You are on page 1of 9

Clinica Chimica Acta 487 (2018) 117–125

Contents lists available at ScienceDirect

Clinica Chimica Acta


journal homepage: www.elsevier.com/locate/cca

Review

Dyslipidemias in clinical practice T


a a a,b,⁎
Manuel Castro Cabezas , Benjamin Burggraaf , Boudewijn Klop
a
Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045 PM Rotterdam, the Netherlands
b
Department of Cardiology, Erasmus MC, ‘s Gravendijkwal 230, 3015 CE Rotterdam, the Netherlands

ARTICLE INFO ABSTRACT

Keywords: Most dyslipidemic conditions have been linked to an increased risk of cardiovascular disease. Over the past few
Lipoprotein years major advances have been made regarding the genetic and metabolic basis of dyslipidemias. Detailed
lipid disorder characterization of the genetic basis of familial lipid disorders and knowledge concerning the effects of en-
hyperlipidemia vironmental factors on the expression of dyslipidemias have increased substantially, contributing to a better
hypercholesterolemia
diagnosis in individual patients. In addition to these developments, therapeutic options to lower cholesterol
treatment
levels in clinical practice have expanded even further in patients with familial hypercholesterolemia and in
subjects with cardiovascular disease. Finally, promising upcoming therapeutic lipid lowering strategies will be
reviewed. All these advances will be discussed in relation to current clinical practice with special focus on
common lipid disorders including familial dyslipidemias.

1. Introduction elevated LDL-C and plasma TG. Clinicians should be aware of the fact
that TG-rich lipoproteins (very low-density lipoproteins (VLDL) and
The term dyslipidemia refers to a disturbance of the lipid profile, chylomicrons and their remnants) also carry cholesterol molecules. This
including both hyperlipidemia and hypolipidemia. The latter may be explains why severely elevated TG are always accompanied by elevated
associated in some cases to deficiencies of fat-soluble vitamins, like the plasma cholesterol levels. One should exclusively use the term “hy-
homozygous form of abetalipoproteinemia and hypobetalipoprotei- pertriglyceridemia” when LDL-C levels are normal or low and the ele-
nemia [1], which are characterized by the absence of apolipoprotein vated plasma cholesterol is confined to the TG-rich lipoproteins. In this
(apo) B in plasma, or disorders like chylomicron retention disease in case plasma apo B is always within normal limits and this helps to
which there is an impaired secretion of intestinally-derived lipoproteins discriminate between hypertriglyceridemia (with the elevated risk of
[2]. Recently, a novel type resulting in reduced levels of low-density pancreatitis) and combined or mixed hyperlipidemia (with increased
lipoprotein cholesterol (LDL-C), triglycerides (TG) and high-density li- cardiovascular risk).
poprotein cholesterol (HDLeC) has been described, the so called “fa- Finally, a distinct type of dyslipidemia is mainly observed in type 2
milial combined hypolipidemia” [3]. In the heterozygous forms, these diabetes mellitus (DM) or situations associated to the metabolic syn-
types of dyslipidemias do not have a clinical expression and are drome and insulin resistance: the high TG-low HDL-C dyslipidemia, also
therefore, infrequently diagnosed. Others like hypoalfalipoproteinemia called “atherogenic dyslipidemia”.
(low HDLeC) may in some instances be linked to increased cardio- While the recognition of a dyslipidemic condition is not so difficult,
vascular risk [4]. clinicians should always try to establish the diagnosis underlying the
In clinical practice, the most frequent and therefore the most re- phenotypic expression. This is of extreme importance to determine the
levant dyslipidemias are the hyperlipidemias [5]. Most of these dysli- prognosis and the choice of treatment. Furthermore, in the case of fa-
pidemic conditions have been linked to an increased risk of cardio- milial dyslipidemias, genetic counselling and family screening need to
vascular disease. In the case of severe hypertriglyceridemia (with be implemented. First, one should differentiate primary from secondary
plasma TG levels > 10 mmol/l) there is also an increased risk of pan- hyperlipidemias. The latter are always the consequence of a different
creatitis. The nomenclature is rather simple: the term hypercholester- condition like for example obesity, hypothyroidism, nephrotic syn-
olemia is used when only plasma cholesterol is elevated, usually due to drome, obstructive liver disease, chronic renal failure or the use of
high LDL-C levels, hypertriglyceridemia is used when only plasma TG medication [5]. When these causes have been ruled out, one should
are increased and combined or mixed hyperlipidermia refers to both consider primary hyperlipidemia in which a differentiation can be


Corresponding author at: Department of Cardiology, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, the Netherlands.
E-mail address: boudewijn.klop@gmail.com (B. Klop).

https://doi.org/10.1016/j.cca.2018.09.010
Received 6 May 2018; Received in revised form 6 September 2018; Accepted 6 September 2018
Available online 07 September 2018
0009-8981/ © 2018 Elsevier B.V. All rights reserved.
M. Castro Cabezas et al. Clinica Chimica Acta 487 (2018) 117–125

Fig. 1. Schematic overview of lipid metabolism of apolipoprotein B containing lipoproteins. Abbreviations: ANGPTL = angiopoietin-like proteins, A-
V = apolipoprotein A-V, B48 = apolipoprotein B-48, B100 = apolipoprotein B-100, C = cholesterol, CM = chylomicron, CM-R = chylomicron remnant, C-
II = apolipoprotein C-II, C-III = apolipoprotein C-III, E = apolipoprotein E, FFA = free fatty acids, GPIHBP1 = glycosylphosphatidylinositol-anchored high-density-
binding protein 1, HSPG = heparan sulphate proteoglycans, LDL = low-density lipoprotein, LDLR = low-density lipoprotein receptor, LPL = lipoprotein lipase,
LRP1 = low-density lipoprotein like receptor 1, MAG = mono-acylglycerols, MTP = microsomal triglyceride transfer protein, NPC1L1 = Niemann-Pick-C1-like 1,
PCSK9 = proprotein convertase subtilisin/kexin type 9, TG = triglycerides, VLDL = very low-density lipoprotein.

made between well-defined genetic hyperlipidemias and the so-called, contrast to NPC1L1, the ATP binding cassette transporter-G5 and G8
most frequent, polygenic hypercholesterolemia in which environmental (ABCG5/8) mediates cholesterol efflux from the enterocyte to the in-
influences play a major role superimposed on a genetic predisposition. testinal lumen contributing to transintestinal cholesterol excretion [8].
Food-derived TG are taken up as free fatty acids (FFA) and 2-
monoacylglycerols (MAG) by the enterocytes after hydrolysis has taken
2. Lipid metabolism of the atherogenic lipoproteins and their
place in the intestinal lumen. These are taken up by enterocytes through
respective therapeutic targets
passive diffusion and specific proteins, like CD36 and various fatty acid
transport proteins [9]. Once inside the endoplasmatic reticulum of the
Knowledge of lipid metabolism is necessary in order to understand
enterocyte, FFA and MAG are assembled into TG again and packed with
the pathophysiologic mechanisms of the different dyslipidemias but
cholesterol, phospholipids and apo B48 and apo A-IV to form lipid-rich
also to understand which therapy will be most appropriate. Therefore,
chylomicrons, which consist for 85–92% of TG and 1–3% of cholesterol
lipoprotein metabolism will be discussed together with the clinically
[10,11]. In this process, the microsomal triglyceride transfer protein
most relevant pharmacological targets (Fig. 1).
(MTP) and the editing enzyme complex are paramount factors [12,13].
Especially the latter, which in humans is only present in the intestine, is
2.1. Intestinal chylomicron synthesis crucial for the synthesis of apo B48, the structural protein of chylomi-
crons. Apo B48 is synthesized from the apo B mRNA strand following
All atherogenic lipoproteins are synthesized either by the intestine posttranscriptional editing with enzymatic deamination at nucleotide
or the liver and are initially TG-rich lipoproteins. After food intake 6666, which results in truncation of apo B codon 2153, so that the
cholesterol is actively transported from the intestinal lumen into the mature protein consists of only 48% of the apo B100. After assembling,
enterocyte via the Niemann-Pick-C1-like 1 (NPC1L1) transporter [6]. the chylomicrons can have a diameter up to 1000 nm. They are secreted
NPC1L1 is the primary target of ezetimibe, which inhibits intestinal into the lymphatics and enter the circulation through the thoracic duct.
cholesterol absorption resulting in 20% reductions of circulating LDL-C Within the circulation apo A-IV is exchanged for two other apolipo-
levels. Ezetimibe has shown to reduce the absolute risk for major car- proteins, namely apo C-II and apo E [14]. These are involved in TG
diovascular events by 2.0% in patients with an acute coronary syn- lipoprotein lipase (LPL)-mediated hydrolysis and receptor-mediated
drome when added on top of a statin over a median of 6 years [7]. In uptake, respectively.

118
M. Castro Cabezas et al. Clinica Chimica Acta 487 (2018) 117–125

2.2. Hepatic synthesis of VLDL LRP1 and its binding affinity to the LDL-receptor is 20 times less than
apo E. In addition, only one apo B100 is present per LDL, whereas
In fasting physiological conditions, plasma TG are mainly de- several apo E molecules are present on every chylomicron remnant and
termined by the VLDL concentrations secreted by the liver. This is a VLDL [23]. Finally, LDL is internalized after binding to the LDL-re-
continuous process, which increases postprandially when TG and FFA ceptor where it is catabolized within the cell. The unesterified choles-
derived from food reach the liver. The VLDL assembly resembles chy- terol is then used for the synthesis of bile acids and the regulation of
lomicron synthesis with an important role for MTP, but in this case, apo pathways to limit cellular accumulation of cholesterol.
B100 is the structural protein of the lipoprotein. Human hepatocytes do Statins reduce the hepatic biosynthesis of cholesterol by competi-
not have an editing complex and therefore the apo B mRNA is fully tively inhibiting HMG-CoA-reductase and therefore stimulating in-
translated. The availability of TG is the most critical regulator for the creased LDL-receptor expression, as elegantly demonstrated by the
synthesis of VLDL since it lipidates the VLDL and its availability pre- Nobel prize laureates Goldstein and Brown [24]. Subsequently, hepatic
vents the proteosomal degradation of apo B100 [15]. VLDL-size may uptake of LDL from the circulation, but also of the TG-rich lipoproteins,
reach diameters up to 200 nm and nascent lipoproteins are transported is increased. This results in both lower levels of plasma LDL-C and
from the endoplasmatic reticulum of the hepatocyte to its Golgi system remnant-C with significant clinical benefit. A meta-analysis from 26
for translational modifications. Subsequently VLDL are secreted and statin trials with almost 170,000 study participants have shown that
enter the circulation via the space of Disse. statin use was associated with a 22% proportional reduction in cardi-
Nicotinic acid, or vitamin B3, is a water-soluble vitamin that in- ovascular events per mmol/l LDL-C reduction over a median of 5 years
hibits the formation of TG and lipolysis of TG from adipocytes, limiting [25,26].
the release of TG into the circulation and therefore impairing VLDL Once the LDL-receptor-bound LDL is internalized into the hepato-
secretion [12,16]. MTP-inhibitors have also been developed but did not cyte, the receptor is degraded in the lysosome when proprotein con-
reach clinical practice due to intrahepatic fat accumulation. vertase subtilisin/lexin type 9 (PCSK9) forms a complex with the LDL-
receptor and LDL. However, if PCSK9 is absent the PCSK9-mediated
2.3. Intravascular hydrolysis degradation of the LDL-receptor is inhibited allowing recycling of the
LDL-receptor and subsequent increased uptake of LDL [27]. The re-
Both, chylomicrons and VLDL are essential for the delivery of FFA to cently developed PCSK9-inhibitors are specific polyclonal antibodies
mainly the heart, skeletal muscle and adipose tissue for energy ex- against PCSK9 to increase the expression of the LDL-receptor by im-
penditure or storage. Hydrolysis of TG in the circulation is the main pairing the PCSK9-initiated LDL-receptor degradation thereby lowering
determinant of FFA delivery to tissues. Many proteins are involved in LDL-C by approximately 50–60% on top of statin therapy [28–30]. A
the hydrolysis of TG with the most important proteins being lipoprotein large randomized, double-blind, placebo-controlled trial successfully
lipase (LPL) and its co-factor apo C-II. LPL is mainly synthesized by demonstrated a further reduction in major cardiovascular events with
myocytes and adipocytes, which secrete LPL into the surrounding in- evolocumab on top of high-intensity statin therapy and results with
terstitial spaces. LPL is transported from the interstitial space to the another compound, alirocumab are promising [30,31]. Both, ezetimibe
capillary lumen after binding to heparan sulphate proteoglycans and bile acid sequestrants indirectly increase hepatic LDL-receptors.
(HSPG). At the endothelial cell surface of small capillaries LPL is co- The reduced intestinal cholesterol absorption by ezetimibe also leads to
anchored by glycosylphosphatidylinositol-anchored high-density- an increase in LDL-receptor expression. In contrast, bile acid-binding
binding protein 1 (GP1HBP1), where LPL serves as a docking station for resins remove bile acids from the enterohepatic circulation leading to
TG-rich lipoproteins [17]. Apo C-II, which is carried by chylomicrons increased hepatic production of bile acids and subsequent cholesterol
and VLDL, serves as a co-factor for LPL as is apo A-V, which facilitates catabolism, which results in a compensatory increase in hepatic LDL-
the binding of TG-rich lipoproteins to the endothelial surface via HSPG receptors, thereby stimulating LDL removal from the circulation. Since
and GPIHBP1 [18]. In contrast, apo C-III serves as an inhibitor of LPL. the introduction of statins, bile acid sequestrants have become less
Recently, it was discovered that apo C-III also impairs hydrolysis via popular in clinical practice, mainly due to weaker effects on LDL-C le-
different LPL-independent pathways [19]. The angiopoietin-like pro- vels and the gastrointestinal side effects of these drugs [16,28]. A low
teins (ANGPTL) 3, 4 and 8, which share their molecular structure with cholesterol diet also results in a similar compensatory increase in LDL-
the members of angiopoietin protein family have also been described to receptors due to its feedback regulation by the sterol regulatory ele-
reduce hydrolysis by inhibiting LPL [20]. Hydrolysis results in TG de- ment-binding preotein-1 (SREBP-1). SREBP-1 activates LDL-receptor
pleted chylomicrons and VLDL, which shrink in diameter and also be- transcription when intracellular cholesterol levels fall below a certain
come enriched with cholesterol due to the interaction with high-density threshold, which lowers plasma LDL-C levels [24].
lipoproteins (HDL) by the action of cholesterol ester transfer protein
(CETP) and other transfer proteins [21]. 3. Primary versus secondary lipid disorders
Fibrates affect different genes regulating hydrolysis including LPL,
APOC3 and APOA5 by binding to the peroxisome proliferator-activated The first step in managing lipid disorders is establishing the diag-
receptor-α. Many studies have shown that fibrates upregulate the LPL nosis. Primary dyslipidemias should be distinguished from secondary
and downregulate APOC3 expression, facilitating hydrolysis and re- lipid disorders, but environmental factors are always important de-
sulting in increased clearance of chylomicron remnants and VLDL to- terminants of the phenotypic expression. The most common lipid dis-
gether with concomitant reduced secretion of VLDL [12,16]. orders will be discussed, although it should be noted that overlap be-
tween the different lipid disorders may exist [32]. The prevalence of the
2.4. Hepatic uptake of lipoproteins most common primary lipid disorders are shown in Table 1.

Further hydrolysis of the lipoprotein remnants by hepatic lipase 3.1. Familial hypercholesterolemia
(HL), which is only present in the capillary endothelium in the liver, is
necessary for hepatic uptake by engaging the LDL-receptor (which re- Familial hypercholesterolemia (FH) is an autosomal-dominant lipid
cognizes both, apo B and apo E as ligands), HSPG or LDL receptor- disorder, which can be caused by mutations in different genes. > 90%
related protein (LRP1) via their receptor-binding apo E. This process is of the mutations are found in the LDL-receptor (LDLR) gene causing
completely sufficient for chylomicron remnants, but not for VLDL, either receptor deficiency or defective receptors, but mutations in genes
which are either taken up by the liver or loose apo E and become LDL encoding for apo B (APOB) or PCSK9 (PCSK9) can also cause FH, albeit
after HL-mediated hydrolysis [12,18,22]. Apo B100 does not bind to much less frequent (< 10%). Mutations in APOB gene lead to impaired

119
M. Castro Cabezas et al. Clinica Chimica Acta 487 (2018) 117–125

Table 1 Table 3
The prevalence of the most common primary lipid disorders in the general Causes of polygenic hypercholesterolemia.
population.
Contributing causes of polygenic hypercholesterolemia
Prevalence References
Diet High saturated fat intake
Familial hypercholesterolemia 1:250 [36] High trans fat intake
Familial combined hyperlipidemia 1:100 to 1:50 [52] High cholesterol intake
Familial hypertriglyceridemia 1:100 to 1:50 [62,122] Lifestyle Sedentary lifestyle
(TG > 10 mmol/l) Obesity
Familal dysbetalipoproteinemia 1:1000 to 1:250 [67] Diseases Cushing syndrome
Lipoprotein(a) hyperlipoproteinemia 1:20 to 1:12 [89,91] Diabetes mellitus
HIV-infection
Hypothyroidism
Insulin resistance
binding affinity with the receptor, whereas gain of function mutations
Metabolic syndrome
in the PCSK9 gene lead to enhanced degradation of receptors [33]. Nonalcoholic fatty liver disease
Currently, > 1800 mutations have been described in familial hyperch- Medication Amiodarone, betablockers, corticosteroids, cyclosporine, estrogens,
olesterolemia, but approximately 50% need further evidence before thiazide diuretics
Genetics > 30 SNPs
they can be confirmed as disease-causing mutations [34]. The national
and international registries for FH have shown to be of great relevance
to further clarify the relationship between genotypes and phenotypes
(http://www.circl.ubc.ca/english/web_fh.html) [39]. Lipid lowering
[35]. The heterozygous form of FH is one of the most common genetic
therapy is focussed on LDL-C lowering using statins, ezetimibe and, if
disorders associated with cardiovascular disease with a prevalence of
necessary PCSK9-inhibitors. In addition, treatment of other con-
approximately 1:250 [36]. Recently, it has been shown that 8.3% of
comitant cardiovascular risk factors is important to further lower the
patients with atherosclerotic cardiovascular disease have FH [37]. In
residual cardiovascular risk. Finally, cascade screening is very im-
contrast, the homozygous form of FH is very rare (1 per million). In FH,
portant to search for other family members with FH since early primary
elevated LDL-C levels may cause xanthomas and atherosclerosis with a
prevention with lipid lowering therapy is most beneficial in reducing
3- to 13-fold increased risk for the development of cardiovascular dis-
lifetime risk for cardiovascular disease. Statin therapy is generally in-
ease. The individual risk increases further when other risk factors are
itiated between the ages of 8 to 10 years in children with FH in order to
present such as obesity, insufficiently controlled hypertension, Lp(a)
achieve lifelong cardiovascular risk reduction [28]. This should only be
levels > 50 mg/dl, smoking and a history of cardiovascular disease
done in specialized centers with experience treating families.
[38].
The FH diagnostic criteria from the Dutch Lipid Clinic Network are
one of the most frequently used to establish the diagnosis. Priority is 3.2. Polygenic hypercholesterolemia
given to genetic testing, but also non-genetic criteria composed of
clinical and laboratory variables are included (Table 2). One drawback Polygenic hypercholesterolemia is the most frequent form of hy-
of these criteria is the use of untreated LDL-C levels, since establishing percholesterolemia. In comparison with FH, polygenic hypercholester-
the diagnosis is also important in patients who are already treated with olemia is caused by accumulation of small effects of lipid-affecting al-
lipid lowering therapy because of the necessity for cascade screening leles often in combination with secondary lipid modifying factors
and optimizing medical therapy once the diagnosis is established. Re- including diet, lifestyle, underlying diseases or medication (Table 3)
cently, a validated online application has been developed to impute [40,41]. Interestingly, in approximately 60% of patients with clinically
baseline LDL-C in patients already treated with a statin or ezetimibe diagnosed FH no pathogenic mutations in candidate genes can be
identified, but in up to 88% of those patients the presence of multiple
Table 2 alleles with LDL-C raising effects has been observed [42,43]. To date,
Diagnostic criteria for establishing the diagnosis familial hypercholesterolemia over 100 of single nucleotide polymorphisms (SNPs) have been dis-
(FH) using the Dutch Lipid Network Criteria. A total of > 8 points is definite covered associated with lipid disturbances including 30 SNPs affecting
FH, 6–8 points is probable FH, 3–5 points possible FH and < 3 points is unlikely LDL-C concentrations [43–46]. These include several specific SNPs in
FH [28,123]. APOB, LDLR and PCSK9 genes among others. Recently, it has been
Dutch network lipid criteria for FH Points shown that patients with clinically diagnosed FH who do not carry a
monogenic mutation may have a milder lipid phenotype and lower
Family history degree of subclinical atherosclerosis than genetically defined FH. In
First-degree relative with known premature atherosclerotic vascular 1
disease OR first degree relative with known LDL-C above 95th
addition, LDL-C had a normal distribution in those families in contrast
percentile. to a bimodal distribution in FH families, which supports the polygenic
First-degree relative with tendinous xanthomata and/or arcus 2 concept [47,48]. The contribution of risk alleles for high LDL-C varies
cornealis OR children aged < 18 years with LDL-C above 95th greatly among families underscoring the complexity and heterogeneity
percentile.
of polygenic hypercholesterolemia and the importance of secondary
Clinical history
Patient with premature coronary artery diseasea 2 lipid modifying factors [49].
Patient with premature cerebral or peripheral vascular diseasea 1 Polygenic hypercholesterolemia is characterized by moderately
Physical examination elevated levels of LDL-C, normal to mildly elevated TG but no clinical
Tendinous xanthomata 6 features such as xanthomas. The cardiovascular risk is less severely
Arcus cornealis prior to age 45 years 4
Untreated cholesterol levels
elevated than in FH [50,51]. It is important to rule out secondary causes
LDL-C ≥ 8.5 mmol/l (330 mg/dL) 8 of hypercholesterolemia. These should be eliminated or optimally
LDL-C 6.5–8.4 mmol/l (250–329 mg/dL) 5 treated before starting lipid-lowering medication. Current guidelines
LDL-C 5.0–6.4 mmol/l (190–249 mg/dL) 3 recommend to treat patients with clinically ‘definite’ or ‘probable’ FH
LDL-C 4.0–4.9 mmol/l (155–189 mg/dL) 1
on a polygenic basis, similarly intensive as patients with genetically
D0NA analysis
Functional mutation in the LDLR, APOB or PCSK9 gene 8 defined FH. Initiation of medical lipid-lowering therapy in patients with
polygenic hypercholesterolemia and a less severe phenotype should be
a
Premature ≤ 55 years in men and < 60 years in women. considered according to risk score charts [28].

120
M. Castro Cabezas et al. Clinica Chimica Acta 487 (2018) 117–125

3.3. Familial combined hyperlipidemia 3.5. Familial dysbetalipoproteinemia

The term familial combined hyperlipidemia (FCH) derives from the Familial dysbetalipoproteinemia (FD) is a recessive autosomal dis-
classical studies by Goldstein and co-workers in 1973 [52]. The pre- order caused by homozygous mutations in the apolipoprotein E gene
senting phenotype is a combined hyperlipidemia with elevated LDL-C (APOE), however 10% is caused by an autosomal dominant mutation in
and plasma TG but also elevated plasma apo B is necessary to establish APOE [67–69]. Patients with the autosomal dominant variant have the
the diagnosis [53]. FCH is characterized by increased hepatic VLDL same clinical presentation as those with the more common autosomal
secretion, predominance of small-dense LDL, impaired postprandial recessive form of FD. Three common alleles of APOE exist (E2, E3 and
chylomicron remnant and FFA metabolism and insulin resistance [54]. E4) with the E2/2 genotype present in 91% of the patients with FD,
Recently it has been shown that apo C-III, insulin, adiponectin and apo resulting in decreased binding of intermediate density lipoproteins
A-II are major determinants that affect VLDL composition in FCH after (IDL) to the LDL-receptor and remnant receptors. However, only 15% of
adjusting for waist circumference [55]. the people with the E2/2 genotype develop FD [67,69,70]. Therefore, it
This condition has been shown to have an autosomal dominant in- is thought that the development of FD is associated with additional
heritance pattern in the past, but there is no monogenic component. genetic and environmental factors, such as gender, obesity, hypothyr-
Some individuals carry a high burden of small-effect polymorphisms oidism, insulin resistance and dietary factors [71–73].
while others carry a large effect mutation in the LDLR or LPL gene. In Apo E is present on the TG-rich lipoproteins (chylomicrons and
addition, non-genetic lifestyle factors such as obesity, diet and alcohol VLDL and their respective remnants) and functions both as a structural
consumption add to the continuous quantitative trait. Therefore, the protein and as a ligand for the LDL-receptor and remnant receptors. Due
phenotype between family members can vary and also within in- to mutations, binding between apo E and its receptor is impaired with
dividuals over time [56]. FH should be included in the differential di- decreased clearance of chylomicrons, VLDL and their respective rem-
agnosis since approximately 4.8% of the patients with FCH showed a nants in combination with reduced conversion of VLDL to LDL. This will
loss of function mutation in LDLR [57]. lead to increased transfer of TG from the TG-rich lipoproteins to HDL
Therapy should focus on reducing the hepatic VLDL overproduction and transfer of cholesterolesters vice versa, leading to an increase in
and the plasma LDL concentration. For this purpose, lifestyle inter- non-HDL-C and decrease in HDLeC. This was recently demonstrated in
vention always in combination with drug therapy is warranted. Weight a clinical study, showing that patients with FD have a low HDL-C to apo
loss of approximately 9% has been shown to reduce endogenic cho- A-II ratio. In addition, non-HDL-C levels were almost doubled in FD
lesterol synthesis with a reduction of non-HDL-C levels by 9% and TG whereas apo B was only increased with 20% [67,74]. Recently, it was
by 27% [58]. The drugs of choice are statins, sometimes in combination also shown that uptake of remnants by HSPG is reduced in FD, in
with fibrates. One should be aware of the risk of myopathy and rhab- contrast to subjects with similar genotypes but a normal phenotype.
domyolysis when using this combination. In addition, periodic Therefore, it seems that remnant clearance is adequately compensated
screening for the development of T2DM is necessary since patients with in those subjects with a normal lipid phenotype but not in patients
FCH have a significantly higher risk for developing T2DM with a 10- expressing the disease [69]. All these metabolic disturbances typically
year risk of 20% when compared to age-matched patients with FH [59]. lead to elevated TG, total cholesterol and non-HDL-C levels with con-
comitant low LDL-C and HDL-C together with relatively normal apo B
3.4. Familial hypertriglyceridemia levels. Therefore, we suggest that a mismatch between elevated non-
HDL-C with a relatively normal apo B may be suggestive of FD. It
The major clinical risk in familial hypertriglyceridemia (FHTG) is should also be noted that LDL-C can be falsely elevated when the re-
pancreatitis and not cardiovascular disease [60,61]. In contrast to FCH, ported LDL-C is calculated using the Friedewald formula.
the phenotype of the dyslipidemia is not an elevated number of FD is clinically relevant since it is associated with an increased risk
atherogenic apo B containing lipoproteins, but rather the presence of for premature cardiovascular disease in index patients and their kin-
large TG-rich lipoproteins. Typically, plasma apo B is normal in FHTG dred [75,76]. Clinical features of FD are tubero-eruptive xanthomas on
reflecting normal numbers of circulating atherogenic lipoproteins. the elbows and xanthoma striatum palmare (orange and red dis-
FHTG is a polygenic disorder in contrast to the very rare familial chy- coloration on the palms) [77,78]. In patients with a mixed hyperlipi-
lomicronemia syndrome, which is a monogenic disorder with a loss of demia and these skin abnormities, DNA genotyping should be per-
function mutation in LPL or APOCII. Identification of pathogenic mu- formed. Recently it has been shown that undertreatment is common in
tations in FHTG remains challenging with rare variants found in only FD [79]. Therapy includes reduction of lipid levels by dietary and
12.3% of patients in a Spanish population study [62]. Overlap exists lifestyle modifications with special emphasis on carbohydrate and cal-
with FCH with multiple genes involved with common genetic variants. orie restriction [80,81] and lipid lowering therapy with statins and or
In FHTG, these variants have a larger effect on TG metabolism than on fibrates. Fibrates have been shown to reduce postprandial hyperlipi-
the cholesterol pathways [56,63]. In our opinion, the biggest difference demia in FD in addition to fasting lipid levels [69,82]. Non-HDL-C is
with FCH is the fact that VLDL production is not increased in FHTG. The favored over LDL-C as treatment target since FD is characterized by a
most likely reason is the fact that FFA metabolism, and specially per- mixed hyperlipidemia.
ipheral fatty acid trapping, may not be impaired in FHTG, leading to a
lesser flux of FFA to the liver. However, detailed studies on this subject 3.6. Diabetic dyslipidemia
in FHTG are lacking.
The clinical relevance to identify patients with FHTG is the asso- In uncontrolled type 1 DM the presenting dyslipidemia is usually
ciated increased risk for pancreatitis, which warrants treatment. It has hypertriglyceridemia, due to the lack of insulin and therefore severely
been shown that 5.4% of patients with TG levels > 10 mmol/l devel- impaired hydrolysis of TG-rich lipoproteins in the circulation. This is
oped acute pancreatitis one year after the diagnosis [64]. The therapy exacerbated by an uninhibited adipose tissue lipolysis causing a mas-
of choice in FHTG is lifestyle intervention with restriction of alcohol, sive release of FFA, which in the liver will give rise to large VLDL [83].
saturated fats and carbohydrates. In addition, TG lowering therapy with Of course, the most efficient and necessary therapy is insulin adminis-
fibrates or nicotinic acid should also be considered when TG remain tration and short-term starvation. In most instances this will result in a
markedly elevated [12,65]. Genetic analysis is not routinely advised rapid and dramatic decrease of plasma TG. The reason that not all
due to the low frequency of rare mutations and should only be con- subjects with type 1 DM express such a severe hypertriglyceridemia
sidered in case of strong clinical criteria for a monogenic disorder suggests that there must also be a genetic predisposition in the TG-
causing severe hypertriglyceridemia [62,66]. handling pathway for such a presentation to occur.

121
M. Castro Cabezas et al. Clinica Chimica Acta 487 (2018) 117–125

The most frequently observed diabetic dyslipidemia is the high TG/ damage, severe chronic kidney disease or a 10-year cardiovascular risk
low HDL-C type, which is characteristic for type 2 DM. The underlying of ≥10%). It is recommended to use apo B or non-HDL-C as secondary
mechanism may be an impairment of fat storage and mobilization in treatment target in the case of a combined hyperlipidemia since apo B
adipose tissue [84]. In this case subjects usually have increased levels of takes all atherogenic lipoproteins into account and non-HDL-C includes
small-dense LDL, which contributes to the increased cardiovascular remnant cholesterol besides LDL-C. The treatment target of apo B
risk. It is now generally accepted that statins reduce the cardiovascular is < 1.0 g/l for high risk patients and < 0.8 g/l in the case of very high
risk significantly in T2DM. However, some evidence has also been risk, whereas the treatment target of non-HDL-C is 0.8 mmol/l higher
provided for the use of fibrates in this condition, especially in the high than the advised LDL-C treatment target [28]. The ESC/EAS guidelines
TG/low HDL-C presentation [85]. Current guidelines recommend low- focus strongly on treatment targets, whereas the guidelines from the
ering LDL-C as primary treatment target, which automatically will de- American College of Cardiology (ACC) and American Heart Association
crease the concentrations of small-dense LDL and remnant cholesterol (AHA) focus more on the treatment process without specific treatment
[28]. Since LDL-C is not invariably elevated in diabetic dyslipidemia, targets [99].
we and others have recommended to use apo B or non-HDL-C as a Both, the ESC and ACC have recently published updates for their
secondary treatment target [28,86]. existing guidelines with special emphasis on clinical guidance for when
to consider PCSK9-inhibition in patients with cardiovascular disease or
3.7. Lipoprotein(a) hyperlipoproteinemia FH [100,101]. PCSK9-inhibition may be considered when LDL-C re-
mains > 3.6 mmol/l in patients with cardiovascular disease despite a
Lipoprotein(a) (Lp(a)) is a highly polymorphic lipoprotein particle high-intensity statin in combination with ezetimibe or when LDL-C
that consists of a large, hydrophilic glycoprotein apo (a) attached to apo remains > 2.6 mmol/l in the case of additional risk factors (rapidly
B-100 on LDL. Plasma concentrations of Lp(a) and isoform size of the progressive atherosclerotic cardiovascular disease, DM, complex mul-
apo (a) are mainly determined by genetic variation in the LPA gene tivessel or polyvascular atherosclerotic disease). A PCSK9-inhibitor may
encoding apo (a), which can vary up to 1000-fold between individuals. be considered in FH when LDL-C remains > 4.5 mmol/l on maximum
It is an independent, causal and heritable cardiovascular risk factor treatment or > 3.6 mmol/l in the case of additional risk factors
with Lp(a) plasma levels > 30 mg/dl considered atherogenic [87,88]. (marked hypertension, smoking, Lp(a) > 50 mg/dl, presence of pre-
Recently, it was shown that a 10 mg/dl increase in Lp(a) was associated mature cardiovascular disease in first degree relatives, DM with target
with a 2–4% higher likelihood of having a myocardial infarction below organ damage, > 40 years of age without treatment). The rationale
the age of 60 years [89]. In addition, Lp(a) adds to an increased car- behind these thresholds is a balance between health care costs and a
diovascular risk especially in patients with other risk factors like T2DM higher absolute risk reduction in case of higher LDL-C levels when in-
or the co-existence of FH or FCH [87,90,91]. Although Lp(a) is an es- itiating PCSK9-inhibition although cost-effectiveness studies are
tablished cardiovascular risk factor, the exact atherogenic mechanisms lacking.
of Lp(a) have not been clarified, although there is evidence that Lp(a) Patients with markedly elevated TG (≥10 mmol/l) have an in-
has antifibrinolytic and pro-inflammatory effects and promotes oxida- creased risk for pancreatitis, which warrants treatment. A paradoxical
tive stress. increase in LDL-C is frequently observed after effective TG lowering
Treatment of high levels of Lp(a) remains challenging since Lp(a) with lifestyle intervention and fibrates due to improved hydrolysis of
hyperlipoproteinemia is resistant to lifestyle interventions and there are VLDL. Subsequently, the need for additional cholesterol lowering
no specific drugs available yet to lower Lp(a) [92], besides androgens therapy can be determined by calculating the 10-year cardiovascular
and nicotinic acid. Statins and ezetimibe have little to no clinically risk. However, it was recently shown that patients with
relevant effect on Lp(a), whereas nicotinic acid and PCSK-9-inhibitors TG > 3.0 mmol/l who were not eligible for statin therapy according to
lower Lp(a) by approximately 30%, which is probably still not sufficient current guidelines showed a similar risk as statin-eligible patients in
in the case of very high Lp(a) levels [92–94]. The only available therapy primary prevention [102].
to effectively lower plasma Lp(a) levels is weekly lipoprotein apheresis,
which is recommended for patients with Lp(a) > 60 mg/dl and pro- 5. Potential new pharmaceutical strategies to treat dyslipidemias
gressive cardiovascular disease despite LDL-C within target range. Li-
poprotein apheresis reduces Lp(a) by approximately 70% with potential 5.1. Mipomersem
regression of atherosclerosis, but no prospective randomized studies on
cardiovascular outcomes have been performed yet [95–98]. Currently, Mipomersem is an antisense oligonucleotide that binds to the mRNA
the most reasonable option to treat patients with Lp(a) hyperlipopro- responsible for the synthesis of apo B100 and therefore inhibits the
teinemia is an intensified treatment of modifiable cardiovascular risk production of apo B100. It is dosed subcutaneously once weekly and
factors including lifestyle management and LDL-C lowering [28]. lowers non-HDL-C, LDL-C, TG and Lp(a) since apo B100 is an essential
component of VLDL and LDL. A phase 3 trial showed LDL-C reduction of
4. General recommendations regarding treatment of 37% in patients with hypercholesterolemia [103]. Currently, mipo-
dyslipidemias mersem is only approved in the United States for the treatment of
homozygous FH, in particular when current standard of care fails [104].
The guidelines from the European Society of Cardiology (ESC) and The most common side effects are injection site reactions, flu like
European Atherosclerosis Society (EAS) for the management of dysli- symptoms and elevations of plasma alanine aminotransferase.
pidemias advice to estimate the cumulative 10-years cardiovascular risk
in order to determine the strategy and intensity of treatment. However, 5.2. Bempedoic acid
patients with documented cardiovascular disease, T2DM, very high
levels of individual risk factors including FH or chronic kidney disease Bempedoic acid is the first in its class. It is an oral agent that inhibits
are automatically classified as high or very high risk making active adenosine triphosphate citrate lyase, which is an enzyme involved in
treatment necessary. A treatment target of LDL-C < 2.5 mmol/l is ad- the cholesterol synthesis by catalysing acetyl-CoA. It lowers LDL-C by
vised for patients with high risk (markedly elevated total cholesterol approximately 25% as monotherapy or on top of a statin and up to 48%
of > 8 mmol/l, FH, most patients with T2DM or in subjects with a 10- when combined with ezetimibe [105–107]. Efficacy and safety of
year cardiovascular risk of ≥5% to < 10%). A more strict treatment bempedoic acid on top of ezetimibe was recently confirmed in statin
target of LDL-C < 1.8 mmol/l is recommended for patients with very intolerant patients, showing similar adverse events to placebo [108].
high risk (known cardiovascular disease, T2DM with end-organ Currently, the effects of bempedoic acid on cardiovascular morbidity

122
M. Castro Cabezas et al. Clinica Chimica Acta 487 (2018) 117–125

and mortality are evaluated in the CLEAR OUTCOMES trial, which constitutive expression promotor. Up to 64 intra-muscular injections
randomizes 180 mg of bempedoic acid versus placebo in an estimated were given in major skeletal muscle groups under ultrasound guidance.
12,600 high-risk, statin intolerant patients [109]. Plasma TG reductions up to 40% were achieved at three to six weeks
after injections, but TG levels returned to baseline after 12 weeks. A
5.3. Volanesorsen 50% reduction in pancreatitis events was observed after 2 years after
treatment when compared with a historic cohort despite the dis-
A second-generation antisense inhibitor of APOC3 synthesis, vola- appearance of its TG lowering effect after 12 weeks [120]. Nevertheless,
nesorsen, lowers TG by approximately 70% in patients with hyper- alipogene tiparvovec was withdrawn in 2017 due to poor commercial
triglyceridemia [110,111]. It specifically degrades APOC3 mRNA in prospects and uncertainties about reimbursement [121].
order to prevent the production of the apo C-III protein. Therefore, li-
polysis of TG-rich lipoproteins is enhanced with concomitant reduction 6. Conclusions
in plasma TG. Several phase 3 trials have now been completed in-
cluding the APPROACH trial (in patients with familial chylomicronemia The first step in clinical care of patients with dyslipidemias is es-
syndrome) and the COMPASS trial (in patients with hypertriglycer- tablishing the diagnosis. This will determine the prognosis, the choice
idemia), which have successfully met their primary endpoint with sig- of treatment and the necessity of genetic counselling. Many dyslipide-
nificant reductions in TG of 71.2% and 77% after 26 weeks and mias have a polygenic basis interacting with lifestyle factors. Therefore,
52 weeks of treatment, respectively [112–114]. These results are pro- genetic screening is currently only advised when FH or another, but
mising for the treatment of patients with severe hypertriglyceridemia, rare monogenetic lipid disorder is considered (for example FD) due to
but large studies with coronary heart disease and pancreatitis as out- the necessity of family cascade screening with subsequent clinical
come are needed. consequences for affected family members. The pathophysiologic me-
chanism of the dyslipidemia determines the optimal pharmacological
5.4. Antisense oligonucleotides targeting apolipoprotein(a) treatment strategy besides lifestyle management in order to reach the
optimal treatment targets.
Recent advances using antisense oligonucleotides have led to the
promising development of compounds targeting apo (a) to reduce References
plasma Lp(a) concentrations. The first phase 1 study with the second
generation antisense drug to reduce the hepatic synthesis of apo(a), [1] F.K. Welty, Hypobetalipoproteinemia and abetalipoproteinemia, Curr. Opin.
Lipidol. 25 (2014) 161–168.
IONIS-APO(a)-Rx, resulted in a potent, dose-dependent reduction of
[2] J. Julve, J.M. Martin-Campos, J.C. Escola-Gil, F. Blanco-Vaca, Chylomicrons:
plasma Lp(a) up to 77.8% [115]. Its safety and efficacy was confirmed Advances in biology, pathology, laboratory testing, and therapeutics, Clin. Chim.
in a phase 2 trial (N = 64) with plasma Lp(a) reductions up to 71.6%, Acta 455 (2016) 134–148.
[3] A.J. Hooper, J.R. Burnett, Recent developments in the genetics of LDL deficiency,
but injection-site reactions occurred in 12% of the participants [116]. Curr. Opin. Lipidol. 24 (2013) 111–115.
IONIS-APO(a)-Lrx is a variant of IONIS-APO(a)-Rx, which is also in [4] H.Y. Choi, A. Hafiane, A. Schwertani, J. Genest, High-density lipoproteins:
development and allows for rapid and specific uptake within hepato- biology, epidemiology, and clinical management, Can. J. Cardiol. 33 (2017)
325–333.
cytes via the asialoglycoprotein receptor and resulted in a maximally [5] L. Kopin, C. Lowenstein, Dyslipidemia, Ann. Intern. Med. 167 (2017)
92% reduction in Lp(a) without any injection-site reactions. Therefore, ITC81–ITC96.
[6] S.W. Altmann, H.R. Davis Jr., L.J. Zhu, X. Yao, L.M. Hoos, G. Tetzloff,
IONIS-APO(a)-Lrx seems to be the most promising of the two and it may M.P. Graziano, et al., Niemann-Pick C1 Like 1 protein is critical for intestinal
reduce Lp(a) hyperlipoproteinemia-related cardiovascular risk or pro- cholesterol absorption, Science 303 (2004) 1201–1204.
gression of calcified aortic valve stenosis [116]. [7] C.P. Cannon, M.A. Blazing, R.P. Giugliano, A. McCagg, J.A. White, P. Theroux, I.-
I. Investigators, Ezetimibe added to statin therapy after acute coronary syndromes,
N. Engl. J. Med. 372 (2015) 2387–2397.
5.5. CETP-inhibition [8] J.F. de Boer, F. Kuipers, A.K. Groen, Cholesterol transport revisited: a new turbo
mechanism to drive cholesterol excretion, Trends Endocrinol. Metab. 29 (2018)
123–133.
Inhibitors of CETP were developed in order to raise HDL-C levels, [9] X. Pan, M.M. Hussain, Gut triglyceride production, Biochim. Biophys. Acta 1821
but they lower LDL-C as well, albeit to a lesser extent than statins. (2012) 727–735.
[10] J. Iqbal, M.M. Hussain, Intestinal lipid absorption, Am. J. Physiol. Endocrinol.
Despite their favourite modulation of the lipid profile, most CETP-in- Metab. 296 (2009) E1183–E1194.
hibitors have not shown its potential to lower cardiovascular risk. [11] C.M. Mansbach, S.A. Siddiqi, The biogenesis of chylomicrons, Annu. Rev. Physiol.
Torcetrapib, which was the first CETP-inhibitor that reached an ad- 72 (2010) 315–333.
[12] B. Klop, J. Wouter Jukema, T.J. Rabelink, M. Castro Cabezas, A physician's guide
vanced stage of clinical development, showed a 58% increase in mor- for the management of hypertriglyceridemia: the etiology of hypertriglyceridemia
tality and 25% increase in the primary cardiovascular endpoint during determines treatment strategy, Panminerva Med. 54 (2012) 91–103.
[13] G.K. Leung, M.M. Veniant, S.K. Kim, C.H. Zlot, M. Raabe, J. Bjorkegren,
the prematurely stopped ILLUMINATE study [117]. There is evidence
S.G. Young, et al., A deficiency of microsomal triglyceride transfer protein reduces
that the disappointing results of torcetrapib were due to off-target apolipoprotein B secretion, J. Biol. Chem. 275 (2000) 7515–7520.
toxicity and not due to CETP-inhibition itself. Currently, anacetrapib is [14] E.J. Schaefer, M.G. Wetzel, G. Bengtsson, R.O. Scow, H.B. Brewer Jr.,
T. Olivecrona, Transfer of human lymph chylomicron constituents to other lipo-
the only CETP-inhibitor that showed a reduction in cardiovascular protein density fractions during in vitro lipolysis, J. Lipid Res. 23 (1982)
events, with a modest but significant 9% reduction in cardiovascular 1259–1273.
events during a mean follow-up of 4.1 years. This outcome was prob- [15] S.O. Olofsson, J. Boren, Apolipoprotein B: a clinically important apolipoprotein
which assembles atherogenic lipoproteins and promotes the development of
ably mainly driven by its LDL-C lowering effect [118]. It was chosen not atherosclerosis, J. Intern. Med. 258 (2005) 395–410.
to pursue regulatory approval for anacetrapib due to its modest clinical [16] M.S. Kelly, C. Beavers, J.D. Bucheit, E.M. Sisson, D.L. Dixon, Pharmacologic ap-
proaches for the management of patients with moderately elevated triglycerides
benefit. It remains uncertain whether CETP-inhibition with further (150-499 mg/dL), J. Clin. Lipidol. 11 (2017) 872–879.
improved agents will result in a potential cardioprotective strategy [17] B.S. Davies, A.P. Beigneux, R.H. Barnes 2nd, Y. Tu, P. Gin, M.M. Weinstein,
[119]. L.G. Fong, et al., GPIHBP1 is responsible for the entry of lipoprotein lipase into
capillaries, Cell Metab. 12 (2010) 42–52.
[18] A. Wolska, R.L. Dunbar, L.A. Freeman, M. Ueda, M.J. Amar, D.O. Sviridov,
5.6. Gene therapy A.T. Remaley, Apolipoprotein C-II: New findings related to genetics, biochemistry,
and role in triglyceride metabolism, Atherosclerosis 267 (2017) 49–60.
[19] D. Gaudet, D. Brisson, K. Tremblay, V.J. Alexander, W. Singleton, S.G. Hughes,
Alipogene tiparvovec was the first registered gene therapy in Europe J.L. Witztum, et al., Targeting APOC3 in the familial chylomicronemia syndrome,
and approved for the treatment of patients with a chylomicronemia N. Engl. J. Med. 371 (2014) 2200–2206.
[20] Y. Li, C. Teng, Angiopoietin-like proteins 3, 4 and 8: regulating lipid metabolism
syndrome due to LPL-deficiency. Its viral vector was based on an adeno- and providing new hope for metabolic syndrome, J. Drug Target. 22 (2014)
associated virus, which contained the LPL S447X gene construct with a

123
M. Castro Cabezas et al. Clinica Chimica Acta 487 (2018) 117–125

679–687. [49] I. Lamiquiz-Moneo, M.R. Perez-Ruiz, E. Jarauta, M.T. Tejedor, A.M. Bea, R. Mateo-
[21] G.D. Kolovou, K.K. Anagnostopoulou, P.M. Kostakou, D.P. Mikhailidis, Cholesterol Gallego, F. Civeira, et al., Single nucleotide variants associated with polygenic
ester transfer protein (CETP), postprandial lipemia and hypolipidemic drugs, Curr. hypercholesterolemia in families diagnosed clinically with familial hypercholes-
Med. Chem. 16 (2009) 4345–4360. terolemia, Rev. Esp. Cardiol. (Engl. Ed.) 71 (2018) 351–356.
[22] D.L. Brasaemle, K. Cornely-Moss, A. Bensadoun, Hepatic lipase treatment of chy- [50] B.A. Ference, W. Yoo, I. Alesh, N. Mahajan, K.K. Mirowska, A. Mewada, J.M. Flack,
lomicron remnants increases exposure of apolipoprotein E, J. Lipid Res. 34 (1993) et al., Effect of long-term exposure to lower low-density lipoprotein cholesterol
455–465. beginning early in life on the risk of coronary heart disease: a Mendelian rando-
[23] W.A. Bradley, S.L. Hwang, J.B. Karlin, A.H. Lin, S.C. Prasad, A.M. Gotto Jr., mization analysis, J. Am. Coll. Cardiol. 60 (2012) 2631–2639.
S.H. Gianturco, Low-density lipoprotein receptor binding determinants switch [51] H.H. Schmidt, S. Hill, E.V. Makariou, I.M. Feuerstein, K.A. Dugi, J.M. Hoeg,
from apolipoprotein E to apolipoprotein B during conversion of hypertriglyceri- Relation of cholesterol-year score to severity of calcific atherosclerosis and tissue
demic very-low-density lipoprotein to low-density lipoproteins, J. Biol. Chem. 259 deposition in homozygous familial hypercholesterolemia, Am. J. Cardiol. 77
(1984) 14728–14735. (1996) 575–580.
[24] J.L. Goldstein, M.S. Brown, A century of cholesterol and coronaries: from plaques [52] J.L. Goldstein, H.G. Schrott, W.R. Hazzard, E.L. Bierman, A.G. Motulsky,
to genes to statins, Cell 161 (2015) 161–172. Hyperlipidemia in coronary heart disease. II. Genetic analysis of lipid levels in 176
[25] C. Cholesterol Treatment Trialists, C. Baigent, L. Blackwell, J. Emberson, families and delineation of a new inherited disorder, combined hyperlipidemia, J.
L.E. Holland, C. Reith, R. Collins, et al., Efficacy and safety of more intensive Clin. Invest. 52 (1973) 1544–1568.
lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in [53] A.D. Sniderman, M. Castro Cabezas, J. Ribalta, R. Carmena, T.W. de Bruin, J. de
26 randomised trials, Lancet 376 (2010) 1670–1681. Graaf, M.R. Taskinen, et al., A proposal to redefine familial combined hyperlipi-
[26] B.A. Ference, H.N. Ginsberg, I. Graham, K.K. Ray, C.J. Packard, E. Bruckert, daemia – third workshop on FCHL held in Barcelona from 3 to 5 May 2001, during
A.L. Catapano, et al., Low-density lipoproteins cause atherosclerotic cardiovas- the scientific sessions of the European Society for Clinical Investigation, Eur. J.
cular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A Clin. Investig. 32 (2002) 71–73.
consensus statement from the European Atherosclerosis Society Consensus Panel, [54] M. Castro Cabezas, T.W. de Bruin, H.W. de Valk, C.C. Shoulders, H. Jansen,
Eur. Heart J. 38 (2017) 2459–2472. D. Willem Erkelens, Impaired fatty acid metabolism in familial combined hy-
[27] S. Krahenbuhl, I. Pavik-Mezzour, A. von Eckardstein, Unmet Needs in LDL-C perlipidemia. A mechanism associating hepatic apolipoprotein B overproduction
Lowering: When Statins Won't Do!, Drugs 76 (2016) 1175–1190. and insulin resistance, J. Clin. Invest. 92 (1993) 160–168.
[28] A.L. Catapano, I. Graham, G. De Backer, O. Wiklund, M.J. Chapman, H. Drexel, [55] I. Cruz-Bautista, R. Mehta, J. Cabiedes, C. Garcia-Ulloa, L.E. Guillen-Pineda,
E.S.C.S.D. Group, ESC/EAS guidelines for the management of dyslipidaemias, Eur. P. Almeda-Valdes, C.A. Aguilar-Salinas, et al., Determinants of VLDL composition
Heart J. 37 (2016) (2016) 2999–3058. and apo B-containing particles in familial combined hyperlipidemia, Clin. Chim.
[29] E.A. Stein, S. Mellis, G.D. Yancopoulos, N. Stahl, D. Logan, W.B. Smith, Acta 438 (2015) 160–165.
G.D. Swergold, et al., Effect of a monoclonal antibody to PCSK9 on LDL choles- [56] A.J. Brahm, R.A. Hegele, Combined hyperlipidemia: familial but not (usually)
terol, N. Engl. J. Med. 366 (2012) 1108–1118. monogenic, Curr. Opin. Lipidol. 27 (2016) 131–140.
[30] M.S. Sabatine, R.P. Giugliano, A.C. Keech, N. Honarpour, S.D. Wiviott, [57] I. Minicocci, C. Prisco, A. Montali, A. Di Costanzo, F. Ceci, G. Pigna, M. Arca,
S.A. Murphy, et al., Evolocumab and clinical outcomes in patients with cardio- Contribution of mutations in low density lipoprotein receptor (LDLR) and lipo-
vascular disease, N. Engl. J. Med. 376 (2017) 1713–1722. protein lipase (LPL) genes to familial combined hyperlipidemia (FCHL): a re-
[31] J.G. Robinson, M. Farnier, M. Krempf, J. Bergeron, G. Luc, M. Averna, appraisal by using a resequencing approach, Atherosclerosis 242 (2015) 618–624.
O.L.T. Investigators, Efficacy and safety of alirocumab in reducing lipids and [58] R. Mateo-Gallego, S. Perez-Calahorra, M. Cofan, L. Baila-Rueda, A. Cenarro,
cardiovascular events, N. Engl. J. Med. 372 (2015) 1489–1499. E. Ros, F. Civeira, et al., Serum lipid responses to weight loss differ between
[32] K.L. Ellis, J. Pang, D.C. Chan, A.J. Hooper, D.A. Bell, J.R. Burnett, G.F. Watts, overweight adults with familial hypercholesterolemia and those with familial
Familial combined hyperlipidemia and hyperlipoprotein(a) as phenotypic mimics combined hyperlipidemia, J. Nutr. 144 (2014) 1219–1226.
of familial hypercholesterolemia: Frequencies, associations and predictions, J. [59] I. Skoumas, N. Ioakeimidis, C. Vlachopoulos, C. Chrysohoou, C. Michalakeas,
Clin. Lipidol. 10 (2016) 1329–1337 (e1323). C. Georgakopoulos, D. Tousoulis, et al., Statin therapy and risk of diabetes mellitus
[33] K. Sharma, R.R. Baliga, Genetics of dyslipidemia and ischemic heart disease, Curr. in aging patients with heterozygous familial hypercholesterolemia or familial
Cardiol. Rep. 19 (2017) 46. combined hyperlipidemia: a 10-year follow-up, Angiology 69 (2018) 242–248.
[34] M. Bourbon, A.C. Alves, E.J. Sijbrands, Low-density lipoprotein receptor muta- [60] J.D. Brunzell, H.G. Schrott, A.G. Motulsky, E.L. Bierman, Myocardial infarction in
tional analysis in diagnosis of familial hypercholesterolemia, Curr. Opin. Lipidol. the familial forms of hypertriglyceridemia, Metabolism 25 (1976) 313–320.
28 (2017) 120–129. [61] L.H. Deng, P. Xue, Q. Xia, X.N. Yang, M.H. Wan, Effect of admission hyper-
[35] I. Kindt, P. Mata, J.W. Knowles, The role of registries and genetic databases in triglyceridemia on the episodes of severe acute pancreatitis, World J.
familial hypercholesterolemia, Curr. Opin. Lipidol. 28 (2017) 152–160. Gastroenterol. 14 (2008) 4558–4561.
[36] J. Genest, Familial hypercholesterolemia: awareness, appraisal, and action, Can. J. [62] I. Lamiquiz-Moneo, C. Blanco-Torrecilla, A.M. Bea, R. Mateo-Gallego, S. Perez-
Cardiol. 33 (2017) 298–299. Calahorra, L. Baila-Rueda, I. de Castro-Oros, et al., Frequency of rare mutations
[37] G. De Backer, J. Besseling, J. Chapman, G.K. Hovingh, J.J. Kastelein, K. Kotseva, and common genetic variations in severe hypertriglyceridemia in the general
E. Investigators, et al., Prevalence and management of familial hypercholester- population of Spain, Lipids Health Dis. 15 (2016) 82.
olaemia in coronary patients: an analysis of EUROASPIRE IV, a study of the [63] I. De Castro-Oros, A. Cenarro, M.T. Tejedor, L. Baila-Rueda, R. Mateo-Gallego,
European Society of Cardiology, Atherosclerosis 241 (2015) 169–175. I. Lamiquiz-Moneo, F. Civeira, et al., Common genetic variants contribute to pri-
[38] L. Perez De Isla, R. Alonso, N. Mata, C. Fernandez-Perez, O. Muniz, J.L. Diaz-Diaz, mary hypertriglyceridemia without differences between familial combined hy-
P. Mata, et al., Predicting cardiovascular events in familial hypercholesterolemia: perlipidemia and isolated hypertriglyceridemia, Circ. Cardiovasc. Genet. 7 (2014)
the SAFEHEART registry (Spanish Familial Hypercholesterolemia Cohort Study), 814–821.
Circulation 135 (2017) 2133–2144. [64] N. Rashid, P.P. Sharma, R.D. Scott, K.J. Lin, P.P. Toth, Severe hypertriglyceridemia
[39] I. Ruel, S. Aljenedil, I. Sadri, E. de Varennes, R.A. Hegele, P. Couture, J. Genest, and factors associated with acute pancreatitis in an integrated health care system,
et al., Imputation of baseline LDL cholesterol concentration in patients with fa- J. Clin. Lipidol. 10 (2016) 880–890.
milial hypercholesterolemia on statins or ezetimibe, Clin. Chem. 64 (2018) [65] D.J. Rader, New Therapeutic Approaches to the Treatment of Dyslipidemia, Cell
355–362. Metab. 23 (2016) 405–412.
[40] R.A. Hegele, Plasma lipoproteins: genetic influences and clinical implications, Nat. [66] A.B. Cefalu, R. Spina, D. Noto, V. Ingrassia, V. Valenti, A. Giammanco,
Rev. Genet. 10 (2009) 109–121. M.R. Averna, et al., Identification of a novel LMF1 nonsense mutation responsible
[41] N.J. Stone, Secondary causes of hyperlipidemia, Med. Clin. N. Am. 78 (1994) for severe hypertriglyceridemia by targeted next-generation sequencing, J. Clin.
117–141. Lipidol. 11 (2017) 272–281 (e278).
[42] M. Futema, S. Shah, J.A. Cooper, K. Li, R.A. Whittall, M. Sharifi, S.E. Humphries, [67] C. Koopal, A.D. Marais, F.L. Visseren, Familial dysbetalipoproteinemia: an un-
et al., Refinement of variant selection for the LDL cholesterol genetic risk score in derdiagnosed lipid disorder, Curr. Opin. Endocrinol. Diabetes Obes. 24 (2017)
the diagnosis of the polygenic form of clinical familial hypercholesterolemia and 133–139.
replication in samples from 6 countries, Clin. Chem. 61 (2015) 231–238. [68] V.I. Zannis, Genetic polymorphism in human apolipoprotein E, Methods Enzymol.
[43] P.J. Talmud, S. Shah, R. Whittall, M. Futema, P. Howard, J.A. Cooper, 128 (1986) 823–851.
S.E. Humphries, et al., Use of low-density lipoprotein cholesterol gene score to [69] C. Koopal, A.D. Marais, J. Westerink, F.L. Visseren, Autosomal dominant familial
distinguish patients with polygenic and monogenic familial hypercholester- dysbetalipoproteinemia: a pathophysiological framework and practical approach
olaemia: a case-control study, Lancet 381 (2013) 1293–1301. to diagnosis and therapy, J. Clin. Lipidol. 11 (2017) 12–23 (e11).
[44] J.A. Hubacek, V. Adamkova, V. Lanska, D. Dlouha, Polygenic hypercholester- [70] P.N. Hopkins, L.L. Wu, S.C. Hunt, E.A. Brinton, Plasma triglycerides and type III
olemia: examples of GWAS results and their replication in the Czech-Slavonic hyperlipidemia are independently associated with premature familial coronary
population, Physiol. Res. 66 (2017) S101–S111. artery disease, J. Am. Coll. Cardiol. 45 (2005) 1003–1012.
[45] S. Kathiresan, C.J. Willer, G.M. Peloso, S. Demissie, K. Musunuru, E.E. Schadt, [71] F. de Beer, A.F. Stalenhoef, N. Hoogerbrugge, J.J. Kastelein, J.A. Gevers Leuven,
L.A. Cupples, et al., Common variants at 30 loci contribute to polygenic dyslipi- C.M. van Duijn, A.H. Smelt, et al., Expression of type III hyperlipoproteinemia in
demia, Nat. Genet. 41 (2009) 56–65. apolipoprotein E2 (Arg158 → Cys) homozygotes is associated with hyper-
[46] C.J. Willer, K.L. Mohlke, Finding genes and variants for lipid levels after genome- insulinemia, Arterioscler. Thromb. Vasc. Biol. 22 (2002) 294–299.
wide association analysis, Curr. Opin. Lipidol. 23 (2012) 98–103. [72] C. Koopal, Y. van der Graaf, F.W. Asselbergs, J. Westerink, F.L. Visseren, Influence
[47] E. Jarauta, M.R. Perez-Ruiz, S. Perez-Calahorra, R. Mateo-Gallego, A. Cenarro, of APOE-2 genotype on the relation between adiposity and plasma lipid levels in
M. Cofan, M.T. Tejedor, et al., Lipid phenotype and heritage pattern in families patients with vascular disease, Int. J. Obes. 39 (2015) 265–269.
with genetic hypercholesterolemia not related to LDLR, APOB, PCSK9, or APOE, J. [73] E.J. Sijbrands, M.J. Hoffer, A.E. Meinders, L.M. Havekes, R.R. Frants, A.H. Smelt,
Clin. Lipidol. 10 (2016) 1397–1405 (e1392). P. De Knijff, Severe hyperlipidemia in apolipoprotein E2 homozygotes due to a
[48] M. Sharifi, E. Higginson, S. Bos, A. Gallivan, D. Harvey, K.W. Li, S.E. Humphries, combined effect of hyperinsulinemia and an SstI polymorphism, Arterioscler.
et al., Greater preclinical atherosclerosis in treated monogenic familial hyperch- Thromb. Vasc. Biol. 19 (1999) 2722–2729.
olesterolemia vs. polygenic hypercholesterolemia, Atherosclerosis 263 (2017) [74] J.P. Corsetti, C.E. Sparks, S.J.L. Bakker, E.G. Gruppen, R.P.F. Dullaart, Roles of
405–411. high apolipoprotein E blood levels and HDL in development of familial

124
M. Castro Cabezas et al. Clinica Chimica Acta 487 (2018) 117–125

dysbetalipoproteinemia in epsilon2epsilon2 subjects, Clin. Biochem. 52 (2018) guidance for proprotein convertase subtilisin/kexin type 9 inhibition in patients
67–72. with atherosclerotic cardiovascular disease or in familial hypercholesterolaemia,
[75] P. Henneman, F. van der Sman-de Beer, P.H. Moghaddam, P. Huijts, Eur. Heart J. 39 (2018) 1131–1143.
A.F. Stalenhoef, J.J. Kastelein, A.H. Smelt, et al., The expression of type III hy- [101] D.M. Lloyd-Jones, P.B. Morris, C.M. Ballantyne, K.K. Birtcher, D.D. Daly Jr.,
perlipoproteinemia: involvement of lipolysis genes, Eur. J. Hum. Genet. 17 (2009) S.M. Depalma, S.C. Smith Jr.et al., 2017 focused update of the 2016 ACC expert
620–628. consensus decision pathway on the role of non-statin therapies for LDL-cholesterol
[76] P.N. Hopkins, M.N. Nanjee, L.L. Wu, M.G. McGinty, E.A. Brinton, S.C. Hunt, lowering in the management of atherosclerotic cardiovascular disease risk: a re-
J.L. Anderson, Altered composition of triglyceride-rich lipoproteins and coronary port of the American College of Cardiology Task Force on Expert Consensus
artery disease in a large case-control study, Atherosclerosis 207 (2009) 559–566. Decision Pathways, J. Am. Coll. Cardiol. 70 (2017) 1785–1822.
[77] M. Rothschild, G. Duhon, R. Riaz, V. Jetty, N. Goldenberg, C.J. Glueck, P. Wang, [102] C.M. Madsen, A. Varbo, B.G. Nordestgaard, Unmet need for primary prevention in
Pathognomonic Palmar Crease Xanthomas of Apolipoprotein E2 Homozygosity- individuals with hypertriglyceridaemia not eligible for statin therapy according to
Familial Dysbetalipoproteinemia, JAMA Dermatol. 152 (2016) 1275–1276. European Society of Cardiology/European Atherosclerosis Society guidelines: a
[78] A.H. Smelt, F. de Beer, Apolipoprotein E and familial dysbetalipoproteinemia: contemporary population-based study, Eur. Heart J. 39 (2018) 610–619.
clinical, biochemical, and genetic aspects, Semin. Vasc. Med. 4 (2004) 249–257. [103] D.N. Ricotta, W. Frishman, Mipomersen: a safe and effective antisense therapy
[79] C. Koopal, K. Retterstol, B. Sjouke, G.K. Hovingh, E. Ros, J. de Graaf, F.L. Visseren, adjunct to statins in patients with hypercholesterolemia, Cardiol. Rev. 20 (2012)
et al., Vascular risk factors, vascular disease, lipids and lipid targets in patients 90–95.
with familial dysbetalipoproteinemia: a European cross-sectional study, [104] D. Zodda, R. Giammona, S. Schifilliti, Treatment strategy for dyslipidemia in
Atherosclerosis 240 (2015) 90–97. cardiovascular disease prevention: focus on old and new drugs, Pharmacy (Basel)
[80] C.B. Blum, Type III Hyperlipoproteinemia: Still Worth Considering? Prog. 6 (2018).
Cardiovasc. Dis. 59 (2016) 119–124. [105] C.M. Ballantyne, M.H. Davidson, D.E. MacDougall, H.E. Bays, L.A. Dicarlo,
[81] G. Riccardi, O. Vaccaro, G. Costabile, A.A. Rivellese, How Well Can We Control N.L. Rosenberg, R.S. Newton, et al., Efficacy and safety of a novel dual modulator
Dyslipidemias Through Lifestyle Modifications? Curr. Cardiol. Rep. 18 (2016) 66. of adenosine triphosphate-citrate lyase and adenosine monophosphate-activated
[82] C. Koopal, A.D. Marais, J. Westerink, Y. van der Graaf, F.L.J. Visseren, Effect of protein kinase in patients with hypercholesterolemia: results of a multicenter,
adding bezafibrate to standard lipid-lowering therapy on post-fat load lipid levels randomized, double-blind, placebo-controlled, parallel-group trial, J. Am. Coll.
in patients with familial dysbetalipoproteinemia. A randomized placebo-con- Cardiol. 62 (2013) 1154–1162.
trolled crossover trial, J. Lipid. Res. 58 (2017) 2180–2187. [106] C.M. Ballantyne, J.M. McKenney, D.E. MacDougall, J.R. Margulies, P.L. Robinson,
[83] B. Verges, Lipid disorders in type 1 diabetes, Diabetes Metab. 35 (2009) 353–360. J.C. Hanselman, N.D. Lalwani, Effect of ETC-1002 on serum low-density lipopro-
[84] G.F. Lewis, A. Carpentier, K. Adeli, A. Giacca, Disordered fat storage and mobili- tein cholesterol in hypercholesterolemic patients receiving statin therapy, Am. J.
zation in the pathogenesis of insulin resistance and type 2 diabetes, Endocr. Rev. Cardiol. 117 (2016) 1928–1933.
23 (2002) 201–229. [107] P.D. Thompson, D.E. MacDougall, R.S. Newton, J.R. Margulies, J.C. Hanselman,
[85] A.S. Group, H.N. Ginsberg, M.B. Elam, L.C. Lovato, J.R. Crouse 3rd, L.A. Leiter, D.G. Orloff, C.M. Ballantyne, et al., Treatment with ETC-1002 alone and in com-
R.P. Byington, et al., Effects of combination lipid therapy in type 2 diabetes bination with ezetimibe lowers LDL cholesterol in hypercholesterolemic patients
mellitus, N. Engl. J. Med. 362 (2010) 1563–1574. with or without statin intolerance, J. Clin. Lipidol. 10 (2016) 556–567.
[86] P.J. Barter, C.M. Ballantyne, R. Carmena, M. Castro Cabezas, M.J. Chapman, [108] C.M. Ballantyne, M. Banach, G.B.J. Mancini, N.E. Lepor, J.C. Hanselman, X. Zhao,
P. Couture, K.M. Williams, et al., Apo B versus cholesterol in estimating cardio- L.A. Leiter, Efficacy and safety of bempedoic acid added to ezetimibe in statin-
vascular risk and in guiding therapy: report of the thirty-person/ten-country panel, intolerant patients with hypercholesterolemia: A randomized, placebo-controlled
J. Intern. Med. 259 (2006) 247–258. study, Atherosclerosis (2018 Jun 12), https://doi.org/10.1016/j.atherosclerosis.
[87] K.L. Ellis, M.B. Boffa, A. Sahebkar, M.L. Koschinsky, G.F. Watts, The renaissance of 2018.06.002 pii: S0021-9150(18)30309-5.
lipoprotein(a): Brave new world for preventive cardiology? Prog. Lipid Res. 68 [109] G. Polychronopoulos, K. Tziomalos, Novel treatment options for the management
(2017) 57–82. of heterozygous familial hypercholesterolemia, Expert. Rev. Clin. Pharmacol. 10
[88] F. Rigamonti, F. Carbone, F. Montecucco, A. Bonaventura, L. Liberale, F. Burger, (2017) 1375–1381.
L. Pisciotta, et al., Serum lipoprotein (a) predicts acute coronary syndromes in [110] D. Gaudet, V.J. Alexander, B.F. Baker, D. Brisson, K. Tremblay, W. Singleton,
patients with severe carotid stenosis, Eur. J. Clin Invest. 48 (2018). J.J. Kastelein, et al., Antisense inhibition of apolipoprotein C-III in patients with
[89] L.S. Rallidis, G. Pavlakis, A. Foscolou, C. Kotakos, A. Katsimardos, A. Drosatos, hypertriglyceridemia, N. Engl. J. Med. 373 (2015) 438–447.
D.B. Panagiotakos, et al., High levels of lipoprotein (a) and premature acute cor- [111] I. Gouni-Berthold, The role of antisense oligonucleotide therapy against apolipo-
onary syndrome, Atherosclerosis 269 (2018) 29–34. protein-CIII in hypertriglyceridemia, Atheroscler. Suppl. 30 (2017) 19–27.
[90] S. Li, N.Q. Wu, C.G. Zhu, Y. Zhang, Y.L. Guo, Y. Gao, J.J. Li, et al., Significance of [112] N.A. Rocha, C. East, J. Zhang, P.A. McCullough, ApoCIII as a cardiovascular risk
lipoprotein(a) levels in familial hypercholesterolemia and coronary artery disease, factor and modulation by the novel lipid-lowering agent volanesorsen, Curr.
Atherosclerosis 260 (2017) 67–74. Atheroscler. Rep. 19 (2017) 62.
[91] C. Waldeyer, N. Makarova, T. Zeller, R.B. Schnabel, F.J. Brunner, T. Jorgensen, [113] D. Gaudet, A. Digenio, V.J. Alexander, M. Arca, A.F. Jones, E. Stroes, et al., The
S. Blankenberg, et al., Lipoprotein(a) and the risk of cardiovascular disease in the APPROACH study: a randomized, double-blind, placebo-controlled, phase 3 study
European population: results from the BiomarCaRE consortium, Eur. Heart J. 38 of volanesorsen administered subcutaneously to patients with familial chylomi-
(2017) 2490–2498. cronemia syndrome (FCS), J. Clin. Lipidol. 11 (2017) 814–815.
[92] C.A. Scipione, M.L. Koschinsky, M.B. Boffa, Lipoprotein(a) in clinical practice: [114] I. Gouni-Berthold, V. Alexander, A. Digenio, R. Dufour, E. Steinhagen-Thiessen,
New perspectives from basic and translational science, Crit. Rev. Clin. Lab. Sci. 55 S. Martin, E. Stroes, et al., Apolipoprotein C-III inhibition with volanesorsen in
(2018) 33–54. patients with hypertriglyceridemia (COMPASS): a randomized, double-blind,
[93] K. Awad, D.P. Mikhailidis, N. Katsiki, P. Muntner, M. BanachLipid, G. Blood placebo-controlled trial, J. Clin. Lipidol. 11 (2017) 794–795.
Pressure Meta-Analysis Collaboration, Effect of ezetimibe monotherapy on plasma [115] S. Tsimikas, N.J. Viney, S.G. Hughes, W. Singleton, M.J. Graham, B.F. Baker,
lipoprotein(a) concentrations in patients with primary hypercholesterolemia: a J.L. Witztum, et al., Antisense therapy targeting apolipoprotein(a): a randomised,
systematic review and meta-analysis of randomized controlled trials, Drugs 78 double-blind, placebo-controlled phase 1 study, Lancet 386 (2015) 1472–1483.
(2018) 453–462. [116] N.J. Viney, J.C. van Capelleveen, R.S. Geary, S. Xia, J.A. Tami, R.Z. Yu,
[94] B. Enkhmaa, E. Anuurad, W. Zhang, K. Yue, C.S. Li, L. Berglund, The roles of apo S. Tsimikas, et al., Antisense oligonucleotides targeting apolipoprotein(a) in
(a) size, phenotype, and dominance pattern in PCSK9-inhibition-induced reduc- people with raised lipoprotein(a): two randomised, double-blind, placebo-con-
tion in Lp(a) with alirocumab, J. Lipid Res. 58 (2017) 2008–2016. trolled, dose-ranging trials, Lancet 388 (2016) 2239–2253.
[95] B. Hohenstein, U. Julius, P. Lansberg, B. Jaeger, K.P. Mellwig, N. Weiss, [117] P.J. Barter, M. Caulfield, M. Eriksson, S.M. Grundy, J.J. Kastelein, M. Komajda,
W. Ramlow, et al., Rationale and design of MultiSELECt: A European Multicenter et al., Investigators. Effects of torcetrapib in patients at high risk for coronary
Study on the effect of lipoprotein(a) elimination by lipoprotein apheresis on car- events, N. Engl. J. Med. 357 (2007) 2109–2122.
diovascular outcomes, Atheroscler. Suppl. 30 (2017) 180–186. [118] H.T.R.C. Group, L. Bowman, J.C. Hopewell, F. Chen, K. Wallendszus, W. Stevens,
[96] J. Leebmann, E. Roeseler, U. Julius, F. Heigl, R. Spitthoever, D. Heutling, et al., M.J. Landray, et al., Effects of anacetrapib in patients with atherosclerotic vascular
Lipoprotein apheresis in patients with maximally tolerated lipid-lowering therapy, disease, N. Engl. J. Med. 377 (2017) 1217–1227.
lipoprotein(a)-hyperlipoproteinemia, and progressive cardiovascular disease: [119] S.J. Nicholls, CETP-inhibition and HDL-cholesterol: a story of CV risk or CV ben-
prospective observational multicenter study, Circulation 128 (2013) 2567–2576. efit, or both, Clin. Pharmacol. Ther. 104 (2018) 297–300.
[97] B.R. Jaeger, Y. Richter, D. Nagel, F. Heigl, A. Vogt, E. Roeseler, et al., Longitudinal [120] D. Gaudet, E.S. Stroes, J. Methot, D. Brisson, K. Tremblay, S.J. Bernelot Moens,
cohort study on the effectiveness of lipid apheresis treatment to reduce high li- M.J. Bruno, et al., Long-term retrospective analysis of gene therapy with alipogene
poprotein(a) levels and prevent major adverse coronary events, Nat. Clin. Pract. tiparvovec and its effect on lipoprotein lipase deficiency-induced pancreatitis,
Cardiovasc. Med. 6 (2009) 229–239. Hum. Gene Ther. 27 (2016) 916–925.
[98] F. Heigl, R. Hettich, N. Lotz, H. Reeg, T. Pflederer, D. Osterkorn, R. Klingel, et al., [121] R. Chaudhry, A. Viljoen, A.S. Wierzbicki, Pharmacological treatment options for
Efficacy, safety, and tolerability of long-term lipoprotein apheresis in patients with severe hypertriglyceridemia and familial chylomicronemia syndrome, Expert. Rev.
LDL- or Lp(a) hyperlipoproteinemia: Findings gathered from more than 36,000 Clin. Pharmacol. 11 (2018) 589–598.
treatments at one center in Germany, Atheroscler. Suppl. 18 (2015) 154–162. [122] E.S. Ford, C. Li, G. Zhao, W.S. Pearson, A.H. Mokdad, Hypertriglyceridemia and its
[99] I.M. Graham, A.L. Catapano, Management of dyslipidemias in Europe and the pharmacologic treatment among US adults, Arch. Intern. Med. 169 (2009)
USA: same evidence, different conclusions? Can we find common ground? Curr. 572–578.
Cardiol. Rep. 19 (2017) 49. [123] J.C. Defesche, P.J. Lansberg, M.A. Umans-Eckenhausen, J.J. Kastelein, Advanced
[100] U. Landmesser, M.J. Chapman, J.K. Stock, P. Amarenco, J.J.F. Belch, J. Boren, method for the identification of patients with inherited hypercholesterolemia,
A.L. Catapano, et al., 2017 Update of ESC/EAS Task Force on practical clinical Semin. Vasc. Med. 4 (2004) 59–65.

125

You might also like