PN, Stalenhoef AF. The rebound of lipoproteins after
LDL-apheresis. Kinetics and estimation of mean lipoprotein levels. Atherosclerosis.
2000;152(2):519–26. https://doi.org/10.1016/s0021-9150(00)00371-3.
Lamina C, Kronenberg F, for the Lp(a)-GWAS-Consortium. Estimation of the required
lipoprotein(a)-lowering therapeutic effect size for reduction in coronary heart disease out-
comes: a Mendelian randomization analysis. JAMA Cardiol. 2019;4(6):575–9. https://doi.
org/10.1001/jamacardio.2019.1041.
Langsted A, Nordestgaard BG. Lipoprotein(a) as part of the diagnosis of clinical familial
hypercholesterolemia. Curr Atheroscler Rep. 2022;24(4):289–96. https://doi.org/10.1007/
s11883-022-01002-0.
Leebmann J, Roeseler E, Julius U, Heigl F, Spitthoever R, Heutling D, et al. Lipoprotein apheresis in
patients with maximally tolerated lipid-lowering therapy, lipoprotein(a)-hyperlipoproteinemia,
and progressive cardiovascular disease: prospective observational multicenter study. Circulation.
2013;128(24):2567–76. https://doi.org/10.1161/CIRCULATIONAHA.113.002432.
Ma L, Waldmann E, Ooi EMM, Chan DC, Barrett HPR, Watts GF, et al. Lipoprotein(a) and low-
density lipoprotein apolipoprotein B metabolism following apheresis in patients with elevated
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 405
lipoprotein(a) and coronary artery disease. Eur J Clin Investig. 2019;49(2):e13053. https://doi.
org/10.1111/eci.13053.
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, et al. 2019 ESC/EAS
Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular
risk. Eur Heart J. 2019;41:111–88. https://doi.org/10.1093/eurheartj/ehz455.
Madsen CM, Kamstrup PR, Langsted A, Varbo A, Nordestgaard BG. Lipoprotein(a)-lowering by
50 mg/dL (105 nmol/L) may be needed to reduce cardiovascular disease 20% in secondary pre-
vention: a population-based study. Arterioscler Thromb Vasc Biol. 2020;40(1):255–66. https://
doi.org/10.1161/ATVBAHA.119.312951.
Makino H, Koezuka R, Tamanaha T, Ogura M, Matsuki K, Hosoda K, et al. Familial hypercho-
lesterolemia and lipoprotein apheresis. J Atheroscler Thromb. 2019;26(8):679–87. https://doi.
org/10.5551/jat.RV17033.
Mickiewicz A, Marlega J, Kuchta A, Bachorski W, Cwiklinska A, Raczak G, et al. Cardiovascular
events in patients with familial hypercholesterolemia and hyperlipoproteinaemia(a): indications
for lipoprotein apheresis in Poland. J Clin Apher. 2021;36(3):370–8. https://doi.org/10.1002/
jca.21872.
Moriarty PM, Gray JV, Gorby LK. Lipoprotein apheresis for lipoprotein(a) and cardiovascular
disease. J Clin Lipidol. 2019;13(6):894–900. https://doi.org/10.1016/j.jacl.2019.09.010.
Nave AH, von Eckardstein A. Is lipoprotein(a) a risk factor for ischemic stroke and venous throm-
boembolism? Clin Res Cardiol Suppl. 2019;14(Suppl 1):28–32. https://doi.org/10.1007/
s11789-019-00101-8.
Nordestgaard BG, Chapman MJ, Ray K, Boren J, Andreotti F, Watts GF, et al. Lipoprotein(a)
as a cardiovascular risk factor: current status. Eur Heart J. 2010;31(23):2844–53. https://doi.
org/10.1093/eurheartj/ehq386.
O'Donoghue ML, Fazio S, Giugliano RP, Stroes ESG, Kanevsky E, Gouni-Berthold I, et al.
Lipoprotein(a), PCSK9 inhibition, and cardiovascular risk. Circulation. 2019;139(12):1483–92.
https://doi.org/10.1161/CIRCULATIONAHA.118.037184.
Parish S, Hopewell JC, Hill MR, Marcovina S, Valdes-Marquez E, Haynes R, et al. Impact of
apolipoprotein(a) isoform size on lipoprotein(a) lowering in the HPS2-THRIVE study. Circ
Genomic Precis Med. 2018;11(2):e001696. https://doi.org/10.1161/CIRCGEN.117.001696.
Pokrovsky SN, Afanasieva OI, Safarova MS, Balakhonova TV, Matchin YG, Adamova IYU, et al.
Specific Lp(a) apheresis: a tool to prove lipoprotein(a) atherogenicity. Atheroscler Suppl.
2017;30:166–73. https://doi.org/10.1016/j.atherosclerosissup.2017.05.004.
Pokrovsky SN, Afanasieva OI, Ezhov MV. Therapeutic apheresis for management of Lp(a)
hyperlipoproteinemia. Curr Atheroscler Rep. 2020;22(11):68. https://doi.org/10.1007/
s11883-020-00886-0.
Poller WC, Berger A, Dreger H, Morgera S, Enke-Melzer K. Lipoprotein apheresis in patients
with peripheral artery disease and lipoprotein(a)-hyperlipoproteinemia: 2-year follow-up of a
prospective single center study. Atheroscler Suppl. 2017;30:174–9. https://doi.org/10.1016/j.
atherosclerosissup.2017.05.007.
Pouwer MG, Pieterman EJ, Worms N, Keijzer N, Jukema JW, Gromada J, et al. Alirocumab,
evinacumab, and atorvastatin triple therapy regresses plaque lesions and improves lesion com-
position in mice. J Lipid Res. 2020;61(3):365–75. https://doi.org/10.1194/jlr.RA119000419.
Reyes-Soffer G, Ginsberg HN, Berglund L, Duell PB, Heffron SP, Kamstrup PR, et al.
Lipoprotein(a): a genetically determined, causal, and prevalent risk factor for atherosclerotic
cardiovascular disease: a scientific statement from the American Heart Association. Arterioscler
Thromb Vasc Biol. 2022;42(1):e48–60. https://doi.org/10.1161/ATV.0000000000000147.
Roeseler E, Julius U, Heigl F, Spitthoever R, Heutling D, Breitenberger P, et al. Lipoprotein apher-
esis for lipoprotein(a)-associated cardiovascular disease: prospective 5 years of follow-up and
Apo(a) characterization. Arterioscler Thromb Vasc Biol. 2016;36(9):2019–27. https://doi.
org/10.1161/ATVBAHA.116.307983.
Rosenson RS, Burgess LJ, Ebenbichler CF, Baum SJ, Stroes ESG, Ali S, et al. Evinacumab in
patients with refractory hypercholesterolemia. N Engl J Med. 2020;383(24):2307–19. https://
doi.org/10.1056/NEJMoa2031049.
406 U. Julius and S. Tselmin
Ruscica M, Watts GF, Sirtori CR. PCSK9 monoclonal antibodies and lipoprotein apheresis for
lowering lipoprotein(a): making choices in an era of RNA-based therapies. Eur J Prev Cardiol.
2019;26(9):998–1000. https://doi.org/10.1177/2047487319833504.
Safarova MS, Ezhov MV, Afanasieva OI, Matchin YG, Atanesyan RV, Adamova IY, et al. Effect
of specific lipoprotein(a) apheresis on coronary atherosclerosis regression assessed by quan-
titative coronary angiography. Atheroscler Suppl. 2013;14(1):93–9. https://doi.org/10.1016/j.
atherosclerosissup.2012.10.015.
Schatz U, Tselmin S, Muller G, Julius U, Hohenstein B, Fischer S, et al. Most significant reduc-
tion of cardiovascular events in patients undergoing lipoprotein apheresis due to raised Lp(a)
levels—a multicenter observational study. Atheroscler Suppl. 2017;30:246–52. https://doi.
org/10.1016/j.atherosclerosissup.2017.05.047.
Schettler V, Roeseler E, Thode C, Gruetzmacher P, Klingel R, Julius U. Differenzierung der
Indikation zur Lipoproteinapherese bei erhöhtem Lp(a) durch unterschiedliche Messmethoden.
Cardiovasc. 2015;15:41–3.
Schettler V, Bernhardt W, van Buuren F, Gruetzmacher P, Heigl F, Hohenstein B, et al. The
German lipoprotein apheresis registry (GLAR)—almost 5 years on. Clin Res Cardiol Suppl.
2020;12(1):44–9.
Stefanutti C, Morozzi C, Di GS. Italian multicenter study on low-density lipoprotein apher-
esis working group 2009 survey. Ther Apher Dial. 2013;17(2):169–78. https://doi.
org/10.1111/j.1744-9987.2012.01142.x.
Stefanutti C, Pisciotta L, Favari E, Di Giacomo S, Vacondio F, Zenti MG, et al. Lipoprotein(a)
concentration, genetic variants, apo(a) isoform size, and cellular cholesterol efflux in patients
with elevated Lp(a) and coronary heart disease submitted or not to lipoprotein apheresis: an
Italian case-control multicenter study on Lp(a). J Clin Lipidol. 2020;14(4):487–97.e1. https://
doi.org/10.1016/j.jacl.2020.05.002.
Stiekema LCA, Prange KHM, Hoogeveen RM, Verweij SL, Kroon J, Schnitzler JG, et al. Potent
lipoprotein(a) lowering following apolipoprotein(a) antisense treatment reduces the pro-
inflammatory activation of circulating monocytes in patients with elevated lipoprotein(a). Eur
Heart J. 2020;41(24):2262–71. https://doi.org/10.1093/eurheartj/ehaa171.
Szarek M, Bittner VA, Aylward P, Baccara-Dinet M, Bhatt DL, Diaz R, et al. Lipoprotein(a)
lowering by alirocumab reduces the total burden of cardiovascular events independent of
low-density lipoprotein cholesterol lowering: ODYSSEY OUTCOMES trial. Eur Heart
J. 2020;41(44):4245–55. https://doi.org/10.1093/eurheartj/ehaa649.
Thompson GR. The scientific basis and future of lipoprotein apheresis. Ther Apher Dial.
2022;26(1):32–6. https://doi.org/10.1111/1744-9987.13716.
Thompson G, Parhofer KG. Current role of lipoprotein apheresis. Curr Atheroscler Rep.
2019;21(7):26. https://doi.org/10.1007/s11883-019-0787-5.
Tmoyan NA, Ezhov MV, Afanasieva OI, Klesareva EA, Afanasieva MI, Balakhonova TV, et al.
Association of lipoprotein(a) with ischemic stroke and stenotic carotid atherosclerosis.
Zh Nevrol Psikhiatr Im S Korsakova. 2020;120(3. Vyp. 2):42–8. https://doi.org/10.17116/
jnevro202012003242.
Trenkwalder E, Gruber A, Konig P, Dieplinger H, Kronenberg F. Increased plasma concentrations of
LDL-unbound apo(a) in patients with end-stage renal disease. Kidney Int. 1997;52(6):1685–92.
https://doi.org/10.1038/ki.1997.503.
Tselmin S, Muller G, Schatz U, Julius U, Bornstein SR, Hohenstein B. Kinetics of lipoprotein(a) in
patients undergoing weekly lipoprotein apheresis for Lp(a) hyperlipoproteinemia. Atheroscler
Suppl. 2017;30:209–16. https://doi.org/10.1016/j.atherosclerosissup.2017.05.033.
Tsimikas S. A test in context: lipoprotein(a): diagnosis, prognosis, controversies, and emerging
therapies. J Am Coll Cardiol. 2017;69(6):692–711. https://doi.org/10.1016/j.jacc.2016.11.042.
Tsimikas S. Potential causality and emerging medical therapies for lipoprotein(a) and its associ-
ated oxidized phospholipids in calcific aortic valve stenosis. Circ Res. 2019;124(3):405–15.
https://doi.org/10.1161/CIRCRESAHA.118.313864.
23 Lipoprotein Apheresis for Reduction of Lipoprotein(a) 407
Cardiovascular diseases (CVDs) are the leading cause of death globally (World
Health Organization 2021). Lp(a), pronounced “Lp little a,” is an LDL (low-density
lipoprotein)-like, fatty, sticky lipoprotein particle with an additional protein
apolipoprotein(a) [apo(a)] wrapped around it and found in blood serum. It is an
inherited atherogenic lipoprotein and an independent risk factor for atherosclerotic
cardiovascular disease, vascular thrombosis, stroke, and calcific aortic stenosis
(Nordestgaard et al. 2010; Bennet et al. 2088; Erqou et al. 2009; Kampstrup et al.
2009; Boffa and Koschinsky 2016; Rogers and Aikawa 2015; Langsted et al. 2019).
Lp(a) is one of the strongest genetically determined risk factors for cardiovascular
disease (Kronenberg and Utermann 2013; CARDIoGRAMplusC4D Consortium
and Deloukas 2013; Thanassoulis et al. 2013). Approximately 1 in 5 people have
inherited high Lp(a), more than 1 billion people globally and 63 million in the
United States who are unaware they have up to a 60% increased risk for coronary
artery disease (Nordestgaard et al. 2010; Kamstrup et al. 2009). High Lp(a) is
80–90% genetically determined (Schmidt et al. 2016). Diet and exercise have little
to no impact on high Lp(a) (Mackinnon et al. 1997). Most people fully express the
LPA gene by the time they are 2 years old, reach adult levels by five, and for the
most part maintain the same Lp(a) levels for a lifetime, although Lp(a) levels tend
to increase with age in females after menopause (Wilson et al. 2019; Bittner 2002).
Unfortunately, there are no visible symptoms, such as xanthomas, to indicate high
Lp(a), and traditional cholesterol tests miss 8% of people who have a cardiovascular
event whose only risk factor is high Lp(a) (Mortensen et al. 2015; Bittner 2015).
S. R. Tremulis (*)
Redwood City, CA, USA
The traditional cardiovascular lipid screening does not include high Lp(a). However,
a simple blood test performed once in a person’s lifetime could be the first step in
preventing up to 120,000 cardiovascular events every year (Wilson et al. 2019;
Mortensen et al. 2015). Unfortunately, the first sign of cardiovascular disease often
is a heart attack or a stroke. In 2003, I almost died of a heart attack despite having a
healthy lifestyle, annual preventative health screenings, and no significant risk fac-
tors except my family history of cardiovascular disease. Bloodwork ordered after
my heart event identified high Lp(a) as the only potentially significant contributing
risk factor. I, therefore, propose expanding the current standard cardiovascular pre-
vention lipid screening panel to include high Lp(a) testing for everyone to provide
patients, families, and their healthcare providers with a more accurate prediction of
their overall risk for premature cardiovascular disease and death (Wilson et al. 2019;
Mortensen et al. 2015; Bittner 2015).
eveal Lp(a): “You Must Have Had Some Really Bad Habits
R
When You Were Younger”
Heart disease is the leading cause of death for men, women, and people of most
racial and ethnic groups in the United States (Table 24.1) (Centers for Disease
Control and Prevention 2018) and costs the United States about $363 billion each
year from 2016 to 2017 (Virani et al. 2021). This includes the cost of healthcare
services, medicines, and lost productivity due to death. There is no cure for cardio-
vascular disease; it is a chronic, systemic disease.
These daunting heart disease statistics have provoked fear in me for decades. My
father died young of a heart attack; he had his first heart event at age 30, femoral
bypass surgery at age 40, and a fatal heart attack at age 50. I was 22 years of age
when he died. I never got over it; I just adjusted to it. Do you have a family history
of cardiovascular disease? What are your personal inherited cardiometabolic risk
factors for cardiovascular disease? I thought, heart attack, it was never going to
Table 24.1 2020 Top causes of death in the United States—Centers for Disease Control
2020 Top causes of death in the United States—Centers for Disease Control
Heart disease: 696,962
Cancer: 602,350
COVID-19: 350,831
Accidents: 200,955
Stroke (cerebrovascular diseases): 160,264
Alzheimer’s disease: 134,242
The table was created from data in Mortality in the United States, 2020, data table for Figure 4
Murphy SL, Kochanek KD, Xu JQ, Arias E. Mortality in the United States, 2020. NCHS Data
Brief, no 427. Hyattsville, MD: National Center for Health Statistics. 2021. DOI: https://doi.
org/10.15620/cdc:112079. Copyright 2020 CDC/National Center for Health Statistics
24 Elevated Lp(a): Why Should I Test For It, If I Cannot Treat It? A Patient’s Perspective 411
happen to me. I was a young female, and I thought heart disease primarily impacted
older men, like my father. I was in the medical device industry, knowledgeable
about heart disease, and working on cutting-edge technology to help families faced
with a cardiovascular disease diagnosis and helping to make a difference. I was
proactive about my cardiovascular health. Because of my family history, I had a
lifelong commitment to fitness and health, never missing my annual checkups. I
thought I was doing everything right, and according to my Framingham and
Reynold’s risk score, I was!
Table 24.2 below is my Framingham Risk Score from 2003; it gave me a 1%
chance of having a cardiovascular event.
Furthermore, Table 24.3 is my Reynolds Risk Score, an assessment that predicts
cardiovascular disease, gave me a 1% chance of having a cardiovascular event, and
includes family history in their risk calculation algorithm.
However, based on the Bruneck Study, if you add Lp(a) to the Framingham and
Reynolds Risk Score, I would have been reclassified with my high Lp(a) into a
higher risk category like 20% of patients (Willeit et al. 2014).
In 2003, at 39 years of age, I went out for my usual 5-mile run, got one block
down the road, and physically felt I could not go any further. I experienced fatigue
and mild tingling in my chest upon exertion. Even still, like many people, I rational-
ized away my symptoms. I taught fitness classes as a hobby and had run a marathon
the year before. I thought my tiredness was due to a potential thyroid issue, early
Table 24.2 Framingham Risk Score including Sandra Revill’s data from 2003
Information about your risk score
Age 39
Gender Female
Total 136 mg/dL (3.52 mm/L)
cholesterol
HDL 30 mg/dL (0.77 mm/L)
cholesterol
Smoker No
Systolic blood 117 mm/Hg
pressure
On medication No
for HBP
Risk scorea Less than 1%
The score means less than 1 in 100 people with this level of risk will have a
heart attack in the next 10 years
a
Your risk score was calculated using an equation. Other NCEP products, such
as printed ATP III materials, use a point system to determine a risk score close
to the equation score
The table was created from the Framingham Heart Study https://www.framinghamheartstudy.org/
fhs-risk-functions/cardiovascular-disease-10-year-risk/using the 2003 personal medical data from
Sandra Revill. Copyright for the Framingham Risk Score Calculator, D’agostino RB, Vasan RS,
Pencina MJ, Wolf PA, Cobain M, Massaro JM, Kannel WB. General cardiovascular risk profile for
use in primary care. Circulation. 2008 Feb 12;117:743–53. PMID:18212285
412 S. R. Tremulis
Table 24.3 Reynold’s Risk Score including Sandra Revill’s data from 2003
Information about your risk score
Age 39a
Gender Female
Total cholesterol 136 mg/dL (3.52 mm/L)
HDL cholesterol 30 mg/dL (0.77 mm/L)
Smoker No
Systolic blood pressure 117 mm/Hg
High-sensitivity C-reactive 0.16 mg/L
protein (hsCRP)
Did your mother or father Yes
have a heart attack before age
60?
Risk score a
Note the value you entered for age is outside the lower range.
The result is based on age 45
As shown in the graph below, age 45, your chance of having a
heart attack, stroke, or other heart disease event at some point in
the next 10 years is 1%
a
The table was created from the Reynolds Risk Calculator, Calculating Heart and Stroke Risk for
Men and Women http://www.reynoldsriskscore.org/Default.aspx using the 2003 personal medical
data from Sandra Revill. Copyright for Reynolds Risk Calculator, Journal of the American Medical
Association (Ridker PM, Buring JE, Rifai N, Cook NR. Development and validation of improved
algorithms for the assessment of global cardiovascular risk in women: The Reynolds Risk Score.
JAMA 2007;297:611–619)
menopause, or the flu. It never occurred to me that I might have a heart problem.
However, I knew something was wrong, so I scheduled an appointment with my
family practice physician, who had a comprehensive overview of my family history
of cardiovascular disease. The physician I saw wanted me to have a stress test in the
emergency room, but they were too busy that day. Not thinking it was emergent, he
suggested I was doing too much and recommended scheduling a treadmill test with
the cardiology group. A couple of weeks later, I passed the treadmill test and got
approval to teach my indoor cycling class that evening. Nevertheless, something
was still wrong; I had to stop riding three times during class due to an overwhelming
feeling of fatigue.
I still thought I had the flu! Later that week, I left on a business trip to Washington,
DC, to attend a major medical device conference with all the top cardiologists in the
world. I exercised in the hotel, as I usually did, but this time I felt terrible. I had the
same fatigue and mild tingling in my chest as I ran on the treadmill, and when I
slowed down, the tingling went away. I had swollen feet, so I started to take aspirin
to try and reduce the swelling in my feet. I was aware of the risk of blood clots when
flying long distances and that aspirin could help reduce that risk.
After I arrived home, compelled to seek a second opinion, I referred myself to an
interventional cardiologist I knew in my professional role as a product marketing
manager in the vascular business of a major medical device company. He listened to
my history and informed me that he would be conservative and order a Nuclear
24 Elevated Lp(a): Why Should I Test For It, If I Cannot Treat It? A Patient’s Perspective 413
Stress Test because of my family history and symptoms. He felt I might have
exercise-induced angina. I completed the Nuclear Stress Test, and I could not
believe his diagnosis of potential cardiovascular disease. I had been physically fit
my whole life and watched my diet because of losing my father at such an early age
and diligent about my annual medical screenings, so what had I done wrong? He
wanted me to take blood thinners overnight. He stated, “There are several abnor-
malities on your test, and I need you in hospital first thing tomorrow morning for a
heart catheterization.” I arrived at the hospital and was shocked when they asked me
if I had a Will. I was a young, single female who owned property and had never
dreamed I would need a Will at my age. I felt a sense of impending doom as I signed
the consent papers at check-in to immediately convert me to bypass surgery should
it be necessary if they could not stent the potential blockages in my heart arteries. I
knew there might be limitations reaching the blockages with the current portfolio of
stents because of my smaller female anatomy. I was facing this life crisis alone; my
family was overseas without time to reach me. Petrified, they wheeled me down to
the catheterization lab for the procedure.
Upon injection of dye into my coronary arteries, I heard a collective expression
of surprise in the Cath Lab as it revealed a 95% occluded proximal left anterior
descending coronary artery. This type of blockage is commonly referred to as the
“widow maker” in the cardiology world. I remember asking, “Can you fix it?” The
doctor said, “I think your father is sitting on your shoulder because I am not sure
how you are still here.” They inserted a drug-coated stent to open the blockage.
Interestingly, after my procedure, the Cath Lab nurses asked me, “Strong family
history?” As I think about my journey with heart disease, they were a few of the
people with empathy who instinctively, based on their experience, realized the
inherited nature of my premature heart disease. During my follow-up visit with my
cardiologist, I cried, and I asked him, “What did I do wrong?”
I felt this way because of the public misconception that a poor lifestyle is the
only reason people get cardiovascular disease.
The cardiologist said he wanted to understand the cause of my premature heart dis-
ease and ran more cardiometabolic bloodwork. That is the moment I discovered I
had high Lp(a). He told me I did not do anything wrong, and I inherited this from
my parents. I had an uncontrollable genetic risk factor for my premature heart dis-
ease, but surprisingly, it was a huge relief not to carry the shame that I could have
prevented this event somehow. Finally, I could give my disorder a name. I was able
to have a sense of control over an uncontrollable situation. He also assured me there
would be significant advances in heart disease and not worry about my future.
Some would argue that testing for high Lp(a) is pointless in that “why test for it
if you cannot treat it.” Others would argue the benefit that testing for high Lp(a)
uncovers a hidden genetic risk factor. I am one of the faces of high Lp(a) who had
414 S. R. Tremulis
an inaccurate prediction of my cardiovascular disease risk and feel that Lp(a) testing
provides patients and their families a better risk prediction for premature cardiovas-
cular disease and death. A personalized prescription for more aggressive primary or
secondary prevention, including optimizing all cardiometabolic risk factors, can be
initiated for an at-risk individual. Due to the thrombogenic nature of Lp(a), the doc-
tor thought the aspirin I took when I felt my symptoms may have saved my life on
the plane journey home by reducing the risk of a flow-limiting coronary thrombosis
and a heart attack. In the Women’s Health Study, carriers of the rare LPA gene vari-
ant (rs3798220) had a relative 56% risk reduction in ASCVD risk in carriers on
aspirin therapy versus noncarriers (Chasman et al. 2009). More research should be
conducted to improve the risk calculators and on aspirin use for primary and sec-
ondary prevention for patients with high Lp(a) (Mortensen et al. 2015; Zheng and
Roddick 2019). I would also support risk-based versus trial-based calculators
because many other factors decide enrollment criteria in randomized controlled
clinical trials.
Over the next 10 years after my stent procedure, I would go through the grieving
process for my former self as I recovered and returned to my new normal. I became
an advocate for women’s heart disease, but little information was shared about
inherited cardiovascular disease. During these years, I had a child, and just as there
is a 30-year deficit of data on women and heart disease, there was a total deficit of
data to manage a woman through the reproductive years of her life with diagnosed
heart disease and high Lp(a). Nevertheless, this was the beginning of my journey to
learn about high Lp(a) and become educated and empowered and protect my own
life. I lived with the trauma from my father’s death and from my event, but it
appeared nothing had advanced in the field of Lp(a) research in the 10 years since
my heart event, and I wanted to know why. I later learned that the lack of implemen-
tation of a US and global standardized Lp(a) assay had hampered the progression of
Lp(a) research, but now there are exciting new developments with a mass
spectrometry-based approach for Lp(a) measurement. I would support the rapid
adoption of a global standardized Lp(a) assay because time is measured in lives for
patients.
A pivotal moment occurred when I went to see a leading lipid researcher at a
major medical institution for a consultation. I wanted to know about the latest
research on Lp(a). The appointment with the consultant took 2 h and cost $600. He
concluded with a very clinical and dogmatic statement, “You have a malignant fam-
ily history; there is no treatment for what you have. It is prohibitively expensive to
research because each different ethnic group has a different normal level of Lp(a),
and the child you risked your life having has a 50% chance of inheriting it!” I was
motivated to make a difference and said to my husband, “I have nothing to lose
except my life and everything to gain; I want us to make a difference. I do not want
another family to suffer. I want to save lives by educating everyone about the health
consequences of high Lp(a) and empower them to take action to reduce their risk
and save lives.” So, in 2013, I founded the Lipoprotein(a) Foundation.
24 Elevated Lp(a): Why Should I Test For It, If I Cannot Treat It? A Patient’s Perspective 415
I reviewed the published research focusing on evidence-based data and found that
20% (one in five people) have inherited high Lp(a), the most prevalent genetic risk
factor for cardiovascular disease; more than one billion people worldwide are
unaware they have at least a 60% increased risk for cardiovascular disease or death
(Nordestgaard et al. 2010; Kampstrup et al. 2009; Emerging Risk Factors
Collaboration et al. 2009; Patel et al. 2021). This lack of awareness was an unac-
ceptable situation that could not continue. The first sign of the disease, for some
people, is a heart attack or stroke. More than one billion families worldwide are
unaware of their actual risk. I thought I was rare and an outlier, but, as I discovered,
high Lp(a) is not a rare disorder (Nordestgaard et al. 2010).
When I mention these statistics, most people are shocked. Repeatedly, I hear
heart disease is 80% preventable, but what about the 20% who have inherited
an uncontrollable risk factor such as high Lp(a), familial hypercholesterolemia,
hypertriglyceridemia, and homocysteinemia and their age or gender? During
my research, it became increasingly clear we needed a dedicated charity to
raise awareness and help educate families about their genetic risk from the
fatty, sticky, Lp(a) particle in their blood on which diet and exercise have little
to no impact. These families, including friends, neighbors, or loved ones, may
not die from this inherited Lp(a) risk if we fund more awareness, advocacy,
community support, and research programs. Very few people talk about inher-
ited cardiometabolic disease, and even fewer people are diagnosed with it. I
would support adding comprehensive cardiometabolic genetic testing to the
risk calculators.
Our promise of value to our members was as follows:
“Guided by evidenced-based data on Lp(a), we help educate and empower our members to
save lives.”
• Fifty percent of hospital admissions for coronary artery disease have a normal
LDL-C <100 mg/dL (Sachdeva et al. 2009).
• Lp(a) is currently the strongest, single genetic risk factor for coronary heart dis-
ease and aortic stenosis (Kronenberg and Utermann 2013).
• Increasing evidence reveals that high Lp(a) is a genetic, independent, and causal
risk factor for coronary artery heart disease, atherosclerosis, thrombosis, stroke,
and aortic stenosis (Nordestgaard et al. 2010).
• Approximately sixty-three million people in the United States are unaware of
their risk from high Lp(a), one in five Americans and more than a one billion
people globally (Nordestgaard et al. 2010).
• The Lp(a) blood test is not part of the regular lipid panel.
• Traditional lifestyle preventative measures including diet and exercise have little
or no impact on Lp(a) levels (Mackinnon et al. 1997).
• High Lp(a) levels occur in all ethnic groups, but it is more common among
African Americans, South Asians, and Hispanics (Paré et al. 2019).
The increasing clinical evidence high Lp(a) is a causal risk factor for cardiovascular
disease and calcific aortic stenosis.
• Epidemiological studies/meta-analyses (Emerging Risk Factors Collaboration
et al. 2009)
• Mendelian randomized studies (Kampstrup et al. 2009)
• Genetic association studies (Clarke et al. 2009)
• Insights from UK Biobank (Patel et al. 2021)
Randomized controlled clinical trials (RCT)—patients with high Lp(a) levels are
randomized to potential therapy. As of 2019, there are now at least three clinical
trials underway for a specific therapy to lower Lp(a) (Viney et al. 2016).
Kare Berg discovered Lp(a) in human serum in 1963. After 60 years, there still
is no FDA-approved therapy for lowering high Lp(a). With the launch of recent
Lp(a) clinical trials, there is hope on the horizon for patients with high Lp(a).
The Lipoprotein(a) Foundation was a patient-founded and patient-focused organi-
zation that helped reveal the impact of high Lp(a). It was supported by a team of
researchers, healthcare practitioners, and patient advocates who volunteered their
knowledge and passion for helping others. We were honored to have Lp(a) key opin-
ion leaders, both from the US and international arena, sharing their expertise and
research insights with the Lipoprotein(a) Foundation. Our success was rooted in pas-
sion, empathy, innovation, and commitment. The foundation took pride in its innova-
tive and grassroots approach to making real change for 20% of the global population
24 Elevated Lp(a): Why Should I Test For It, If I Cannot Treat It? A Patient’s Perspective 417
living with or at risk of cardiovascular disease or death due to high Lp(a). Based on
feedback from our member community, I identified a set of strategic program areas
for the foundation. Specific objectives were set within each program to help overcome
the barriers to adoption for Lp(a) testing, which included executing an integrated mar-
keting and communications plan within 5 years prioritizing grassroots efforts due to
minimal funding and resources. The Lipoprotein(a) Foundation and its community
made a measurable impact in these key strategic areas. Since 2013, the Lipoprotein(a)
Foundation has delivered impactful programs driving awareness, advocacy, commu-
nity support, and research to effect change and address unmet needs (Table 24.4).
Table 24.4 Overview of the foundation’s key accomplishments from 2013 to 2020 (The
Lipoprotein(a) Foundation 2020)
Then (2013) Now (2020)
No ICD codes for ICD-CM Codes E78.41 and Z83.430 approved—56% increase in
Lp(a) individuals and 71% increase in families diagnoseda
No Lp(a) contact 8000+ enrolled; helped enroll three phase 1 and one phase 2 clinical
registry trials. Published market research study on participating in clinical trials
during COVID-19 (Swerdlow et al. 2021)
No Lp(a) awareness 500+ million impressions from PR activities, 500+ online headline
postings, top Google ranking, featured in New York Times, USA Today,
Fox News, American Airlines, Martha Stewart Living, plus others.
Community outreach focused on high-priority gender, ethnic, and
disease state groups
No Lp(a) support Growing community online and in person—social media, patient forum
community program, community events, and support phone line
No Lp(a)-focused 140,000+ visitors from 166 countries to the website each year offering
website Lp(a) expert physician location services
No professional Lp(a) in ACC/AHA, NLA, ESC/EAS, and cholesterol guidelines as a
guidelines risk factor (Virani et al. 2022)
No group advocating Seven years of Lp(a) advocacy with NIH, CDC, and others
for Lp(a)
Little attention and NIH strategic research proposal (Tsimikas et al. 2018)—$400K grant
funding for Lp(a) awarded to Columbia University Medical Center
No gathering of Thirty top Lp(a) experts on SAB and CAB advisory board, including
experts representation from all our prioritized groups
No treatment options Five potential innovative treatments in development, three in clinical
trials; helped enroll three phase 1 (Akcea/Ionis, Amgen, Silence
Therapeutics) and one phase 2 clinical trial (Akcea/Ionis/Novartis)
No directory of Lp(a) 600+ physicians registered with the foundation
specialists
No standardized NHLBI/CDC working group conducted on global standardization of
Lp(a) blood test Lp(a) assay in humans (Lijuan et al. 2019)
The table was created from data from the 2019 Impact Report for the Lipoprotein(a) Foundation
L00012US 6/20. Copyright 2020 Lipoprotein(a) Foundation EIN: 46-3024812 a nonprofit, 501(c)3
patient advocacy organization
a
Data provided by Vladimir Polony from the Green Button team to the Lipoprotein(a) Foundation,
led by Nigam Shah at the Stanford Center for Biomedical Informatics Research from a representa-
tive sample database
418 S. R. Tremulis
Patients and their families need an accurate prediction of their risk for premature
cardiovascular disease to prevent the first symptom from being death. A 2016 study
by Mortensen et al. looked at statin eligibility and 5-year cardiovascular disease
outcomes in 37,892 individuals (57% women) aged 40–75 years of age in the
Copenhagen General Population Study (Mortensen et al. 2015). The study limita-
tions include that it only looked at Caucasian subjects and was limited to a 5-year
follow-up. It would have been more informative to include higher-risk Hispanic,
Black, and South Asian populations with high Lp(a). The study used the 2013
American College of Cardiology/American Heart Association (ACC/AHA) risk
prediction tool. In the results of their study, as noted by Dr. Vera Bittner (the
University of Alabama at Birmingham) in an accompanying editorial, “The study
suggests that Lp(a) levels might help identify the 8% of individuals who had an
event despite being ineligible for statins.” She noted, “Comprehensive risk factor
control is associated with improved prognosis, and our challenge is to develop care
models that will allow us to achieve such control.” Another perspective accompany-
ing the article notes, “Future research should be directed toward developing more
accurate risk prediction tools.” In the editorial accompanying this study, Dr. Valentin
Fuster, JACC editor-in-chief (Icahn School of Medicine at Mount Sinai, New York),
suggested, “Let’s begin to pay attention to high Lp(a) because it may explain cardio-
vascular events in patients who otherwise do not have a significant risk factor pro-
file.” (Bennet et al. 2088) There are 1.5 million people in the United States who have
a cardiovascular event every year; 8% of that number is 120,000 people (Erqou
et al. 2009).
The public appears to have little empathy for cardiovascular disease because it is
often perceived as self-inflicted. But would not it be good to reduce the emotional
and financial impact on US society of an estimated 120,000 people with only iso-
lated high Lp(a) having a cardiovascular event every year and many more globally?
Individuals can be diagnosed with a simple inexpensive Lp(a) blood test, but you
can also identify a family potentially at risk for generations to come. It is a simple,
blood test once in a person’s life and annual bloodwork is not required. We encour-
age healthcare practitioners to pursue continued education about this inherited lipid
risk. In medical practices, one in five individuals and their families already have
high Lp(a) and face at least a 60% increased risk of a cardiovascular event
(Nordestgaard et al. 2010; Kamstrup et al. 2009). Educating and empowering
patients about high Lp(a) does save lives. In 2019, the Lipoprotein(a) Foundation
was named a Top-Ranked Nonprofit by the leading platform for community-sourced
stories about nonprofits. The foundation received this award for successfully achiev-
ing the objectives of our 5-year strategic plan and because of community feedback
on our programs. A patient advocate, stated, “The Lipoprotein(a) Foundation has
helped save my life! It was the beginning of my journey, guiding me through what
I needed to test to identify what ended up being significant heart disease.” This tes-
timonial was just one of the many mission moments that occurred as we fulfilled our
objectives for the foundation.
24 Elevated Lp(a): Why Should I Test For It, If I Cannot Treat It? A Patient’s Perspective 419
Conclusion
Awareness
So, how can I have had normal LDL-cholesterol and almost died of a heart attack
despite having a healthy lifestyle, annual preventative health screening, and no sig-
nificant risk factors except my family history of cardiovascular disease? It is my
opinion that, unfortunately, there is low public awareness for personal inherited
cardiovascular disease risk (Sanderson et al. 2011). The data to generate that aware-
ness for high Lp(a) has been inconsistent and largely missing due to the lack of a
standardized Lp(a) assay for research purposes, drug target development, and level
1 data from clinical trials for a therapy to improve outcomes for patients. Level 1
data is the trigger to include a risk target into the global cholesterol guidelines if
they are trial-based versus risk-based, and those guidelines are periodically updated
(Marcovina et al. 2003). Often, the public perception is that cardiovascular disease
is entirely self-inflicted, and the stigma attached to it is similar to AIDS and lung
cancer, which reduces the funding and empathy that often drives awareness of a
disease state (Benson 2021). This is also the case for women’s heart disease due to
a 30-year deficit of women and heart disease data (Garcia et al. 2016). I was aware
of my family history of heart disease but not my risk as a young woman with a
strong family history. The global focus and funding imperative for COVID-19 vac-
cines show how an enormous-focused response might finally eliminate the insur-
mountable global burden of cardiovascular disease. All stakeholders involved in the
cardiometabolic disease industry should focus on driving awareness for personal-
ized, inherited cardiometabolic disease.
In addition, we must do better for women in healthcare and recognize the unique
and important differences between men and women; women’s more subtle symp-
toms may be ignored or treated less aggressively than male patients (Garcia et al.
2016). I was one of those women with subtle symptoms treated less aggressively.
Many biases can impair diagnostic accuracy by humans. Availability bias, a cogni-
tive bias, can lead to diagnosis errors (Yagoda 2018).
Without having the latest medical research data available on-demand to healthcare
providers at the point of care within their institution, a physician cannot be informed
about the latest evidence-based data to aid in care (Lenaerts et al. 2021). With the
420 S. R. Tremulis
advent of artificial intelligence (AI), there is promise for AI tools such as the Human
Diagnosis Project, also known as Human Dx, aiding in diagnosis if the human
biases do not become embedded in the AI tools (Human Diagnosis Project 2022).
It would benefit patients and their families to establish focused clinical centers of
excellence providing equal access to specialized treatment and care for inherited
cardiometabolic disease with a priority given to underserved minority groups at
increased risk from high Lp(a). Developing a standard of care to direct families with
inherited cardiometabolic disorders to these clinical centers of excellence would
simplify access to state-of-the-art research and care (Elrod and Fortenberry Jr. 2017).
We have known about high Lp(a) for 60 years and still do not have a globally stan-
dardized Lp(a) assay, which has hampered research and the progression of the body
of scientific evidence in this area (Marcovina et al. 2003). The bloodwork identify-
ing my only inherited, hidden, significant contributing risk factor, high Lp(a), was
performed after my life-threatening event. Including a globally standardized test for
high Lp(a) in the standard preventative lipid screening and risk calculators would
provide a more accurate prediction of risk for patients and their families, initiating
more aggressive primary and secondary prevention, which otherwise may not have
been identified (Mortensen et al. 2015).
24 Elevated Lp(a): Why Should I Test For It, If I Cannot Treat It? A Patient’s Perspective 421
There is no path of vigilance for inherited high Lp(a) as there is for other diseases
from birth onward. The risk calculators that guide the standard of care for cardiovas-
cular disease are designed for population medical care and not personalized medi-
cine. They do not include inherited cardiometabolic risk markers such as high Lp(a)
or factor in premature cardiovascular disease at an age younger than 45 (Semaev
and Shakhtshneider 2020). I was one of those young people with high Lp(a) missed
by the risk calculators.
Precision Medicine
Personalized preventative medical care is costly to adopt for the general population.
Researching the benefit of aspirin use for high Lp(a), including aspirin resistance
and other forms of blood clotting disorders, could provide a cost-effective preventa-
tive solution for patients with high Lp(a) (Greving et al. 2008). It will not prevent
cardiovascular disease due to high Lp(a) but might save lives. By chance, I took
aspirin during my heart event due to public awareness information about blood clots
when flying.
burden of premature cardiovascular disease in the United Sates and help prioritize
funding (Bilimoria et al. 2008). The healthcare industry could invest savings gener-
ated by preventing cardiovascular events and heart damage for 20% of the global
population with high Lp(a) into the cost of precision, preventative cardiometabolic
care. Improving the survival rate of a heart event is good but often moves the costs
along to managing the chronic condition of heart failure after the heart is damaged
(Heidenreich et al. 2013).
References
Bennet A, Di A, Erqou S, et al. Lp(a) levels and risk of future coronary heart disease: large-scale
prospective data. Arch Intern Med. 2088;168:598–608.
Benson L. This survivor wants to change the narrative around lung cancer [Internet]. Next Avenue.
2021. https://www.nextavenue.org/lung-cancer-stigma/. Accessed 22 Feb 2022.
Bilimoria KY, Stewart AK, Winchester DP, Ko CY. The National Cancer Data Base: a powerful
initiative to improve cancer care in the United States. Ann Surg Oncol. 2008;15(3):683–90.
Bittner V. Lipoprotein abnormalities related to women’s health. Am J Cardiol. 2002;90(8A):
77i–84i.
Bittner V. Selecting patients for statin therapy in primary prevention if we could predict the future.
J Am Coll Cardiol. 2015;66:2710–1.
Boffa MB, Koschinsky ML. Lipoprotein(a): truly a direct prothrombotic factor in cardiovascular
disease? J Lipid Res. 2016;57(5):745–57.
24 Elevated Lp(a): Why Should I Test For It, If I Cannot Treat It? A Patient’s Perspective 423
Patel AP, Wang M, Pirruccello JP, Ellinor PT, Ng K, Kathiresan S, Khera AV. Lp(a) (Lipoprotein[a])
concentrations and incident atherosclerotic cardiovascular disease: new insights from a large
national biobank. Arterioscler Thromb Vasc Biol. 2021;41(1):465–74.
Rogers MA, Aikawa E. A not-so-little role for lipoprotein(a) in the development of calcific aortic
valve disease. Circulation. 2015;132:621–3.
Sachdeva A, Cannon CP, Deedwania PC, et al. Lipid levels in patients hospitalized with coronary
artery disease: an analysis of 136,905 hospitalizations in Get With the Guidelines. Am Heart
J. 2009;157:111–7.
Sanderson SC, · Waller J · Humphries SE · Wardle J Public awareness of genetic influence on
chronic disease risk: are genetic and lifestyle causal beliefs compatible? Public Health
Genomics 2011;14:290–297.
Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of
lipoprotein(a). J Lipid Res. 2016;57:1339–59.
Semaev S, Shakhtshneider E. Genetic risk score for coronary heart disease: review. J Pers Med.
2020;10(4):239. https://doi.org/10.3390/jpm10040239.
Swerdlow D, Tremulis S, et al. Patients with raised lipoprotein(a) are committed to being involved
in clinical trials to develop a treatment to lower Lp(a): results of a survey distributed via the
lipoprotein(a) foundation. J Clin Lipidol. 2021;15(5):e17.
Thanassoulis G, et al. Genetic associations with valvular calcification and aortic stenosis. N Engl
J Med. 2013;368(6):503–12.
The Lipoprotein(a) Foundation. Lipoprotein(a) Foundation Impact Report—how you have made a
difference in the lives of families living with or at risk from high Lp(a) [Internet]. Yumpu. 2020.
https://www.yumpu.com/en/document/read/63585647/the-lipoproteina-foundation-2019-
impact-report. Accessed 2 Feb 2022.
Tsimikas S, Fazio S, Ferdinand KC, Ginsberg HN, Koschinsky ML, Marcovina SM, Moriarty PM,
Rader DJ, Remaley AT, Reyes-Soffer G, Santos RD, Thanassoulis G, Witztum JL, Danthi S,
Olive M, Liu L. NHLBI working group recommendations to reduce lipoprotein(a)-mediated
risk of cardiovascular disease and aortic stenosis. J Am Coll Cardiol. 2018;71(2):177–92.
Viney NJ, van Capelleveen JC, Geary RS, Xia S, Tami JA, Yu RZ, Marcovina SM, Hughes SG,
Graham MJ, Crooke RM, Crooke ST, Witztum JL, Stroes ES, Tsimikas S. Antisense oligo-
nucleotides targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised,
double-blind, placebo-controlled, dose-ranging trials. Lancet. 2016;388(10057):2239–53.
Virani SS, Alonso A, Aparicio HJ, Benjamin EJ, Bittencourt MS, Callaway CW, et al. Heart
Disease and Stroke Statistics—2021 update: a report from the American Heart Association.
Circulation. 2021;143:e254–743.
Virani S, Koschinsky M, Maher L, Mehta A, Orringer C, Santos R, Shapiro M, Saseen J. Global
think tank on the clinical considerations and management of lipoprotein(a): the top questions
and answers regarding what clinicians need to know. Prog Cardiovasc Dis. 2022;73:32–40.
https://doi.org/10.1016/j.pcad.2022.01.002.
Willeit P, Kiechl S, Kronenberg F, Witztum JL, 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(9):851–60.
Wilson DP, Jacobson TA, Jones PH, Koschinsky ML, McNeal CJ, Nordestgaard BG, Orringer
CE. Use of lipoprotein(a) in clinical practice: a biomarker whose time has come. A scientific
statement from the National Lipid Association. J Clin Lipidol. 2019;13(3):374–92.
World Health Organization. Cardiovascular diseases (CVDs) [internet]. World Health Organization
2021. 2021. https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds).
Accessed 11 Jun 2021.
Yagoda B. The cognitive biases tricking your brain. Atlantica. 2018;
Zheng SL, Roddick AJ. Association of aspirin use for primary prevention with cardiovascular
events and bleeding events: a systematic review and meta-analysis. JAMA. 2019;321:277.
Chapter 25
Unresolved Questions
It is now almost exactly 60 years since Kare Berg first described an extra pre-β band
found in lipid electrophoresis that later was named sinking pre-β and finally Lp(a).
There was a continuous up and down in Lp(a) research that was mainly driven by
actual research findings related to Lp(a) function, metabolism, correlation to cardiac
risk, and epidemiology. The following are four key findings that caused a major
boost in the interest for Lp(a) that led to a flurry in publications:
1. Cloning of LPA by McLean and Lawn demonstrating homology of the apo(a)
protein and the LPA gene with plasminogen (McLean et al. 1987; Utermann 2001).
2. The unique size polymorphism caused by variations in the number of K-IV2
repeats that paved the way for consecutive genetic epidemiological studies by
the group of Utermann [reviewed in (Utermann 2001; Kamstrup et al. 2009)].
3. The demonstration of the causal relationship of elevated Lp(a) levels with ath-
erosclerosis and coronary heart diseases by Mendelian randomization in the
Copenhagen Heart Study (Kamstrup et al. 2009; Graham et al. 2016).
4. The development of a very efficient therapy for elevated-Lp(a) with antisense
oligonucleotide (ASO) therapy by the group of Tsimikas (Graham et al. 2016).
All these exciting milestones in Lp(a) research cannot change the fact that our
knowledge in all areas of Lp(a) research is still fragmented. This is due to a lack of
knowledge in the following areas:
1. Function
2. Metabolism
G. M. Kostner (*)
Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
e-mail: [email protected]
K. Kostner
Department of Cardiology, Mater Hospital and University of Queensland, Brisbane, Australia
3. Pathophysiology
4. Lp(a) measurement in clinical laboratories
5. Significance in diseases other than related to atherosclerosis
6. Therapy
Many of these points have been already discussed in the previous chapters and
will therefore only be summarized here:
Nature rarely designs complex structures such as apo(a) without any physiological
function in mind. This may not necessarily be true for the whole human population
but may be only for some ethnicity. Thus, it has been speculated that individuals
exposed to dangerous parasites or bacterial and viral infections may have an advan-
tage if they have high Lp(a) plasma concentrations. The actual mechanism behind
this is far from being clear, yet it may explain why populations originating from
African countries where such diseases prevail have significantly higher Lp(a) levels
than Europeans and Asians (Schmidt et al. 2006; Sandholzer et al. 1992).
Another function of Lp(a) might relate to aging and longevity. In fact, lipids and
lipoproteins have been implicated in life span regulation (Joshi et al. 2017), and in
the list of genes suggested in previous research to code for such factors, LPA, APOE,
and APOAI are found. We addressed this question in early investigations in view of
age-related diseases and hypothesized that assuming that Lp(a) might be a signifi-
cant risk factor for atherosclerosis and MI, individuals with high Lp(a) should die
earlier than individuals with low Lp(a). Thus, we first measured Lp(a) in a family
kindred within three generations and anticipated that Lp(a) values in the older gen-
eration might be lower than in the younger one (Pagnan et al. 1982). In fact, the
opposite turned out to be the case. In another study, we measured Lp(a) in octo-
nonagenarians (Zuliani et al. 1995), and although we could not confirm a correla-
tion of Lp(a) with age, to our surprise, the plasma Lp(a) concentration of “very old”
individuals and more importantly the apo(a) isoform distribution did not differ sig-
nificantly from that of young individuals (Zuliani et al. 1995). Comparable studies
have been also published from other investigators (Wood and Schumacher 1995).
The question obviously arises about the physiological relevance of these obser-
vations—or in other words—does this relate to a physiological function of Lp(a).
We studied this possibility by asking whether apo(a) might be involved in angiogen-
esis (Schulter et al. 2001). Angiogenesis has been found to be important not only for
tumor growth but also for cancer metastasis. O’Reilly et al. (1994) were first to
demonstrate that angiostatin, a proteolytic cleavage product of plasminogen secreted
into urine, has very high angiostatic properties. Since proteolytic fragments from
apo(a) are found in urine as well, we purified these fragments and tested their angio-
static properties in vitro in a tube forming assay: indeed apo(a) from urine that
25 Unresolved Questions 427
Metabolism
Biosynthesis and assembly: The chapter authored by Dan Rader and John Miller in
this book gives an excellent overview on the current concepts of Lp(a) metabolism.
As these authors point out, our research group was first to demonstrate that the
Lp(a) metabolism is distinct from that of LDL: other than for LDL, VLDL is not a
precursor of Lp(a) (Krempler et al. 1979). We also published that other than for
LDL, plasma Lp(a) concentrations are governed by the rate of biosynthesis—or in
other words, individuals with high Lp(a) concentrations show a high rate of apo(a)
expression (Krempler et al. 1980). The expression of apo(a) is driven by
428 G. M. Kostner and K. Kostner
transcription factors and nuclear receptors. In silico search in the APOA promoter
revealed more than 70 binding regions for known transcription factors; two of such
response elements, ETS −1630 to −1615 and DR-1 −826 to −814, turned out to be
of particular importance as they are strongly turned off by FXR signaling
(Chennamsetty et al. 2011). We also identified several cAMP response elements that
were responsible for the Lp(a) lowering effect of nicotinic acid (Chennamsetty et al.
2012). But how about the role of all the other response elements that we identified
in the apo(a) promoter? This is an ample research field that deserves much further
attention.
Following APOA transcription, translation, and glycosylation, the mature apo(a)
protein assembles with LDL to form Lp(a). The individual steps in assembly have
been addressed in numerous studies in the past, yet there is currently no general
agreement on the site where this might occur. Whereas some data favor an intrahe-
patic assembly, other data point toward an assembly on the surface of liver cells, and
even others suggest an assembly in circulating blood. Undoubtedly there are further
studies needed to clarify the location of the assembly of Lp(a).
Another fully open field is the role of the APOA expression in the brain and testes
(McLean et al. 1987): does this have any physiological relevance? Nobody has ever
studied this rather interesting phenomenon in detail.
Catabolism: In our early experiments in man, we found that FH patients lacking
LDL receptors catabolize Lp(a) to the same extent than healthy controls. The cata-
bolic rate in both, however, was markedly slower as compared to LDL (Krempler
et al. 1980). This led us to conclude that LDL receptor-mediated catabolism plays
little role in Lp(a) removal from circulation. Since then, a wealth of publications
appeared that found Lp(a) binding to almost any specific lipoprotein receptor
including the apoE receptor, the VLDL receptor, the remnant receptor, LRP recep-
tor, the asialoglycoprotein receptor, the plasminogen receptor, several scavenger
receptors, and possibly others. Fact is that in all animal studies even in hedge hogs,
approximately 50% of intravenously injected Lp(a) is taken up by the liver (Kostner
et al. 1997). In addition to the liver, also the kidney plays an important role in Lp(a)
metabolism (see chapter of H. Dieplinger in this book). Apo(a) is fragmented in the
blood by Ca2+-dependent proteases, and even large fragments are secreted into urine
(Frank et al. 2001). The significance of this pathway has never been clarified so far.
We know more about the pathophysiology of Lp(a) than about its physiology. Lp(a)
consists of an LDL particle with all its proatherogenic properties. In addition, Lp(a)
gets into atherosclerotic plaques by interacting with proteoglycans, which causes
foam cell formation and inflammation; Lp(a) also carries OxPhos that trigger
inflammation. Due to the homology of apo(a) with plasminogen, Lp(a) has also
25 Unresolved Questions 429
The early laboratory methods for measuring Lp(a) were based on immunochem-
istry. Almost all immunochemical methods including radial immune-diffusion
(Ouchterlony test), rocket electrophoresis, radioimmunoassay (RIA), ELISA
(enzyme-linked immunosorbent assay), DELFIA (dissociation-enhanced lantha-
nide fluorescence immunoassay), and more methods have been applied. Today,
high-throughput methods for Lp(a) quantitation are based on immune-turbidime-
try or immune-nephelometry. Unfortunately, commercial methods are far from
being harmonized and subject to drastic improvements. There is currently no vali-
dated reference material commercially available, and reference methods for typ-
ing Lp(a) standards are still under development. All the problems with Lp(a)
quantitation in the clinical laboratory are impressively documented in the chap-
ters from S. Marcovina and C. Cobbaert and D. Sullivan in this book. Another
article that highlights this thematic was recently published by F. Kronenberg
(Kronenberg 2022). Kronenberg looks at this topic from practical point of view
and stresses the point that due to the great genetic heterogeneity of LPA, it will be
hardly possible to have a validated routine method for high-throughput Lp(a)
measurement at reasonable costs available that fulfills all standard requirements
of ISO 17511:2020. Former studies where commercial assays for Lp(a) were
evaluated revealed a significant number of outliers that at present time cannot be
explained (Scharnagl et al. 2019). C. Cobbaert established an IFCC (International
Federation of Clinical Chemistry)-sponsored working group with experts in the
field of mass spectrometry and laboratory medicine with the goal to develop a
reference method based on LC-MS (liquid chromatography-mass spectrometry)
http://www.ifcc.org/ifcc-scientific-division/sd-working-groups/wg-apo-ms/. In
addition, this group works on the preparation of a harmonized SI-traceable refer-
ence material that shall be used by industry to standardize their commercial
assays. Although the IFCC working group has been operational for more than 5
years, it may take another 2–3 years to come up with a practicable reference
method and a reference material.
430 G. M. Kostner and K. Kostner
Apart from LDL apheresis therapy, it is currently not clear whether lowering of
Lp(a) reduces hard cardiovascular endpoints. Several phase 2 and 3 trials with anti-
sense and siRNA-targeted therapies are exploring this currently. Most lipidologists
25 Unresolved Questions 431
antisense and RNA technologies are more specific and show much larger Lp(a)
reductions. Clinical outcome trials are currently underway (HORIZON,
NCT04023552; OCEAN(a)-DOSE; NCT04270760).
Monoclonal PCSK9 antibodies, as well as RNA-based inhibitors of PCSK9,
which lower LDL-C, can also reduce Lp(a) by up to 35%. While they are not reim-
bursed through Medicare for Lp(a) treatment, elevated Lp(a) is often treated coinci-
dentally in patients with FH or progressive ASCVD whose LDL-C remains elevated
despite maximal statin and ezetimibe therapy. Sub-analysis of the major outcomes
trials for PCSK9 inhibitors has shown greater relative and absolute risk reduction in
patients with elevated Lp(a). In Cardiovascular Outcomes Research With PCSK9
Inhibition in Subjects With Elevated Risk (FOURIER) trial, reduction in risk of
major acute coronary events (MACE) with evolocumab was associated with base-
line and change in Lp(a) levels (O’Donoghue et al. 2019). In the Evaluation of
Cardiovascular Outcomes After an Acute Coronary Syndrome During Treatment
With Alirocumab (ODYSSEY OUTCOMES) trial, reduction in risk of total cardio-
vascular events with alirocumab was also associated with baseline and change in
Lp(a) levels (Szarek et al. 2020). Reduction in risk of major adverse limb events
(MALE) with alirocumab was also associated with baseline and change in Lp(a)
levels (Schwartz et al. 2020). These trials support the conclusion that elevated Lp(a)
is a major driver of residual risk.
Statins do have a variable effect on plasma Lp(a). Although most statins are able to
lower Lp(a) to some extent, there are numerous patients who do not respond to
statins at all or even show an increase of Lp(a) on statin therapy (Kostner et al.
1989). The mechanisms responsible have never been defined. The important fact to
remember is that statins are beneficial in patients with elevated Lp(a) by removing
LDL, which reduces some of the CV risks associated with elevated Lp(a).
References
Bittner VA, Szarek M, Aylward PE, Bhatt DL, Diaz R, Edelberg JM, Fras Z, Goodman SG,
Halvorsen S, Hanotin C, Harrington RA, Jukema JW, Loizeau V, Moriarty PM, Moryusef A,
Pordy R, Roe MT, Sinnaeve P, Tsimikas S, Vogel R, White HD, Zahger D, Zeiher AM, Steg
PG, Schwartz GG. Committees OO and investigators. Effect of alirocumab on lipoprotein(a)
and cardiovascular risk after acute coronary syndrome. J Am Coll Cardiol. 2020;75:133–44.
Blencowe C, Hermetter A, Kostner GM, Deigner HP. Enhanced association of platelet-activating
factor acetylhydrolase with lipoprotein(a) in comparison with low density lipoprotein. J Biol
Chem. 1995;270:31151–7.
Boffa MB. Beyond fibrinolysis: the confounding role of Lp(a) in thrombosis. Atherosclerosis.
2022;349:72–81.
Chennamsetty I, Claudel T, Kostner KM, Baghdasaryan A, Kratky D, Levak-Frank S, Frank S,
Gonzalez FJ, Trauner M, Kostner GM. Farnesoid X receptor represses hepatic human APOA
gene expression. J Clin Invest. 2011;121:3724–34.
Chennamsetty I, Kostner KM, Claudel T, Vinod M, Frank S, Weiss TS, Trauner M, Kostner
GM. Nicotinic acid inhibits hepatic APOA gene expression: studies in humans and in trans-
genic mice. J Lipid Res. 2012;53:2505–412.
Frank S, Hrzenjak A, Blaschitz A, Dohr G, Kostner GM. Role of various tissues in apo(a) fragmen-
tation and excretion of fragments by the kidney. Eur J Cin Invest. 2001;31:504–12.
Gencer BG, Giugliano RP, Tang M, Mach F, Stroes ES, Gouni-Berthold I, Ceska R, Ezhov M,
Jukema JWW, Jensen H, Tokgozoglu LS, Huber K, Sabatine MS, O’Donoghue ML. Clinical
safety outcomes in relation to lipoprotein(a) concentrations: insights from the FOURIER trial.
Circulation. 2021;144(Suppl_1):A11043.
Gorges R, Hofer G, Sommer A, Stutz H, Grillhofer H, Kostner GM, Paltauf F, Hermetter
A. Transfer of phospholipase A-resistant pyrene-dialkyl-glycerophosphocholine to plasma
lipoproteins: differences between Lp[a] and LDL. J Lipid Res. 1995;36:251–9.
Graham MJ, Viney N, Crooke RM, Tsimikas S. Antisense inhibition of apolipoprotein(a) to lower
plasma lipoprotein(a) levels in humans. J Lipid Res. 2016;57:340–51.
Hermann PM, Watson SN, Wildering WC. Phospholipase A2—nexus of aging, oxidative stress,
neuronal excitability, and functional decline of the aging nervous system? Insights from a
snail model system of neuronal aging and age-associated memory impairment. Front Genet.
2014;5:419. https://doi.org/10.3389/fgene.2014.00419.
434 G. M. Kostner and K. Kostner
Joshi PK, Pirastu N, Kentistou KA, Fischer K, Hofer E, Schraut KE, Wilson JF. Genome-wide
meta-analysis associates HLA-DQA1/DRB1 and LPA and lifestyle factors with human longev-
ity. Nat Commun. 2017;8(1):910. https://doi.org/10.1038/s41467-017-00934-5.
Kamstrup PR, Tybjaerg-Hansen A, Steffensen R, Nordestgaard BG. Genetically elevated
lipoprotein(a) and increased risk of myocardial infarction. JAMA. 2009;301:2331–9.
Koren MJ, Moriarty PM, Neutel J, Baum SJ, Hernandez-Illas M, Weintraub HS, 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(Suppl_3):A13951.
Kostner GM, Gavish D, Leopold B, Bolzano K, Weintraub MS, Breslow JL. HMG-CoA
reductase inhibitors lower LDL cholesterol without reducing Lp(a) levels. Circulation.
1989;1989(80):1313–0.
Kostner GM, Wo X, Frank S, Zimmermann R, Steyrer E. Metabolism of Lp(a): assembly and
excretion. Clin Genet. 1997;52:347–54.
Kostner KM, Kostner GM, Wierzbicki AS. Is Lp(a) ready for prime time use in the clinic? A pros-
and-cons debate. Atherosclerosis. 2018;274:16–22.
Krempler F, Kostner G, Bolzano K, Sandhofer F. Lipoprotein(a) is not a metabolic product of other
lipoproteins containing apolipoprotein B. Biochim Biophys Acta. 1979;575:63–70.
Krempler F, Kostner GM, Bolzano K, Sandhofer F. Turnover of lipoprotein(a) in man. J Clin
Invest. 1980;65:1483–90.
Kronenberg F. Lipoprotein(a) measurement issues: are we making a mountain out of a molehill?
Atherosclerosis. 2022;349:123–35.
McLean JW, Tomlinson JE, Kuang WJ, Eaton DL, Chen EY, Fless GM, et al. cDNA sequence of
human apolipoprotein(a) is homologous to plasminogen. Nature. 1987;330(6144):132–7.
Moriarty PM, Gray JV, Gorby LK. Lipoprotein apheresis for lipoprotein(a) and cardiovascular
disease. J Clin Lipidol. 2019;13(6):894–900.
O’Donoghue M, Fazio S, Stroes E, et al. Lipoprotein(a), PCSK9 inhibition and cardiovascular risk.
Insights from the Fourier trial. Circulation. 2019;39:1483–92.
O'Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Moses M, Lane WS, Cao Y, Sage
EH, Folkman J. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of
metastases by a Lewis lung carcinoma. Cell. 1994;79(2):315–28.
Pagnan A, Kostner G, Braggion M, Ziron L. Relationship between ‘sinking pre-beta-lipoprotein’
(Lp(a) lipoprotein) and age in a family kindred. Gerontology. 1982;28(6):381–5.
Sandholzer C, Boerwinkle E, Saha N, Tong MC, Utermann G. Apolipoprotein(a) phenotypes,
Lp(a) concentration and plasma lipid levels in relation to coronary heart disease in a Chinese
population: evidence for the role of the apo(a) gene in coronary heart disease. J Clin Invest.
1992;89:1040–6.
Sattler W, Kostner GM, Waeg G, Esterbauer H. Oxidation of lipoprotein Lp(a). A comparison with
low-density lipoproteins. Biochim Biophys Acta. 1991;1081:65–74.
Scharnagl H, Stojakovic T, Dieplinger B, Dieplinger H, Erhart G, Kostner GM, Herrmann M, et al.
Comparison of lipoprotein(a) serum concentrations measured by six commercially available
immunoassays. Atherosclerosis. 2019;289:206–13.
Schmidt K, Kraft HG, Parson W, Utermann G. Genetics of the Lp(a)/apo(a) system in an autoch-
thonous Black African population from the Gabon. Eur J Hum Genet. 2006;14:190–201.
Schulter V, Koolwijk P, Peters E, Frank S, Hrzenjak A, Graier WF, van Hinsbergh VW, Kostner
GM. Impact of apolipoprotein(a) on in vitro angiogenesis arteriosclerosis. Thromb Vasc Biol.
2001;21(3):433–8.
Schwartz GG, Steg GG, Szarek M, et al. Peripheral artery disease and venous thromboem-
bolic events after acute coronary syndrome. Role of lipoprotein(a) and modification by ali-
rocumab. Prespecified analysis of the Odyssey outcomes randomized clinical trial. Circulation.
2020;141:1608–17.
Sukocheva O, Menschikowski M, Hagelgans A, Yarla NS, Siegert G, Reddanna P, Bishayee
A. Current insights into functions of phospholipase A2 receptor in normal and cancer cells:
more questions than answers. Semin Cancer Biol. 2019;56:116–27.
25 Unresolved Questions 435
Szarek M, Bittner V, Aylward P, et al. Lipoprotein(a) lowering by alirocumab reduces the total
burden of cardiovascular events independent of LDL lowering: ODYSSEY OUTCOMES trial.
Eur Heart J. 2020;75:133–44.
Utermann G. Lipoprotein(a). In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The metabolic
and molecular bases of inherited disease. 8th ed. New York: McGraw-Hill; 2001. p. 2753–87.
Viney NJ, van Capelleveen JC, Geary RS, Xia S, Tami JA, Yu RZ, et al. Antisense oligonucleotides
targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind,
placebo-controlled, dose-ranging trials. Lancet. 2016;388(10057):2239–53.
Wood WG, Schumacher M. Lipoprotein(a) concentrations in non-selected hospitalized patient
between 18 and 100 years of age: comparison with cholesterol fractions and triacylglycerols in
patients with lipid status requests. Eur J Clin Chem Clin Biochem. 1995;33:903–14.
Yeang C, Witztum JL, Tsimikas S. Novel method for quantification of lipoprotein(a)-cholesterol:
implications for improving accuracy of LDL-C measurements. J Lipid Res. 2021;62:100053–62.
Zuliani G, Bader G, Imbastaro T, Mezzetti A, Taddeo U, Kostner GM, Fellin R. Lipoprotein(a)
plasma levels and apo(a) isoforms are not associated with longevity or disability in a sample of
Italian octo-nonagenarians. Associazione Medica Sabin. Aging. 1995;7(5):385–91.
Index
© The Editor(s) (if applicable) and The Author(s), under exclusive license to 437
Springer Nature Switzerland AG 2023
K. Kostner et al. (eds.), Lipoprotein(a), Contemporary Cardiology,
https://doi.org/10.1007/978-3-031-24575-6
438 Index
C
CAD, see Coronary artery disease (CAD) D
Calcific aortic stenosis, 409 Danger associated molecular patterns
Calcific aortic valve disease (CAVD), 173, (DAMPs), 263
275, 290 Deep vein thrombosis, 145
Calcific aortic valve stenosis (CAVS), 148, DELFIA, 429
149, 162 Denka Seiken based assays, 307
Candidate reference measurement procedure The Denka Seiken reagents, 306
(cRMP), 338, 339 Direct assays, 235
Cardiovascular disease (CVD), 135, 138, 148, Dresden Center for Extracorporeal Therapy,
150, 162–166, 220, 221, 328 386, 395
Cardiovascular mortality, 164
Cardiovascular outcomes trials, 354
Cardiovascular risk management, 349 E
Catabolism, 428 Electrospray differential ion mobility
CD14++CD16+ intermediate monocyte analysis, 310
subpopulation, 266 Endogeneous fibrinolytic system, 190, 195
CD16+ monocytes, 264 Enzyme-linked immunosorbent assay
CDC-Clinical Standardization Program (ELISA), 304, 328, 429, 430
(CSP), 342 E-selectin, 162
Cell surface receptors European ancestry, 235
kidney receptors, 130 European Society of Cardiology (ESC)/
liver receptors, 127–130 European Atherosclerosis Society
Lp(a) (EAS) guidelines, 278, 347
assembly, 127 European Union, 362
catabolism, 127 Evinacumab, 400
structure, 125–127 Evolocumab, 351
macrophages receptors, 131 Extracorporeal therapy, 269, 381
Center for Disease Control (CDC), 303 Ezetimibe, 350
Cerebral venous thrombosis, 191, 193
CETP inhibitors, 352
Cholesterol Reference Method Laboratory F
Network, 298 Familial hypercholesterolaemia (FH), 235,
Cholesteryl ester transfer protein 276, 289, 432
(CETP), 85, 352 Fibrinolysis, 141, 142, 145, 147, 152,
Clot lysis, 195 179–181
Coagulation, 195–196
Index 439