Next Article in Journal
Legionella spp. Monitoring in the Water Supply Systems of Accommodation Facilities in Sardinia, Italy: A Two-Year Retrospective Analysis
Previous Article in Journal
Family Resilience and Dyadic Coping during the Outbreak of the COVID-19 Pandemic in Italy: Their Protective Role in Hedonic and Eudaimonic Well-Being
Previous Article in Special Issue
Factors Associated with Symptoms of Depression among People with Obesity: Analysis of a 3-Year-Peruvian National Survey
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lipoprotein(a) as a Risk Factor for Cardiovascular Diseases: Pathophysiology and Treatment Perspectives

1
Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy
2
SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy
3
Institute for Maternal and Child Health, I.R.C.C.S “Burlo Garofolo”, 34137 Trieste, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally.
Int. J. Environ. Res. Public Health 2023, 20(18), 6721; https://doi.org/10.3390/ijerph20186721
Submission received: 13 April 2023 / Revised: 31 July 2023 / Accepted: 9 August 2023 / Published: 6 September 2023

Abstract

:
Cardiovascular disease (CVD) is still a leading cause of morbidity and mortality, despite all the progress achieved as regards to both prevention and treatment. Having high levels of lipoprotein(a) [Lp(a)] is a risk factor for cardiovascular disease that operates independently. It can increase the risk of developing cardiovascular disease even when LDL cholesterol (LDL-C) levels are within the recommended range, which is referred to as residual cardiovascular risk. Lp(a) is an LDL-like particle present in human plasma, in which a large plasminogen-like glycoprotein, apolipoprotein(a) [Apo(a)], is covalently bound to Apo B100 via one disulfide bridge. Apo(a) contains one plasminogen-like kringle V structure, a variable number of plasminogen-like kringle IV structures (types 1–10), and one inactive protease region. There is a large inter-individual variation of plasma concentrations of Lp(a), mainly ascribable to genetic variants in the Lp(a) gene: in the general po-pulation, Lp(a) levels can range from <1 mg/dL to >1000 mg/dL. Concentrations also vary between different ethnicities. Lp(a) has been established as one of the risk factors that play an important role in the development of atherosclerotic plaque. Indeed, high concentrations of Lp(a) have been related to a greater risk of ischemic CVD, aortic valve stenosis, and heart failure. The threshold value has been set at 50 mg/dL, but the risk may increase already at levels above 30 mg/dL. Although there is a well-established and strong link between high Lp(a) levels and coronary as well as cerebrovascular disease, the evidence regarding incident peripheral arterial disease and carotid atherosclerosis is not as conclusive. Because lifestyle changes and standard lipid-lowering treatments, such as statins, niacin, and cholesteryl ester transfer protein inhibitors, are not highly effective in reducing Lp(a) levels, there is increased interest in developing new drugs that can address this issue. PCSK9 inhibitors seem to be capable of reducing Lp(a) levels by 25–30%. Mipomersen decreases Lp(a) levels by 25–40%, but its use is burdened with important side effects. At the current time, the most effective and tolerated treatment for patients with a high Lp(a) plasma level is apheresis, while antisense oligonucleotides, small interfering RNAs, and microRNAs, which reduce Lp(a) levels by targeting RNA molecules and regulating gene expression as well as protein production levels, are the most widely explored and promising perspectives. The aim of this review is to provide an update on the current state of the art with regard to Lp(a) pathophysiological mechanisms, focusing on the most effective strategies for lowering Lp(a), including new emerging alternative therapies. The purpose of this manuscript is to improve the management of hyperlipoproteinemia(a) in order to achieve better control of the residual cardiovascular risk, which remains unacceptably high.

1. Introduction

Cardiovascular disease (CVD) remains a significant global health challenge, despite considerable advancements in its prevention and treatment. One crucial risk factor associated with CVD is elevated levels of lipoprotein(a) [Lp(a)], which operates independently and contributes to what is known as residual cardiovascular risk. Remarkably, even when low-density lipoprotein cholesterol (LDL-C) levels fall within recommended ranges, high Lp(a) can still increase the risk of developing cardiovascular events. Lp(a) is a unique LDL-like particle found in human plasma, comprising a large plasminogen-like glycoprotein, called apolipoprotein(a) [Apo(a)], covalently bound to Apo B100 via a disulfide bridge. Apo(a) consists of plasminogen-like kringle V and variable kringle IV structures (up to 40 copies per allele), along with an inactive protease region. The plasma concentrations of Lp(a) exhibit significant inter-individual variability, primarily attributed to genetic variants in the Lp(a) gene, resulting in levels ranging from <1 mg/dL to >1000 mg/dL in the general population. Moreover, Lp(a) concentrations vary among different ethnicities.
Lp(a) has emerged as a crucial risk factor implicated in the development of atherosclerotic plaque. Higher Lp(a) concentrations have been associated with an increased risk of various cardiovascular conditions, including ischemic CVD, aortic valve stenosis, and heart failure. A threshold value of 50 mg/dL has been established for clinical significance, but even levels above 30 mg/dL may escalate the risk. While a robust and well-established link exists between elevated Lp(a) levels and coronary as well as cerebrovascular diseases, the evidence concerning incident peripheral arterial disease (PAD) and carotid atherosclerosis remains less conclusive. Standard lifestyle modifications and lipid-lowering therapies, such as statins, niacin, and cholesteryl ester transfer protein (CETP) inhibitors, have demonstrated limited efficacy in reducing Lp(a) levels. This limitation has spurred increased interest in developing novel drugs to address this issue.
Among potential therapeutic avenues, PCSK9 inhibitors have shown promise in reducing Lp(a) levels by 25–30%, offering a potential breakthrough. Another approach involves mipomersen, an antisense oligonucleotide (ASO) that can reduce Lp(a) levels by 25–40%, but its use is hampered by significant side effects. Currently, lipoprotein apheresis stands out as the most effective and well-tolerated treatment for individuals with high Lp(a) plasma levels. This extracorporeal method selectively removes lipoproteins containing Apo B100, resulting in an over 50% reduction in atherogenic lipoprotein. In addition to apheresis, emerging therapies involving antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), and microRNAs hold great promise in reducing Lp(a) levels by targeting RNA molecules and regulating gene expression as well as protein production levels.
The aim of this comprehensive review is to provide an updated insight into the pathophysiological mechanisms of Lp(a), with a particular focus on the most effective strategies for lowering Lp(a) levels, including the investigation of new and emerging alternative therapies. By addressing hyperlipoproteinemia(a) more effectively, this review aims to contribute to the better management of residual cardiovascular risk, which continues to present a significant challenge in clinical practice.

Search Strategy

The authors searched three bibliographic databases—PubMed, MEDLINE, and Embase—from the inceptions of the databases until Jul 2023, using a combination of terms related to Lp(a) prevalence, risk factors for cardiovascular diseases, pathophysiology, and treatment, without restrictions on language or publication date.

2. Hyperlipoproteinemia(a), the Hidden CV Risk Factor

Despite all of the progress achieved in terms of prevention and treatment, CVDs remain the primary cause of morbidity and mortality in the world population. Indeed, even if all of the best evidence-based strategies are applied (such as tailored pharmacological therapy plus lifestyle modifications, leading to the full achievement of goals for LDL-C reduction and blood pressure as well as glycemia normalization, according to the current guidelines), the observed residual cardiovascular (CV) risk remains high. Therefore, independent risk factors should be considered. Hyperlipoproteinemia(a) represents a widespread health problem in the global population: indeed, levels of Lp(a) >50 mg/dL have been found in 10–30% of the world population, with an estimated 1.43 billion people affected in the world, of which 148 million are in Europe [1] (Table 1).

2.1. Structure and Metabolism

Lp(a) is a low-density lipoprotein (LDL) variant where a large plasminogen-like glycoprotein, i.e., apolipoprotein(a), or Apo(a), is covalently bounded to Apo B100 via one disulfide bridge. Apo(a) contains one plasminogen-like kringle V structure, a variable number of plasminogen-like kringle IV structures (types 1–10), and one inactive protease region [2] (Figure 1). Lipoprotein (a) is synthesized in the liver [3]; its catabolism, however, is not completely understood [4]. Indeed, Lp(a) has a longer plasma half-life than LDL, suggesting a distinct metabolic pathway involved in its degradation. Because of its configuration and composition, Lp(a) is able to interact with different receptors, such as the LDL receptor protein megalin, very-low-density lipoprotein (VLDL) receptor, galactose-specific asialoglycoprotein receptor (ASGPR), plasminogen receptor, and macrophage receptors [5].

2.2. The Role of Genetic and “Non-Genetic” Factors in Lp(a) Metabolism

Lp(a) plasma concentration shows a wide inter-individual variation in the general population, ranging from <1 mg/dL to >1000 mg/dL [6]. Nonetheless, Lp(a) plasma concentration remains relatively constant over the course of life in men, while in women it tends to increase with age after menopause, with levels peaking during late peri- and post-menopause [7]. Lpa(a) concentrations also differ among ethnicities: it tends to be higher in individuals of African descent when compared to populations with European or Asian heritage [8,9]. Some authors have hypothesized that the differences among populations may be due to different Lp(a) gene variants [9]. Therefore, a higher Lp(a) plasmatic concentration could be considered a genetic feature largely controlled by genetic variants of the Lp(a) gene. The heritability of Lp(a) has been shown to be very high by twin and family studies, assessed as being from 70% to over 90% [10,11,12]. Indeed, differently from other lipoprotein concentrations, Lp(a) is minimally responsive to lifestyle or behavior changes, suggesting that Lp(a) levels are mostly genetically determined [13].
The gene responsible for producing Lp(a) is situated at positions 26 and 27 on the long arm of chromosome 6 (6q26-27). This gene is characterized by a high degree of polymorphism and consists of a variable exonic repeat. The gene encodes a protein domain referred to as “kringle” (k) [14], which evolved from the plasminogen gene (PLG). The plasminogen gene contains one protease domain and five different types of kringle domains. In about 40 million years the PLG gene has been remodeled into the Lp(a) gene; in the process it lost KI, KII, and KIII, kept only one copy of KV, and its protease domain lost activity [15,16].
The most important predictors of Lp(a) levels are an array of LPA genetic polymorphisms that mainly account for different isoforms of the protein, which contribute to the variation in Lp(a) concentrations in a range of 40% to 70% [17]. In fact, polymorphisms of the Lp(a) gene result in lipoprotein isoforms of different sizes, and studies have revealed that the size of Apo(a) changes in relation to kringle IV type 2 (KIV-2) copy number variations, with a clear inverse correlation between the number of KIV-2 repeats and Lp(a) concentration [18,19,20]. According to the Copenhagen General Population Studies, up to 27% of the Lp(a) concentration variation depends on the population studied, but also on the Lp(a) gene KIV-2 repeats number. Beyond KIV-2 repeats, a high Lp(a) plasma level is also associated with the pentanucleotide repeat polymorphism (PNRP) of the promoter 5′ control region of the Apo(a) gene, as well as several single-nucleotide polymorphisms (SNPs) [21,22]. To date, only a few SNPs (including rs3798220 and rs10455872 SNPs) have shown a significant association with increased plasma Lp(a) levels and CVD [23]. Nonetheless, the fact that multiple SNPs with different, mostly small, effect sizes, associated with both high and low Lp(a) levels, may contribute collectively to the variance of the trait should be considered [24]. Obviously, the contribution of other genes requires further investigations.
Although plasma Lp(a) concentration levels are strongly genetically determined, some evidence suggests that nongenetic factors, including renal function, gender, hormones, and inflammation, may also affect Lp(a) [25]. The kidney has been shown to play a role in Lp(a) catabolism, and Lp(a) levels are associated with kidney disease [26]. Several studies have also evaluated how Lp(a) concentration is affected by hemodialysis in patients with end-stage renal disease (ESRD). This clinical condition results in Lp(a) levels 5 to 10 times higher than those in patients with early-stage renal disease. Furthermore, Lp(a) concentration returns to normal levels after a kidney transplantation, probably because of improved clearance [27]. With regard to the hormonal regulation of Lp(a) levels, it has been observed that they are usually higher in postmenopausal women, suggesting an estrogen-related inhibiting effect [28,29]. Lp(a) has been studied in both normal pregnancies and in those complicated by various pathological conditions, such as pre-eclampsia, showing that there is a two-fold increase in Lp(a) during normal pregnancy [30]. Androgens, such as endogenous testosterone levels, may affect Lp(a); men with low testosterone levels have higher Lp(a) levels, and they seem to have a greater risk of heart disease [31,32]. Furthermore, growth hormone (GH) has been linked to a strong Lp(a)-stimulating effect, both in patients with acromegaly and in GH therapy [33,34]. Thyroid hormones also seem to have a modulatory effect on Lp(a), but the mechanism is still unclear. Acute inflammatory states, including sepsis, inflammatory bowel disease, gallbladder fistula, and acute myocardial infarction, have been shown to increase Lp(a) concentration [35]. Furthermore, a direct correlation has been found between the plasma concentration of Lp(a) and some inflammatory proteins, such as IL-6, CRP, and α 1 antitrypsin [36]. The relationship between Lp(a) and diabetes remains unclear: prospective findings demonstrate a strong inverse association between Lp(a) levels and the risk of developing type 2 diabetes [37,38]. On the other hand, data collected by several studies show that ethanol and tobacco consumption, regardless of different Apo(a) isoforms, reduce, on a dose-dependent basis, Lp(a) concentration by up to 60% and 20%, respectively [39,40,41,42]; the mechanism behind this phenomenon is, though, still unclear.

3. Pathophysiology of Lp(a)

As of now, a clear physiological function of Lp(a) has not been clearly recognized, but the homology between Apo(a) and PLG suggests that Lp(a) may represent a link between cholesterol transport and the fibrinolytic system, with a role in the modulation of blood clotting and the fibrinolytic process. Indeed, Lp(a) could enhance wound healing by carrying cholesterol to an injury site and play a role in hemostasis via the inhibition of fibrinolysis [43].
When there is an injury or wound in the body, Lp(a) may play a role in the healing process by transporting cholesterol to the site of the injury. Cholesterol is essential for cell membrane formation and repair, and it also serves as a building block for various molecules involved in tissue repair and regeneration. Therefore, the presence of Lp(a) at the wound site may aid in providing the necessary cholesterol for the healing process [44].
Lp(a) has been suggested to play a role in hemostasis via inhibiting fibrinolysis. In other words, Lp(a) may help stabilize blood clots by preventing their premature breakdown, which could be beneficial in situations where maintaining a stable clot is necessary for effective wound healing and preventing excessive bleeding [45].
It is important to note that, while these potential roles of Lp(a) in wound healing and hemostasis have been proposed, research on this topic is ongoing, and the exact mechanisms and implications are not fully understood.
An elevated Lp(a) level is a strong, causal, and independent risk factor for CVD through multiple pathogenetic mechanisms: proatherogenic, prothrombotic, and proinflammatory [46].
Experimental evidence from in vitro studies and animal models shows Lp(a)’s promotion of atherosclerotic plaque formation through various mechanisms, such as smooth muscle cell proliferation, foam cells formation, and increased expression of IL-8, a key mediator of plaque formation, from inflammatory cells [47,48]. Lp(a) can interact with the major components of the extracellular matrix (fibrin, fibronectin, proteoglycans, tretranectin, and beta2-glycoprotein) [49]. The binding of Lp(a) to fibrin has been proposed as the mechanism that allows for the delivering of cholesterol to sites of injury, where it is involved in the repairing of the vascular wall. It has also been observed that Lp(a) is able to bind oxidized phospholipids (OxPls) in plasma by forming covalent bonds. Levels of Lp(a) and OxPls in plasma predicted the risk of the incidence of CVD [50].
In order to cause CVD, Lp(a) needs to be able to enter into the intima of arteries as well as aortic valve leaflets and accumulate. Studies on human and rabbit kinetics have revealed that Lp(a) can enter normal and atherosclerotic arteries at a comparable rate to LDL [51]; however, Lp(a) has a tendency to accumulate throughout the intima, whereas the accumulation of LDL cholesterol and other lipoproteins containing apoB is typically restricted to atherosclerotic lesions. It has been observed that Lp(a) accumulates two- to three-fold greater than LDL in the arterial wall at sites of injury (balloon-injured rabbit arteries). Lp(a) and Apo(a) have also been found in end-stage aortic valve stenosis and in lesioned intima of the coronary artery bypass [52,53,54]. Lp(a) appears to have a greater capacity than LDL to bind fibrin and glycosaminoglycans, two proteins exposed at sites of injury [55,56]. In conclusion, it is possible that the intimal accumulation of Lp(a) is driven by a different mechanism than that involving LDL and other apoB-containing lipoproteins.

4. Laboratory Assessment

Lp(a) measurements are mainly obtained by immunoassays; however, some major issues can affect the accuracy of Lp(a) quantification and consequently the clinical interpretation of the data. Size variability of Apo(a) isoforms is one of the most critical problems, since it may lead to the over- or under-estimation of Lp(a) levels [57]. In fact, there are two types of immunoassays with which to determine Lp(a) concentration: those that are “isoform-dependent” and those that are “isoform-independent”. Lp(a) concentrations have historically been described as total mass concentration (mg/dL). In the 1970s, Albers et al. purified Lp(a) from plasma; they individually measured protein, lipid, and carbohydrate components, and they used the sum of the components as an assay calibrator, with a value in mg/dL. All of the subsequent immunoassays were then calibrated in mg/dL, although they only measured the Apo(a) component and not the total Lp(a) mass [58]. Indeed, the “isoform-dependent” method uses monoclonal or polyclonal antibodies in order to evaluate the whole lipoprotein mass (mg/dL), and it is highly related to the KIV-2 copies number. At present it is well known that, with this method, Lp(a) concentrations are overestimated in patient samples containing large isoforms and underestimated in samples containing small isoforms; however, recent methods that use five independent calibrators with a large range of Lp(a) levels and distribution of apo(a) isoforms are able to reduce the confounding impact of Apo(a) size if the values of the assay calibrators are well validated [59]. Although this method shows higher precision, it does not completely eliminate the Apo(a) isoform bias. On the other hand, the isoform-independent method (nmol/L) uses monoclonal antibodies able to bind KIV-9, the unique non-variable Apo(a) domain, reflecting the number of Lp(a) particles.
The ELISA method was approved to be the gold standard with which to measure Lp(a) concentration by the World Health Organization (WHO), and it is calibrated in nmol/l, as has been standardized by the International Federation of Clinical Chemistry and Laboratory Medicine (IFCC) [60,61,62].
The two immunoassay approaches report Lp(a) levels using two different units: mg/dL and nmol/L. Indeed, Lp(a) concentrations should not be converted from nmol/L into mg/dL, or vice versa, as all conversion factors are isoform-dependent [61,63]. Previous studies have tried to convert mg/dL or mg/L into nmol/L by using a correction factor equal to 2.4, but this approach is not recommended since it does not consider the wide heterogeneity of Apo(a) type. The existence of two different units for expressing Lp(a) levels can be confusing for both clinicians and patients.
Nowadays, none of the current commercially available assays for Lp(a) measurement are completely inherently isoform-insensitive [62,64]. Innovation in the development of isoform-insensitive Lp(a) assays is currently an unmet need. The development of non-antibody-based methods may provide the right approach with which to avoid the bias from the size variability of Apo(a). There are currently three methods with which to assess the KIV repeats number and Apo(a) isoform size at the DNA level: pulsed field gel electrophoresis (PFGE) and fiber-fluorescence in situ hybridization (FISH), which allow the evaluation of KIV-2 repeats number in separate alleles, while the quantitative polymerase chain reaction (qPCR) evaluates the sum of KIV-2 copies in examined genomes [65].

5. Lp(a) in Vascular Diseases

5.1. Cardiovascular Risk

Coronary heart disease (CHD) is a major CVD, and it is one of the leading causes of death in both developed and developing countries [66,67]. CHD is an atherosclerotic disease whose clinical manifestations include stable angina, unstable angina, myocardial infarction (MI), or sudden cardiac death [68]. Modifiable known risk factors for CAD include diabetes mellitus, hypertension, smoking, hyperlipidemia, obesity, chronic kidney disease, hyperhomocysteinemia, and psychosocial stress [69].
High Lp(a) concentrations have been consistently linked to an increased risk of ischemic CVD, aortic valve stenosis, and heart failure. Both the American Heart Association/American College of Cardiology (AHA/ACC) and the European Society of Cardiology (ESC) guidelines have suggested a threshold of 50 mg/dL of Lp(a) to mitigate an increased risk of CVD; however, some experts believe that the risk of CVD may already increase when Lp(a) plasma levels exceed 30 mg/dL [70]. The 2019 ESC/EAS guidelines recommend that individuals undergo at least one Lp(a) plasma level test in their lifetime. It is worth noting that individuals with congenitally high Lp(a) levels (e.g., >180 mg/dL) may have an ASCVD (atherosclerotic CVD) risk equivalent to those with a family history of premature cardiovascular disease or those with heterozygous familial hypercholesterolemia [71]. Although there is no clear biochemical mechanism to explain how higher Lp(a) levels increase the risk of CHD, researchers have proposed several potential mechanisms, including the involvement of LDL-C [72], inhibition of the conversion of plasminogen into plasmin [73], and the ability to transport proinflammatory oxidized phospholipids as well as disseminate them as carriers [74].
To contribute to CVD, Lp(a) must have the ability to accumulate in the intima of arteries. This accumulation in locations of vascular damage is thought to be one of the primary mechanisms through which elevated Lp(a) leads to CVD [73,74]. Additionally, research suggests that Lp(a) may also contribute to foam cell development through macrophage uptake, a mechanism that is also associated with LDL and triglyceride-rich lipoproteins, as well as the development of atherosclerosis [75,76,77].
A meta-analysis conducted by Wendy Craig and colleagues in 1998 found that Lp(a) concentrations were higher in individuals who developed ischemic heart disease compared to those who did not [78]. This conclusion was based on data from population-based prospective cohort studies and nested case–control studies [79,80,81,82,83].
John Danesh and colleagues confirmed a clear association between Lp(a) and CHD by updating this meta-analysis with an added follow-up of 10 years inclusive of 5436 additional deaths from CHD [84]. The INTERHEART study and the Copenhagen City Heart Study examined the contribution of Lp(a) concentration to MI risk in the general population and assessed that concentrations of Lp(a) >50 mg/dL were associated with an increased risk of MI [85].
Even large Mendelian randomization studies, which are free from confounding bias and reverse causation, strongly support the notion that elevated Lp(a) represents an independent, genetic causal factor of CVD [86,87]. Although the genomic determinants of Lp(a) and their impact on the risk of coronary disease are not well understood, the argument for direct, genetic causality in CVD is much more compelling for Lp(a) than for the majority of other cardiovascular risk factors [88]. A case–control study recruited 3145 case subjects with coronary artery disease and 3352 control subjects from four European countries; in this study, two common Lp(a) variants (rs10455872 and rs3798220) were shown to be strongly associated with both an increased level of Lp(a) lipoprotein [explaining together 36% of the total variation in Lp(a) concentration] and an increased risk of coronary disease [89].

5.2. Aortic Valve Stenosis

Calcific aortic valve disease (CAVD) is a progressive condition that results from the severe calcification of the aortic valve (i.e., aortic valve stenosis, AS), which has three leaflets and regulates blood flow from the heart. This calcification hinders the valve’s movement and restricts the outflow of blood from the ventricle. CAVD affects about 2% of people over 65 years old, and symptoms usually do not appear until the disease is advanced. Epidemiological research has revealed that high levels of Lp(a) in the blood increase the risk of AS. A study from the 1990s found that genetic variations in the Lp(a) locus were linked to aortic valve calcification, and several studies have shown that an rs10455872 Lp(a) SNP doubles the risk of aortic valve calcification in various ethnic groups [90]. Furthermore, the Lp(a) genotype is associated with an increased risk of aortic valve replacement surgery and degree of aortic valve calcification [91,92,93]. These findings are reported in Figure 2.
Imaging studies (based on both computed tomography calcium scoring and echocardiography) observed increased valve calcification and faster rates of disease progression in elderly patients with high Lp(a) levels [94]. Recently, this association was not confirmed by Kaiser and colleagues, who assessed that Lp(a) might play a role in the initial phase of AS but found no association between Lp(a) levels and the progression of disease; if this evidence is confirmed, the effectiveness Lp(a) of Lp(a)-lowering in affecting the clinical outcomes of AS may be limited to pre-calcific stages of aortic valve disease [95].
The epidemiological and genetic evidence that high Lp(a) levels are linked with the development of AS is quite strong. Lp(a) is an important carrier of oxidized phospholipids (OxPL) that may play a key role in the process [96]. Moreover, Lp(a) may favor the onset and development of CAVD by causing aortic valve endothelial dysfunction, accumulating in the valve, and delivering its OxPL content along with autotaxin (ATX) [97]. This mechanism promotes not only inflammation but also the osteogenic transformation of valvular interstitial cells causing calcium deposition [98]. Valve calcification is characterized by an inflammatory response, which involves the secretion of lipoprotein-associated phospholipase A2 (Lp-PLA2) by macrophages. Lp-PLA2 utilizes OxPLs as a substrate and produces lysophosphatidylcholine (LPC), which is then converted by ATX into lysophosphatidic acid [Lp(a)]. Lp(a) binds to and activates the Lp(a) receptor [Lp(a)R], triggering the activation of NF-κB, leading to increased inflammation and secretion of interleukin-6 (IL-6). This process results in osteogenic differentiation, valve leaflet calcification, and eventually AS [99,100]. Additionally, Apo(a), B, and E have been found in aortic valve lesions [101]. Despite Lp(a) consistently being higher in black individuals than in Caucasians, the prevalence of AS is higher in Caucasians than in Blacks, Hispanics, and Asians in the US [102].
While elevated Lp(a) is acknowledged as a major predictive factor for clinical AS in patients with heterozygous familial hypercholesterolemia (FH) [96], its influence on aortic stenosis in the general population remains to be fully determined.

5.3. Peripheral Arterial Disease

Peripheral arterial disease (PAD) is an atherosclerotic disorder characterized by a progressive narrowing, eventually to the point of occlusion, of large- and medium-sized arteries of the extremities, especially the lower ones [103,104]. PAD is largely diffuse worldwide, and it is associated with increased CV morbidity as well as mortality. As in other atherosclerotic diseases, many risk factors have been established as playing an important role in plaque formation and progression. In addition to traditional risk factors, such as hypertension, hypercholesterolemia, and diabetes mellitus, Lp(a) has also been investigated as a risk factor [105].
Despite the strong and well-established association between elevated Lp(a) levels and coronary as well as cerebrovascular disease, data regarding incident PAD and carotid atherosclerosis are less robust: most studies show a direct correlation between Lp(a) levels and PAD, while in others this association is not definitely confirmed [106].
Genetic, pathophysiologic, and epidemiologic studies, such as the Copenhagen General Population Study, support the notion of Lp(a) as a potential causative factor in femoral stenosis [107]. In the most recent GWAS study, Klarin et al. identified Lp(a) variants strongly associated with PAD [108]. A different study, by Lashkolnig et al., showed a significant association between Lp(a) concentrations low-molecular-weight (LMW) Apo(a) phenotypes, and rs10455872 with PAD, both symptomatic and asymptomatic [109]. Both the MESA study and the InCHIANTI study demonstrated a notable correlation between elevated Lp(a) levels and PAD in the lower extremities [110,111]. The EPIC-Norfolk prospective study also showed an increased risk in terms of a 1.37 HR for PAD per a 2.7-fold increase in Lp(a) levels, an association that proved to be independent of LDL cholesterol levels [112].
Despite the ESC and ACC/AHA guidelines stating that the risk of CVD is significant when Lp(a) >50 mg/dL [113,114], in several studies the cut-off level of Lp(a) for a potentially increased risk of PAD was set to 30 mg/dL. A small study reported that Lp(a) levels greater than 30 mg/dL are associated with a 3.9-fold increase in the risk of premature PAD [115]. Another cross-sectional study involving 557 patients with type 2 diabetes showed similar results, with Lp(a) levels exceeding 30 mg/dL carrying a threefold higher risk of PAD. This study also found an inverse correlation between Lp(a) levels and the ankle–brachial index (ABI) [116]. In a prospective case–control study, Lp(a) levels above 24 mg/dL were linked to a two-fold increased risk of PAD. Furthermore, higher levels of Lp(a) were associated with more severe forms of PAD [117]. A higher risk for ischemic stroke, MI, or limb amputation has been observed in symptomatic PAD patients with Lp(a) >30 mg/dL, compared to similarly symptomatic patients with lower levels [118]. A recent study showed that patients with PAD and Lp(a) >30 mg/dL had a greater need for any PAD operation [119]. Yanaka et al. also showed that a higher Lp(a) level was an independent predictor for the loss of primary patency after endovascular therapy (EVT) [120].
A recent systematic review analyzed 15 studies involving 493,650 patients, and the majority of these studies supported a significant association between high Lp(a) levels and the risk of PAD, according to the authors. High Lp(a) levels were also found to be linked to an increased risk of claudication, PAD progression, restenosis, hospitalization, and death [121] (Figure 2).

5.4. Carotid Atherosclerosis

In 2020, globally, around 28% of individuals aged 30–79 years in the general population were found to have an abnormal carotid intima-media thickness of 1.0 mm and above, which corresponds to slightly over one billion people. Moreover, approximately 21% of people aged 30–79 years had carotid plaque, while 1.5% had carotid stenosis, indicating roughly 816 million individuals with carotid plaque and 58 million with carotid stenosis [122]. Lp(a) has been established as one of the risk factors that play an important role in the development of atherosclerotic plaque [123]. Increased Lp(a) levels seem to be associated with carotid artery stenosis: Klein et al. reported that Lp(a) was an independent predictor of stenosis and occlusion, but not of carotid plaque area [124]. Different studies have shown that elevated Lp(a) levels were independent predictors of increased carotid atherosclerotic burden, and other epidemiological as well as genetic studies have shown a continuous and independent association between Lp(a) and cerebrovascular disease [125]. Lp(a) seems, indeed, to be significantly correlated with stenosis and occlusion, which are frequently the consequences of plaque rupture and thrombosis. This may lead to the hypothesis that the role of Lp(a) in atherogenesis may be largely based on its effect on coagulation and thrombosis.
In patients who have suffered an ischemic stroke, elevated Lp(a) levels are associated with the presence of carotid atherosclerosis [126]. Furthermore, in the AIM-HIGH study, Lp(a) was associated with high-risk plaque features, such as the presence of a mural thrombus, intraplaque hemorrhage, or surface defects [127]. Elevated Lp(a) levels may independently predict the risk of carotid atherosclerosis progression, despite a strict LDL-C control [128].
Contrary to the results mentioned previously, Ooi and colleagues’ research revealed that while plasma Lp(a) concentration was independently linked to the extent and severity of coronary artery disease (CAD) upon angiography, it did not display a significant association with carotid artery plaque. This suggests that the impact of plasma Lp(a) levels may differ between the two vascular conditions [129].
Gender-associated differences in Lp(a) distribution, carotid plaque composition, and the frequency of vulnerable plaques have been shown [130]: while men tend to have more vulnerable plaques than women, a recent study has revealed that in women elevated plasma Lp(a) levels were associated with a higher prevalence of intraplaque hemorrhage (IPH), while in men they were associated with a higher degree of stenosis [131].

5.5. Stroke

The association between Lp(a) and stroke was first reported in the 1980s [132]; since then, a large number of studies have investigated the role of Lp(a) as a risk factor of cerebrovascular disease. In a Danish study that analyzed a large population of about 50,000 individuals [133], high Lp(a) levels correlated with an increased incidence of ischemic stroke, although the specific mechanisms are not fully understood. There is no evidence of different Lp(a) accumulation in carotid artery plaques compared to coronary ones [134], although events triggered by intracerebral vessels or hemorrhagic stroke were associated with lower levels of Lp(a) [126]. The different biology of larger lesions (inflammatory cells’ prevalence, fibrous cap erosion, vessel wall frailty, and superimposed thrombosis) compared to smaller ones and the occurrence of non-atherosclerotic lesions might account for such a discrepancy. Several hypotheses have been advanced to explain the pathophysiology: high levels of Lp(a) could increase the deposition of cholesterol in vessels, while the Apo(a) component, via interfering with fibrinolysis and because of its known proinflammatory properties, could predispose them to plaque. However, the role of Lp(a) in the genesis of other types of stroke is not yet fully clarified: a meta-analysis of Kumar et al. [135], which analyzed the relationship between Lp(a) levels and various subtypes of stroke, shows an association between Lp(a) and stroke secondary to large-vessel atherosclerosis rather than small-vessel and cardioembolic stroke.
There are conflicting results concerning the association between Lp(a) levels and thromboembolic risk in atrial fibrillation (AF) [136]. Igarashi demonstrated that Lp(a) is an independent risk factor for left-atrium thrombosis in patients with chronic AF [137]; more recently, a correlation between elevated Lp(a) levels and thromboembolism in patients with AF and a CHADVASC of less than 2 has been found [138]. Conversely, other works do not find a correlation between Lp(a) levels and cardioembolic stroke [139], suggesting that the different phenotypes of Lp(a) may be involved [140]. Reports are also conflicting with regard to lacunar stroke. Kario et al. demonstrated an association between high levels of Lp(a) and multiple lacunar strokes [141], while other studies [142] have failed to show this correlation.

5.6. Heart Failure

Heart failure (HF) is a global public health problem and a major burden, particularly for economies and health systems of countries with aging populations. As mentioned earlier, Lp(a) is an independent risk factor for CAD and calcific aortic valve stenosis, both of which are underlying causes of heart failure (HF) (see Figure 2). In 2016, a study reported an association between Lp(a) and corresponding Lp(a) risk genotypes with HF. Two cohort studies in the combined Copenhagen General population have investigated the correlation between Lp(a) and HF-related outcomes, revealing that the risk of incident HF increases with rising levels of Lp(a) [143]. These findings were subsequently supported by a paper from the ARIC investigators [144]. In the Copenhagen studies, while the increased risk for HF appeared to be mostly driven by CAD and AS, the association between Lp(a) and HF remained significant even after the exclusion of patients with previous MI or AS [145]. This observation implies that Lp(a) might have a part to play in HF via means of alternative pathophysiological mechanisms besides the two mentioned earlier. These may include arterial stiffness related to atherosclerosis or vascular noncompliance, which could result in heightened cardiac afterload.
The association between HF and Lp(a) levels seems to vary according to the ethnic group studied. In a multiethnic cohort, Lp(a) was found to be a significant risk factor for incident HF in Caucasian individuals alone, as in the MESA study [146], and in the Chinese population alone [147]. Recently, a large Icelandic case–control study found an association between Lp(a) genetic variants with risk of HF [148].
Figure 2 displays adjusted hazard ratios for selected outcomes, comparing participants with high Lp(a) levels to those with lower concentrations, in relation to their risk for various health conditions and mortality. The study, conducted as part of the Copenhagen studies, found an association between high Lp(a) levels and an increased risk of CVD as well as mortality. The study calculated hazard ratios by comparing individuals in the upper percentiles of Lp(a) distribution, specifically the 90–95th percentile (yellow) and >95th percentile (red) for aortic stenosis, 91–99th percentile (yellow) and >99th percentile (red) for MI and HF, and >95th percentile for ischemic stroke, cardiovascular mortality, and all-cause mortality, with those who had lower Lp(a) concentrations, which were below the 22nd percentile for calcific AS, below the 34th percentile for MI, HF, and PAD, and below the 50th percentile for ischemic stroke, cardiovascular mortality, and all-cause mortality. The figure presents an adjusted odds ratio for participants in the >66th percentile compared to those in the <33rd percentile for PAD, which was defined as an ankle–brachial index of ≤0.9. It should be noted that the figure was adapted from a publication by Benoit J. Arsenault and Pia R. Kamstrup in the journal Atherosclerosis in 2022 [96].

6. Pharmacological Treatment

How is elevated Lp(a) treated? The low effectiveness of lifestyle modifications and traditional lipid-lowering therapy, such as statins, niacin, or CETP inhibitors, has aroused greater interest in searching for new drugs that can reduce plasma Lp(a) levels [149]. Statins, which are commonly used to lower cholesterol, do not affect Lp(a) levels and may even increase them slightly. For example, data from JUPITER (Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin) showed an increase in Lp(a) plasma levels of 10–20% in patients treated with rosuvastatin [150]. In a recent study of 3,896 patients treated with various statins (including atorvastatin, pravastatin, rosuvastatin, pitavastatin, and simvastatin/ezetimibe), the mean levels of Lp(a) increased by 11% and OxPL-apoB increased by 24% [151]. Another meta-analysis, involving 5256 patients, found that statins significantly increased plasma Lp(a) levels [152]; however, the underlying mechanisms of this increase are not yet well understood, and further studies are required. Furthermore, ezetimibe treatment, both alone and in combination with statins and lomitapide, did not reduce Lp(a) levels, while the effects of fibrates on Lp(a) levels remain uncertain (Table 2) [153,154].
Over the years, niacin, an essential nutrient involved in the synthesis and metabolism of carbohydrates, proteins, and lipids, has been recognized as an atheroprotective agent because of its capacity to lower the plasma levels of cholesterol, triglycerides, VLDL, and LDLc, as well as being able to raise that of high-density lipoproteins [155]. Niacin has also been reported to reduce the plasma levels of Lp(a) by 38–40% [156]. However, the abandonment of this therapeutic strategy was due to the appearance of side effects, particularly hot flashes, abdominal pain, hepatotoxicity, and the failure to achieve primary cardiovascular event reduction endpoints in two large randomized trials, one with prolonged-release niacin (AIM HIGH) [157] and the other with niacin/laropiprant (HPS2-THRIVE) (Table 2) [158].
Drugs that inhibit CETP — a glycoprotein synthetized mostly by the liver that plays a prominent role in the bidirectional transfer of cholesterol esters and triglycerides (TRG) between lipoproteins [159] — are able to increase HDL-C and also decrease serum LDL-C levels [160]. CEPT inhibitors (CEPTis) are also effective in reducing Lp(a) levels [161]: in particular, torcetrapib therapy decreased Lp(a) by 11%, evacetrapib by up to −40%, and evacetrapib combined with statins by 31%, while dalcetrapib had lower effects on lipids than torcetrapib, evacetrapib, or anacetrapib, and decreased Lp(a) by 5% (Table 2) [162].
PCSK9 inhibitors are human monoclonal antibodies, produced by genetic engineering techniques, that have recently been introduced into clinical practice. These drugs bind and inactivate a particular circulating enzyme that plays a central role in modulating the expression of hepatic receptors for low-density lipoproteins (LDL-R): the proprotein convertase subtilisin/kexin type 9 (PCSK9). The blocking of the PCSK9 protein results in a rapid slowdown of LDL receptors’ turnover with an increase in their number, which leads to a marked reduction in plasma LDL concentration by 50–70% [163]. In addition to the LDL cholesterol-lowering effect, PCSK9 inhibitors are also capable of reducing Lp(a) levels by 25–30% [164]. In particular, a recent study evaluated the effects of alirocumab on serum Lp(a) levels via the use of a pool of data from the Odissey phase 3 studies [165]; these data showed a significant reduction in Lp(a) levels regardless of the initial dose in patients treated with alirocumab and the concomitant use of statins. At 24 weeks of treatment, a reduction of 23% to 27% was observed in those patients who started 75 mg of alirocumab, and one of 29% in patients who received 150 mg of alirocumab. The reduction in Lp(a) levels is independent of race, sex, the presence of familial hypercholesterolemia, basal Lp(a) and LDL-C concentrations, and the use of statins. From a meta-analysis obtained from 10 clinical trials on 3,278 patients, a significant reduction in Lp(a) levels of 24.7% was observed in subjects who received 140 mg of evolocumab every 2 weeks, and 21.7% for subjects treated with a monthly dose of 420 mg [166]; however, it should be noted that both the Fourier and Odissey studies were not specifically designed to enroll and treat patients with high Lp(a) levels [167,168]. Furthermore, the use of these drugs is not currently approved for the specific treatment of isolated hyperlipoproteinemia(a). PCSK9 is also the target of inclisiran, a small interfering RNA (siRNA)-based drug that inhibits the hepatic synthesis of this protein. The ORION 10 trial initially measured a median Lp(a) value of 57 nmol/L, with a reduction rate of 25.6% after 510 days of treatment compared to the placebo group [169]. Similarly, in the ORION 11 study the baseline median Lp(a) level was 42 nmol/L, and the relative reduction was 18.6%. In the ORION 9 investigation, although the initial median Lp(a) concentration was 57 nmol/L, the reduction rate was lower, at −17.2% (Table 2) [170].
Mipomersen, a second-generation antisense oligonucleotide (ASO), was approved by the FDA for use in addition to statin therapy to treat homozygous FH [171]. Its mechanism of action involves inhibiting Apo B synthesis without affecting Apo(a). While it can reduce Lp(a) levels by 25–40%, its therapeutic use is limited due to the serious side effects that it can cause, such as site of injection reactions, hepatic steatosis, and hypertransaminasemia (Table 2) [172].
Table 2. The table summarizes different treatment options for elevated Lp(a) levels and their effects on reducing Lp(a) concentrations.
Table 2. The table summarizes different treatment options for elevated Lp(a) levels and their effects on reducing Lp(a) concentrations.
TreatmentMechanism of ActionEffect on Lp(a) LevelsNotable Findings and Remarks
Statins [150,151,152]Lowers cholesterol levelsNo significant reduction; may increase slightlyStatins do not significantly affect Lp(a) levels and may even lead to a slight increase in some cases. Further research is needed to understand the underlying mechanisms of this increase.
Niacin [156,157,158]Atheroprotective agent, lowers lipidsReduction in Lp(a) levels by 38–40%Niacin has been effective in reducing Lp(a) levels, but its use is limited due to side effects, such as hot flashes, abdominal pain, hepatotoxicity, and a failure to achieve primary cardiovascular event reduction in some trials.
CETP inhibitors [161,162]Reduces CETP activity, increases HDLReduction in Lp(a) levels; varying effectsCETP inhibitors have shown varying effects on Lp(a) levels, with some drugs reducing levels by up to 40%. The effects of fibrates on Lp(a) levels remain uncertain.
PCSK9 inhibitors [164,165,166,167,168]Blocks PCSK9 enzyme, increases LDL receptor expressionReduction in Lp(a) levels by 25–30%PCSK9 inhibitors have been found to be effective in reducing Lp(a) levels by 25–30%, along with lowering LDL-C. Alirocumab has shown promising results in reducing Lp(a) levels independently of factors such as race, sex, FH, and baseline Lp(a) as well as LDL-C concentrations.
Inclisiran [169,170]Inhibits the hepatic synthesis of PCSK9Reduction in Lp(a) levelsInclisiran has shown reductions in Lp(a) levels in clinical trials. The ORION studies demonstrated significant reductions in Lp(a) levels over the course of treatment.
Mipomersen [172]Inhibits apo-B synthesis without affecting apo(a)Reduction in Lp(a) levels by 25–40%Mipomersen can reduce Lp(a) levels but has limited therapeutic use due to serious side effects.
Apheresis [173]Extracorporeal removal of lipoproteinsLowering of LDLc and Lp(a) concentrations by 60–70%Apheresis has been found to be the most efficient and well-tolerated therapy for individuals with Lp(a) hyperlipoproteinemia. It achieves substantial reductions in LDL-C and Lp(a) levels, leading to significant improvements in cardiovascular outcomes, with a 54–90% reduction in cardiovascular events. Apheresis is recommended for patients with progressive coronary disease and Lp(a) levels greater than 60 mg/dL despite maximal lipid-lowering therapy. It has been approved for elevated Lp(a) associated with progressive CVD in Germany. Studies have provided evidence of reduction in cardiovascular risk and long-term efficacy in stabilizing CAD.
Summary of various treatment options for elevated Lp(a) levels and their effects on Lp(a) concentrations. The table provides an overview of different therapeutic approaches, including statins, niacin, CETP inhibitors, PCSK9 inhibitors, inclisiran, mipomersen, and apheresis, along with their respective mechanisms of action and their impact on Lp(a) levels. Notably, statins are found to have limited effectiveness in reducing Lp(a) levels, while niacin shows promise but is hindered by side effects. CETP inhibitors exhibit variable effects, and PCSK9 inhibitors demonstrate significant reductions in Lp(a) levels. Inclisiran, an siRNA-based drug, also lowers Lp(a) concentrations. Mipomersen, an antisense oligonucleotide, reduces Lp(a) levels but faces limitations due to side effects. Apheresis, an extracorporeal method for selective lipoprotein removal, proves to be the most efficient and well-tolerated therapy, achieving substantial reductions in Lp(a) levels, along with LDLc, leading to significant improvements in cardiovascular outcomes.

6.1. Lipoprotein Apheresis

Apheresis has been found to be the most efficient and well-tolerated therapy for individuals with Lp(a) hyperlipoproteinemia (Table 2). The data available on lipoprotein apheresis are very impressive, showing a lowering of LDL-C and Lp(a) concentrations by 60–70%, improving the cardiovascular outcomes of these patients with a 54–90% reduction in cardiovascular events [173]. Apheresis is an extracorporeal method of selective removal, from plasma or whole blood, of lipoproteins containing Apo B100, with a reduction of over 50% of atherogenic lipoprotein [174]. The guidelines of the American Society for Apheresis (ASFA) since 2013 have included Lp(a) hyperlipoproteinemia among the indications for lipoprotein apheresis [175]. The HEART UK Lipoprotein apheresis guidelines recommend that apheresis should be considered for those patients with progressive coronary disease and Lp(a) greater than 60 mg/dL whose LDL-C remains 125 mg/dL despite maximal lipid-lowering therapy [176]. In Germany, lipoprotein apheresis has been approved for elevated Lp(a) associated with progressive CVD; since then, the German Lipoprotein Apheresis Registry (GLAR) provides statistical evidence for the assessment of extracorporeal procedures enacted to lower both LDL-C and Lp(a). In a prospective investigation based on GLAR data over a 5-year period, patients with an LDL-C level lower than 100 mg/dL but Lp(a) level higher than 60 mg/dL showed a significant reduction in major coronary (83%) and noncoronary events (63%) [177]. At present, the FDA has authorized the use of lipoprotein apheresis solely for individuals who have documented CVD progression and increased Lp(a) levels exceeding 60 mg/dL [178]. Studies have provided proof of a reduction in cardiovascular risk in patients undergoing regular apheresis; indeed, lipid apheresis achieves near-normal Lp(a) levels and prevents a MACE (major adverse cardiovascular event) [179]. The Prospective Pro(a)LiFe study also supported the notion that the prevention of cardiovascular events is both a rapid and lasting effect of apheresis in patients with progressive CVD associated with Lp(a) hyperlipoproteinemia [180]. Another multicenter study confirmed that long-term treatment with apheresis was at least able to stabilize CAD in most of the individuals with symptomatic elevated Lp(a) [181]. Furthermore, a multicenter retrospective study carried out by the G.I.L.A. (Gruppo Interdisciplinare Aferesi Lipoproteica) with the aim of analyzing the incidence of adverse cardiovascular events before and during lipoprotein apheresis treatment in subjects with an elevated level of Lp(a) (>60 mg/dL), chronic ischemic heart disease, and maximally tolerated lipid-lowering therapy as well as chronic ischemic heart disease, confirmed that lipoprotein apheresis carried long-term efficacy in terms of cardiovascular morbidity [182].

6.2. Innovative Strategies

Currently, antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), and microRNAs—all drugs that aim to reduce Lp(a) by targeting RNA molecules, regulating gene expression, and modulating protein production—are the most widely explored perspectives in this field. Recent trials involving ASOs, capable of inhibiting the expression of Apo(a), were the first randomized trials designed to assess an Lp(a)-lowering therapy [183,184]. Anti-Apo (a) ASOs, injected subcutaneously, bind to plasma proteins and are captured in the liver, where they bind to their target mRNA. Through this mechanism, the assembly of Lp(a) is blocked and the plasma levels of Lp(a) are reduced by more than 80% [185]. Two clinical trials recently revealed promising results according to the direct inhibition of Apo(a) synthesis by ASOs [185,186]: in the first trial, IONIS-APO(a)Rx was administered subcutaneously at dosages of 100 mg, 200 mg, and 300 mg once weekly for 4 weeks at each dose sequentially in patients with high Lp(a) levels in order to analyze the safety and efficacy of the ASO IONIS-APO(a)Rx. Subjects treated with 125–437 nmol/L and ≥438 nmol/L showed, respectively, a 62.8% and 67.7% reduction in Lp(a) concentrations compared with the placebo group [187].
A subsequent randomized, double-blind, placebo-controlled, and dose-ranging trial investigated the decrease in Lp(a) levels at different doses and intervals of IONIS-APO(a)Rx-Lrx [188]: a significant dose-dependent lowering of Lp(a) levels in all of the doses tested was observed. The highest cumulative dose (20 mg weekly) reduced Lp(a) by a mean of 80%. Phase 3 of this study is still ongoing.
Pelacarsen is an ASO that inhibits apolipoprotein, significantly lowers direct Lp(a), and has a neutral to mild lowering effect on LDL-C [189]. In a phase II clinical trial, pelacarsen was administered to patients through subcutaneous injections with different dosages, including 20 mg every 4 weeks, 20 mg every 2 weeks, 20 mg every week, 40 mg every 4 weeks, and 60 mg every 4 weeks; the circulating concentrations of Lp(a) were found to decrease by 35%, 58%, 80%, 56%, and 72%, respectively. Additionally, the side effects observed were mild and rare, with most being limited to reactions at the injection site [190]. Currently, a phase III study is ongoing and is set to end in 2024. This study aims to evaluate the effect of pelacarsen on cardiovascular endpoints in patients with Lp(a) levels ≥ 70 mg/dL who had previously experienced a cardiovascular event within the last 10 years. Participants are being treated with 80 mg of pelacarsen once per month, or a placebo [190].
The last promising frontier in reducing Lp(a) is siRNA. SiRNA functioning is based on post-transcriptional gene silencing: siRNA molecules are usually specific and efficient in the knockdown of disease-related genes [191], therefore reducing the production of a protein of interest. There are two siRNA-based Lp(a)-directed therapies in clinical development targeting Lp(a) mRNA—OLp(a)siran and SLN360—and both have been shown to lower Lp(a) plasma levels by up to 90% [192].
OLp(a)siran reduces Lp(a) levels by directly inhibiting Lp(a) messenger RNA translation in hepatocytes. Its effectiveness in reducing plasma Lp(a) concentration has been demonstrated in several clinical trials. In a phase 1 dose escalation study, participants tolerated a single dose of OLp(a)siran well, and experienced a reduction in Lp(a) concentration ranging from 71% to 97%. These effects persisted for several months after the administration of doses of 9 mg or higher [193]. Promising results were also obtained in phase 2 studies [194,195,196], which confirmed the efficacy of OLp(a)siran in reducing Lp(a) levels. These studies support the use of hepatocyte-targeted siRNA as a viable approach to reducing Lp(a) levels in individuals with an elevated plasma Lp(a) concentration.
Another promising siRNA is SLN360: this drug was tested in vitro for Lp(a) knockdown in primary hepatocytes and it specifically reduced Lp(a) expression in primary human hepatocytes with no relevant off-target effects; a sizeable (up to 95%) and long-lasting (≥9 weeks) reduction in serum Lp(a) was observed [197]. In a phase 1 study that involved 32 participants with elevated Lp(a), SLN360 was well tolerated; the trial confirmed that a dose-dependent lowering of plasma Lp(a) concentrations was observed [198]. These findings warrant further investigations to determine the safety and effectiveness of this siRNA (Table 3).

7. Conclusions and Perspectives

Genetic and observational evidence support a causal role of lipoprotein(a) in the development of CVD, PAD, CAVS, and HF. Hyperlipoproteinemia(a) represents a widespread health problem in the global population. The inter-individual variation in plasma concentrations of Lp(a) is large in the general population, and ranges from <1 mg/dL to >1000 mg/dL. Concentrations also differ according to ethnicities, with higher concentrations found in individuals of African descent when compared to populations with European or Asian heritage.
The accurate measurement of Lp(a) levels is crucial and requires the use of validated assays with traceability to ensure consistent cut-offs for high concentrations and proper risk assessment. The advancements in genotyping technologies over the past 15 years have allowed for the identification of genetic variations at the Lp(a) locus that are significantly linked to various vascular diseases. The genetically predicted levels of Lp(a) are consistently associated with CAD, calcific aortic valve stenosis, PAD, and, to a lesser degree, carotid atherosclerosis as well as HF. Therefore, Lp(a) is an essential component of residual cardiovascular risk and should be carefully monitored in patients with a history of cardiovascular disease or those at a high risk of developing it.
Lp(a) is minimally responsive to lifestyle or behavior changes, while other lipoprotein concentrations are affected by these factors, suggesting that Lp(a) levels are mostly genetically determined. The low effectiveness of lifestyle modifications and traditional lipid-lowering therapies, such as statins, niacin, or CETP inhibitors, has aroused greater interest in searching for new drugs that can reduce plasma Lp(a) levels. In addition to the LDL cholesterol-lowering effect, PCSK9 inhibitors are also capable of reducing Lp(a) levels by 25–30%. Another option is inclisiran, a siRNA that performs reductions in LDL and Lp(a) concentrations similar to those of PCSK9. It is also worth considering recently discovered novel therapies, such as ASOs, which reduce Lp(a) plasma levels by 60–80%. The last promising frontier in reducing Lp(a) is siRNA, which has been shown to lower Lp(a) plasma levels by to 90%. Apheresis is considered the most efficacious and well-tolerated treatment option for individuals with elevated levels of low-density lipoprotein cholesterol (LDLc) and Lp(a), as it can lead to a significant decrease of 60–70% in both LDL-C and Lp(a) concentrations. This reduction in lipid levels has been shown to result in a significant improvement in cardiovascular outcomes, with a 54–90% decrease in cardiovascular events observed in these patients.

Author Contributions

Conceptualization, P.V. and F.G.D.G.; methodology, N.F.; validation, E.P., A.M. and C.B.; writing—original draft preparation, L.M.T., F.P., C.C., C.R. and C.B.; writing—review and editing, P.V., F.G.D.G., A.M., A.P., M.Z., N.A. and N.F.; supervision, P.S. and G.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tsimikas, S.; Fazio, S.; Ferdinand, K.C.; Ginsberg, H.N.; Koschinsky, M.L.; Marcovina, S.M.; Moriarty, P.M.; Rader, D.J.; Remaley, A.T.; Reyes-Soffer, G.; et al. NHLBI Working Group Recommendations to Reduce Lipoprotein(a)-Mediated Risk of Cardiovascular Disease and Aortic Stenosis. J. Am. Coll. Cardiol. 2018, 71, 177–192. [Google Scholar] [CrossRef]
  2. Kronenberg, F.; Utermann, G. Lipoprotein(a): Resurrected by genetics. J. Intern. Med. 2013, 273, 6–30. [Google Scholar] [CrossRef] [PubMed]
  3. Kamstrup, P.R. Lipoprotein(a) and Cardiovascular Disease. Clin. Chem. 2021, 67, 154–166. [Google Scholar] [CrossRef] [PubMed]
  4. McCormick, S.P.A.; Schneider, W.J. Lipoprotein(a) catabolism: A case of multiple receptors. Pathology 2019, 51, 155–164. [Google Scholar] [CrossRef] [PubMed]
  5. Hrzenjak, A.; Frank, S.; Wo, X.; Zhou, Y.; Van Berkel, T.; Kostner, G.M. Galactose-specific asialoglycoprotein receptor is involved in lipoprotein (a) catabolism. Biochem. J. 2003, 376, 765–771. [Google Scholar] [CrossRef]
  6. Maranhao, R.C.; Carvalho, P.O.; Strunz, C.C.; Pileggi, F. Lipoprotein (a): Structure, pathophysiology and clinical implications. Arq. Bras. Cardiol. 2014, 103, 76–84. [Google Scholar] [CrossRef]
  7. Derby, C.A.; Crawford, S.L.; Pasternak, R.C.; Sowers, M.; Sternfeld, B.; Matthews, K.A. Lipid changes during the menopause transition in relation to age and weight: The Study of Women’s Health Across the Nation. Am. J. Epidemiol. 2009, 169, 1352–1361. [Google Scholar] [CrossRef]
  8. Enkhmaa, B.; Anuurad, E.; Berglund, L. Lipoprotein (a): Impact by ethnicity and environmental and medical conditions. J. Lipid Res. 2016, 57, 1111–1125. [Google Scholar] [CrossRef]
  9. Scholz, M.; Kraft, H.G.; Lingenhel, A.; Delport, R.; Vorster, E.H.; Bickeboller, H.; Utermann, G. Genetic control of lipoprotein(a) concentrations is different in Africans and Caucasians. Eur. J. Hum. Genet. 1999, 7, 169–178. [Google Scholar] [CrossRef]
  10. Boerwinkle, E.; Leffert, C.C.; Lin, J.; Lackner, C.; Chiesa, G.; Hobbs, H.H. Apolipoprotein(a) gene accounts for greater than 90% of the variation in plasma lipoprotein(a) concentrations. J. Clin. Invest. 1992, 90, 52–60. [Google Scholar] [CrossRef]
  11. Rao, F.; Schork, A.J.; Maihofer, A.X.; Nievergelt, C.M.; Marcovina, S.M.; Miller, E.R.; Witztum, J.L.; O’Connor, D.T.; Tsimikas, S. Heritability of Biomarkers of Oxidized Lipoproteins: Twin Pair Study. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 1704–1711. [Google Scholar] [CrossRef]
  12. Schmidt, K.; Noureen, A.; Kronenberg, F.; Utermann, G. Structure, function, and genetics of lipoprotein (a). J. Lipid Res. 2016, 57, 1339–1359. [Google Scholar] [CrossRef] [PubMed]
  13. Nordestgaard, B.G.; Langsted, A. Lipoprotein (a) as a cause of cardiovascular disease: Insights from epidemiology, genetics, and biology. J. Lipid Res. 2016, 57, 1953–1975. [Google Scholar] [CrossRef] [PubMed]
  14. Iannuzzo, G.; Tripaldella, M.; Mallardo, V.; Morgillo, M.; Vitelli, N.; Iannuzzi, A.; Aliberti, E.; Giallauria, F.; Tramontano, A.; Carluccio, R.; et al. Lipoprotein(a) Where Do We Stand? From the Physiopathology to Innovative Terapy. Biomedicines 2021, 9, 838. [Google Scholar] [CrossRef] [PubMed]
  15. McLean, J.W.; Tomlinson, J.E.; Kuang, W.J.; Eaton, D.L.; Chen, E.Y.; Fless, G.M.; Scanu, A.M.; Lawn, R.M. cDNA sequence of human apolipoprotein(a) is homologous to plasminogen. Nature 1987, 330, 132–137. [Google Scholar] [CrossRef]
  16. Lawn, R.M.; Schwartz, K.; Patthy, L. Convergent evolution of apolipoprotein(a) in primates and hedgehog. Proc. Natl. Acad. Sci. USA 1997, 94, 11992–11997. [Google Scholar] [CrossRef]
  17. Utermann, G. Genetic architecture and evolution of the lipoprotein(a) trait. Curr. Opin. Lipidol. 1999, 10, 133–141. [Google Scholar] [CrossRef]
  18. Grinstead, G.F.; Ellefson, R.D. Heterogeneity of lipoprotein Lp(a) and apolipoprotein(a). Clin. Chem. 1988, 34, 1036–1040. [Google Scholar] [CrossRef]
  19. Utermann, G.; Kraft, H.G.; Menzel, H.J.; Hopferwieser, T.; Seitz, C. Genetics of the quantitative Lp(a) lipoprotein trait. I. Relation of LP(a) glycoprotein phenotypes to Lp(a) lipoprotein concentrations in plasma. Hum. Genet. 1988, 78, 41–46. [Google Scholar] [CrossRef]
  20. Kraft, H.G.; Sandholzer, C.; Menzel, H.J.; Utermann, G. Apolipoprotein (a) alleles determine lipoprotein (a) particle density and concentration in plasma. Arterioscler. Thromb. 1992, 12, 302–306. [Google Scholar] [CrossRef]
  21. Lanktree, M.B.; Anand, S.S.; Yusuf, S.; Hegele, R.A.; Investigators, S. Comprehensive analysis of genomic variation in the LPA locus and its relationship to plasma lipoprotein(a) in South Asians, Chinese, and European Caucasians. Circ. Cardiovasc. Genet. 2010, 3, 39–46. [Google Scholar] [CrossRef] [PubMed]
  22. Chretien, J.P.; Coresh, J.; Berthier-Schaad, Y.; Kao, W.H.; Fink, N.E.; Klag, M.J.; Marcovina, S.M.; Giaculli, F.; Smith, M.W. 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, 917–923. [Google Scholar] [CrossRef] [PubMed]
  23. Clarke, R.; Peden, J.F.; Hopewell, J.C.; Kyriakou, T.; Goel, A.; Heath, S.C.; Parish, S.; Barlera, S.; Franzosi, M.G.; Rust, S.; et al. Genetic variants associated with Lp(a) lipoprotein level and coronary disease. N. Engl. J. Med. 2009, 361, 2518–2528. [Google Scholar] [CrossRef] [PubMed]
  24. Mack, S.; Coassin, S.; Rueedi, R.; Yousri, N.A.; Seppala, I.; Gieger, C.; Schonherr, S.; Forer, L.; Erhart, G.; Marques-Vidal, P.; et al. A genome-wide association meta-analysis on lipoprotein (a) concentrations adjusted for apolipoprotein (a) isoforms. J. Lipid Res. 2017, 58, 1834–1844. [Google Scholar] [CrossRef] [PubMed]
  25. Enkhmaa, B.; Berglund, L. Non-genetic influences on lipoprotein(a) concentrations. Atherosclerosis 2022, 349, 53–62. [Google Scholar] [CrossRef]
  26. Hopewell, J.C.; Haynes, R.; Baigent, C. The role of lipoprotein (a) in chronic kidney disease. J. Lipid Res. 2018, 59, 577–585. [Google Scholar] [CrossRef]
  27. Kon, V.; Yang, H.; Fazio, S. Residual Cardiovascular Risk in Chronic Kidney Disease: Role of High-density Lipoprotein. Arch. Med. Res. 2015, 46, 379–391. [Google Scholar] [CrossRef]
  28. Tuck, C.H.; Holleran, S.; Berglund, L. Hormonal regulation of lipoprotein(a) levels: Effects of estrogen replacement therapy on lipoprotein(a) and acute phase reactants in postmenopausal women. Arterioscler. Thromb. Vasc. Biol. 1997, 17, 1822–1829. [Google Scholar] [CrossRef]
  29. Shlipak, M.G.; Simon, J.A.; Vittinghoff, E.; Lin, F.; Barrett-Connor, E.; Knopp, R.H.; Levy, R.I.; Hulley, S.B. Estrogen and progestin, lipoprotein(a), and the risk of recurrent coronary heart disease events after menopause. JAMA 2000, 283, 1845–1852. [Google Scholar] [CrossRef]
  30. Sattar, N.; Clark, P.; Greer, I.A.; Shepherd, J.; Packard, C.J. Lipoprotein (a) levels in normal pregnancy and in pregnancy complicated with pre-eclampsia. Atherosclerosis 2000, 148, 407–411. [Google Scholar] [CrossRef]
  31. Kaur, H.; Werstuck, G.H. The Effect of Testosterone on Cardiovascular Disease and Cardiovascular Risk Factors in Men: A Review of Clinical and Preclinical Data. CJC Open 2021, 3, 1238–1248. [Google Scholar] [CrossRef] [PubMed]
  32. Marcovina, S.M.; Lippi, G.; Bagatell, C.J.; Bremner, W.J. Testosterone-induced suppression of lipoprotein(a) in normal men; relation to basal lipoprotein(a) level. Atherosclerosis 1996, 122, 89–95. [Google Scholar] [CrossRef] [PubMed]
  33. Johannsson, G.; Oscarsson, J.; Rosen, T.; Wiklund, O.; Olsson, G.; Wilhelmsen, L.; Bengtsson, B.A. Effects of 1 year of growth hormone therapy on serum lipoprotein levels in growth hormone-deficient adults. Influence of gender and Apo(a) and ApoE phenotypes. Arterioscler. Thromb. Vasc. Biol. 1995, 15, 2142–2150. [Google Scholar] [CrossRef] [PubMed]
  34. O’Halloran, D.J.; Wieringa, G.; Tsatsoulis, A.; Shalet, S.M. Increased serum lipoprotein(a) concentrations after growth hormone (GH) treatment in patients with isolated GH deficiency. Ann. Clin. Biochem. 1996, 33 Pt 4, 330–334. [Google Scholar] [CrossRef]
  35. Gentile, M.; Iannuzzo, G.; Mattiello, A.; Marotta, G.; Iannuzzi, A.; Panico, S.; Rubba, P. Association between Lp (a) and atherosclerosis in menopausal women without metabolic syndrome. Biomark. Med. 2016, 10, 397–402. [Google Scholar] [CrossRef]
  36. Maeda, S.; Abe, A.; Seishima, M.; Makino, K.; Noma, A.; Kawade, M. Transient changes of serum lipoprotein(a) as an acute phase protein. Atherosclerosis 1989, 78, 145–150. [Google Scholar] [CrossRef]
  37. Mora, S.; Kamstrup, P.R.; Rifai, N.; Nordestgaard, B.G.; Buring, J.E.; Ridker, P.M. Lipoprotein(a) and risk of type 2 diabetes. Clin. Chem. 2010, 56, 1252–1260. [Google Scholar] [CrossRef]
  38. Ye, Z.; Haycock, P.C.; Gurdasani, D.; Pomilla, C.; Boekholdt, S.M.; Tsimikas, S.; Khaw, K.T.; Wareham, N.J.; Sandhu, M.S.; Forouhi, N.G. The association between circulating lipoprotein(a) and type 2 diabetes: Is it causal? Diabetes 2014, 63, 332–342. [Google Scholar] [CrossRef]
  39. Jawi, M.M.; Frohlich, J.; Chan, S.Y. Lipoprotein(a) the Insurgent: A New Insight into the Structure, Function, Metabolism, Pathogenicity, and Medications Affecting Lipoprotein(a) Molecule. J. Lipids 2020, 2020, 3491764. [Google Scholar] [CrossRef]
  40. Richard, F.; Marecaux, N.; Dallongeville, J.; Devienne, M.; Tiem, N.; Fruchart, J.C.; Fantino, M.; Zylberberg, G.; Amouyel, P. Effect of smoking cessation on lipoprotein A-I and lipoprotein A-I:A-II levels. Metabolism 1997, 46, 711–715. [Google Scholar] [CrossRef]
  41. Wersch, J.W.; van Mackelenbergh, B.A.; Ubachs, J.M. Lipoprotein(a) in smoking and non-smoking pregnant women. Scand. J. Clin. Lab. Invest. 1994, 54, 361–364. [Google Scholar] [CrossRef] [PubMed]
  42. Afshar, M.; Pilote, L.; Dufresne, L.; Engert, J.C.; Thanassoulis, G. Lipoprotein(a) Interactions With Low-Density Lipoprotein Cholesterol and Other Cardiovascular Risk Factors in Premature Acute Coronary Syndrome (ACS). J. Am. Heart Assoc. 2016, 5, 003012. [Google Scholar] [CrossRef] [PubMed]
  43. Miles, L.A.; Plow, E.F. Lp(a): An interloper into the fibrinolytic system? Thromb. Haemost. 1990, 63, 331–335. [Google Scholar] [CrossRef] [PubMed]
  44. Criado, P.R.; Espinell, D.P.; Barreto, P.; Di Giacomo, T.H.; Sotto, M.N. Lipoprotein(a) and livedoid vasculopathy: A new thrombophilic factor? Med. Hypotheses 2015, 85, 670–674. [Google Scholar] [CrossRef]
  45. Edelberg, J.M.; Pizzo, S.V. Lipoprotein (a) in the regulation of fibrinolysis. J. Atheroscler. Thromb. 1995, 2 (Suppl. 1), S5–S7. [Google Scholar] [CrossRef] [PubMed]
  46. Enas, E.A.; Varkey, B.; Dharmarajan, T.S.; Pare, G.; Bahl, V.K. Lipoprotein(a): An independent, genetic, and causal factor for cardiovascular disease and acute myocardial infarction. Indian. Heart J. 2019, 71, 99–112. [Google Scholar] [CrossRef]
  47. Boffa, M.B.; Marcovina, S.M.; Koschinsky, M.L. Lipoprotein(a) as a risk factor for atherosclerosis and thrombosis: Mechanistic insights from animal models. Clin. Biochem. 2004, 37, 333–343. [Google Scholar] [CrossRef]
  48. Deb, A.; Caplice, N.M. Lipoprotein(a): New insights into mechanisms of atherogenesis and thrombosis. Clin. Cardiol. 2004, 27, 258–264. [Google Scholar] [CrossRef]
  49. Kapetanopoulos, A.; Fresser, F.; Millonig, G.; Shaul, Y.; Baier, G.; Utermann, G. Direct interaction of the extracellular matrix protein DANCE with apolipoprotein(a) mediated by the kringle IV-type 2 domain. Mol. Genet. Genom. 2002, 267, 440–446. [Google Scholar] [CrossRef]
  50. Boffa, M.B.; Koschinsky, M.L. Oxidized phospholipids as a unifying theory for lipoprotein(a) and cardiovascular disease. Nat. Rev. Cardiol. 2019, 16, 305–318. [Google Scholar] [CrossRef]
  51. Nielsen, L.B.; Nordestgaard, B.G.; Stender, S.; Niendorf, A.; Kjeldsen, K. Transfer of lipoprotein(a) and LDL into aortic intima in normal and in cholesterol-fed rabbits. Arterioscler. Thromb. Vasc. Biol. 1995, 15, 1492–1502. [Google Scholar] [CrossRef] [PubMed]
  52. Rath, M.; Niendorf, A.; Reblin, T.; Dietel, M.; Krebber, H.J.; Beisiegel, U. Detection and quantification of lipoprotein(a) in the arterial wall of 107 coronary bypass patients. Arteriosclerosis 1989, 9, 579–592. [Google Scholar] [CrossRef] [PubMed]
  53. 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 Pathol. Anat. Histopathol. 1990, 417, 105–111. [Google Scholar] [CrossRef]
  54. Nielsen, L.B.; Stender, S.; Kjeldsen, K.; Nordestgaard, B.G. Specific accumulation of lipoprotein(a) in balloon-injured rabbit aorta in vivo. Circ. Res. 1996, 78, 615–626. [Google Scholar] [CrossRef]
  55. Bihari-Varga, M.; Gruber, E.; Rotheneder, M.; Zechner, R.; Kostner, G.M. Interaction of lipoprotein Lp(a) and low density lipoprotein with glycosaminoglycans from human aorta. Arteriosclerosis 1988, 8, 851–857. [Google Scholar] [CrossRef] [PubMed]
  56. Nordestgaard, B.G. The vascular endothelial barrier--selective retention of lipoproteins. Curr. Opin. Lipidol. 1996, 7, 269–273. [Google Scholar] [CrossRef]
  57. Reyes-Soffer, G.; Ginsberg, H.N.; Berglund, L.; Duell, P.B.; Heffron, S.P.; Kamstrup, P.R.; Lloyd-Jones, D.M.; Marcovina, S.M.; Yeang, C.; Koschinsky, M.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, e48–e60. [Google Scholar] [CrossRef]
  58. Albers, J.J.; Hazzard, W.R. Immunochemical quantification of human plasma Lp(a) lipoprotein. Lipids 1974, 9, 15–26. [Google Scholar] [CrossRef]
  59. Marcovina, S.M.; Albers, J.J. Lipoprotein (a) measurements for clinical application. J. Lipid Res. 2016, 57, 526–537. [Google Scholar] [CrossRef]
  60. Wilson, D.P.; Jacobson, T.A.; Jones, P.H.; Koschinsky, M.L.; McNeal, C.J.; Nordestgaard, B.G.; Orringer, C.E. 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, 374–392. [Google Scholar] [CrossRef]
  61. Marcovina, S.M.; Koschinsky, M.L.; Albers, J.J.; 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, 1785–1796. [Google Scholar] [CrossRef] [PubMed]
  62. Tsimikas, S.; Fazio, S.; Viney, N.J.; Xia, S.; Witztum, J.L.; Marcovina, S.M. Relationship of lipoprotein(a) molar concentrations and mass according to lipoprotein(a) thresholds and apolipoprotein(a) isoform size. J. Clin. Lipidol. 2018, 12, 1313–1323. [Google Scholar] [CrossRef] [PubMed]
  63. Cegla, J.; France, M.; Marcovina, S.M.; Neely, R.D.G. Lp(a): When and how to measure it. Ann. Clin. Biochem. 2021, 58, 16–21. [Google Scholar] [CrossRef] [PubMed]
  64. Scharnagl, H.; Stojakovic, T.; Dieplinger, B.; Dieplinger, H.; Erhart, G.; Kostner, G.M.; Herrmann, M.; Marz, W.; Grammer, T.B. Comparison of lipoprotein (a) serum concentrations measured by six commercially available immunoassays. Atherosclerosis 2019, 289, 206–213. [Google Scholar] [CrossRef]
  65. 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. [Google Scholar] [CrossRef]
  66. Collaborators, G.B.D.C.o.D. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980-2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1736–1788. [Google Scholar] [CrossRef]
  67. Nordestgaard, B.G.; Chapman, M.J.; Ray, K.; Boren, J.; Andreotti, F.; Watts, G.F.; Ginsberg, H.; Amarenco, P.; Catapano, A.; Descamps, O.S.; et al. Lipoprotein(a) as a cardiovascular risk factor: Current status. Eur. Heart J. 2010, 31, 2844–2853. [Google Scholar] [CrossRef]
  68. Alvarez-Alvarez, M.M.; Zanetti, D.; Carreras-Torres, R.; Moral, P.; Athanasiadis, G. A survey of sub-Saharan gene flow into the Mediterranean at risk loci for coronary artery disease. Eur. J. Hum. Genet. 2017, 25, 472–476. [Google Scholar] [CrossRef]
  69. Malakar, A.K.; Choudhury, D.; Halder, B.; Paul, P.; Uddin, A.; Chakraborty, S. A review on coronary artery disease, its risk factors, and therapeutics. J. Cell Physiol. 2019, 234, 16812–16823. [Google Scholar] [CrossRef]
  70. Liu, T.; Yoon, W.S.; Lee, S.R. Recent Updates of Lipoprotein(a) and Cardiovascular Disease. Chonnam Med. J. 2021, 57, 36–43. [Google Scholar] [CrossRef]
  71. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A.; et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Eur. Heart J. 2020, 41, 111–188. [Google Scholar] [CrossRef] [PubMed]
  72. Kathiresan, S.; Willer, C.J.; Peloso, G.M.; Demissie, S.; Musunuru, K.; Schadt, E.E.; Kaplan, L.; Bennett, D.; Li, Y.; Tanaka, T.; et al. Common variants at 30 loci contribute to polygenic dyslipidemia. Nat. Genet. 2009, 41, 56–65. [Google Scholar] [CrossRef] [PubMed]
  73. Caplice, N.M.; Panetta, C.; Peterson, T.E.; Kleppe, L.S.; Mueske, C.S.; Kostner, G.M.; Broze, G.J., Jr.; Simari, R.D. Lipoprotein (a) binds and inactivates tissue factor pathway inhibitor: A novel link between lipoproteins and thrombosis. Blood 2001, 98, 2980–2987. [Google Scholar] [CrossRef] [PubMed]
  74. Tsimikas, S.; Brilakis, E.S.; Miller, E.R.; McConnell, J.P.; Lennon, R.J.; Kornman, K.S.; Witztum, J.L.; Berger, P.B. Oxidized phospholipids, Lp(a) lipoprotein, and coronary artery disease. N. Engl. J. Med. 2005, 353, 46–57. [Google Scholar] [CrossRef]
  75. Bottalico, L.A.; Keesler, G.A.; Fless, G.M.; Tabas, I. Cholesterol loading of macrophages leads to marked enhancement of native lipoprotein(a) and apoprotein(a) internalization and degradation. J. Biol. Chem. 1993, 268, 8569–8573. [Google Scholar] [CrossRef]
  76. Keesler, G.A.; Li, Y.; Skiba, P.J.; Fless, G.M.; Tabas, I. Macrophage foam cell lipoprotein(a)/apoprotein(a) receptor. Cell-surface localization, dependence of induction on new protein synthesis, and ligand specificity. Arterioscler. Thromb. 1994, 14, 1337–1345. [Google Scholar] [CrossRef]
  77. Nordestgaard, B.G. Triglyceride-Rich Lipoproteins and Atherosclerotic Cardiovascular Disease: New Insights From Epidemiology, Genetics, and Biology. Circ. Res. 2016, 118, 547–563. [Google Scholar] [CrossRef]
  78. Craig, W.Y.; Neveux, L.M.; Palomaki, G.E.; Cleveland, M.M.; Haddow, J.E. Lipoprotein(a) as a risk factor for ischemic heart disease: Metaanalysis of prospective studies. Clin. Chem. 1998, 44, 2301–2306. [Google Scholar] [CrossRef]
  79. 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. [Google Scholar] [CrossRef]
  80. Jauhiainen, M.; Koskinen, P.; Ehnholm, C.; Frick, M.H.; Manttari, M.; Manninen, V.; Huttunen, J.K. Lipoprotein (a) and coronary heart disease risk: A nested case-control study of the Helsinki Heart Study participants. Atherosclerosis 1991, 89, 59–67. [Google Scholar] [CrossRef]
  81. Alfthan, G.; Pekkanen, J.; Jauhiainen, M.; Pitkaniemi, J.; Karvonen, M.; Tuomilehto, J.; Salonen, J.T.; Ehnholm, C. Relation of serum homocysteine and lipoprotein(a) concentrations to atherosclerotic disease in a prospective Finnish population based study. Atherosclerosis 1994, 106, 9–19. [Google Scholar] [CrossRef] [PubMed]
  82. Klausen, I.C.; Sjol, A.; Hansen, P.S.; Gerdes, L.U.; Moller, L.; Lemming, L.; Schroll, M.; Faergeman, O. Apolipoprotein(a) isoforms and coronary heart disease in men: A nested case-control study. Atherosclerosis 1997, 132, 77–84. [Google Scholar] [CrossRef] [PubMed]
  83. Wild, S.H.; Fortmann, S.P.; Marcovina, S.M. A prospective case-control study of lipoprotein(a) levels and apo(a) size and risk of coronary heart disease in Stanford Five-City Project participants. Arterioscler. Thromb. Vasc. Biol. 1997, 17, 239–245. [Google Scholar] [CrossRef] [PubMed]
  84. Danesh, J.; Collins, R.; Peto, R. Lipoprotein(a) and coronary heart disease. Meta-analysis of prospective studies. Circulation 2000, 102, 1082–1085. [Google Scholar] [CrossRef] [PubMed]
  85. Pare, G.; Caku, A.; McQueen, M.; Anand, S.S.; Enas, E.; Clarke, R.; Boffa, M.B.; Koschinsky, M.; Wang, X.; Yusuf, S.; et al. Lipoprotein(a) Levels and the Risk of Myocardial Infarction Among 7 Ethnic Groups. Circulation 2019, 139, 1472–1482. [Google Scholar] [CrossRef]
  86. Dube, J.B.; Boffa, M.B.; Hegele, R.A.; Koschinsky, M.L. Lipoprotein(a): More interesting than ever after 50 years. Curr. Opin. Lipidol. 2012, 23, 133–140. [Google Scholar] [CrossRef]
  87. Kostner, K.M.; Marz, W.; Kostner, G.M. When should we measure lipoprotein (a)? Eur. Heart J. 2013, 34, 3268–3276. [Google Scholar] [CrossRef]
  88. Jansen, H.; Samani, N.J.; Schunkert, H. Mendelian randomization studies in coronary artery disease. Eur. Heart J. 2014, 35, 1917–1924. [Google Scholar] [CrossRef]
  89. Page, M.M.; Ellis, K.L.; Pang, J.; Chan, D.C.; Hooper, A.J.; Bell, D.A.; Burnett, J.R.; Watts, G.F. Coronary artery disease and the risk-associated LPA variants, rs3798220 and rs10455872, in patients with suspected familial hypercholesterolaemia. Clin. Chim. Acta 2020, 510, 211–215. [Google Scholar] [CrossRef]
  90. Kronenberg, F. Lipoprotein(a) and aortic valve stenosis: Work in progress. Eur. Heart J. 2022, 43, 3968–3970. [Google Scholar] [CrossRef]
  91. Youssef, A.; Clark, J.R.; Koschinsky, M.L.; Boffa, M.B. Lipoprotein(a): Expanding our knowledge of aortic valve narrowing. Trends Cardiovasc. Med. 2021, 31, 305–311. [Google Scholar] [CrossRef] [PubMed]
  92. Thanassoulis, G.; Campbell, C.Y.; Owens, D.S.; Smith, J.G.; Smith, A.V.; Peloso, G.M.; Kerr, K.F.; Pechlivanis, S.; Budoff, M.J.; Harris, T.B.; et al. Genetic associations with valvular calcification and aortic stenosis. N. Engl. J. Med. 2013, 368, 503–512. [Google Scholar] [CrossRef] [PubMed]
  93. Arsenault, B.J.; Boekholdt, S.M.; Dube, M.P.; Rheaume, E.; Wareham, N.J.; Khaw, K.T.; Sandhu, M.S.; Tardif, J.C. Lipoprotein(a) levels, genotype, and incident aortic valve stenosis: A prospective Mendelian randomization study and replication in a case-control cohort. Circ. Cardiovasc. Genet. 2014, 7, 304–310. [Google Scholar] [CrossRef]
  94. Zheng, K.H.; Tsimikas, S.; Pawade, T.; Kroon, J.; Jenkins, W.S.A.; Doris, M.K.; White, A.C.; Timmers, N.; Hjortnaes, J.; Rogers, M.A.; et al. Lipoprotein(a) and Oxidized Phospholipids Promote Valve Calcification in Patients With Aortic Stenosis. J. Am. Coll. Cardiol. 2019, 73, 2150–2162. [Google Scholar] [CrossRef]
  95. Kaiser, Y.; van der Toorn, J.E.; Singh, S.S.; Zheng, K.H.; Kavousi, M.; Sijbrands, E.J.G.; Stroes, E.S.G.; Vernooij, M.W.; de Rijke, Y.B.; Boekholdt, S.M.; et al. Lipoprotein(a) is associated with the onset but not the progression of aortic valve calcification. Eur. Heart J. 2022, 43, 3960–3967. [Google Scholar] [CrossRef] [PubMed]
  96. Arsenault, B.J.; Kamstrup, P.R. Lipoprotein(a) and cardiovascular and valvular diseases: A genetic epidemiological perspective. Atherosclerosis 2022, 349, 7–16. [Google Scholar] [CrossRef] [PubMed]
  97. Hrovat, K.; Rehberger Likozar, A.; Zupan, J.; Sebestjen, M. Gene Expression Profiling of Markers of Inflammation, Angiogenesis, Coagulation and Fibrinolysis in Patients with Coronary Artery Disease with Very High Lipoprotein(a) Levels Treated with PCSK9 Inhibitors. J. Cardiovasc. Dev. Dis. 2022, 9, 211. [Google Scholar] [CrossRef]
  98. Schnitzler, J.G.; Ali, L.; Groenen, A.G.; Kaiser, Y.; Kroon, J. Lipoprotein(a) as Orchestrator of Calcific Aortic Valve Stenosis. Biomolecules 2019, 9, 760. [Google Scholar] [CrossRef]
  99. Capoulade, R.; Chan, K.L.; Yeang, C.; Mathieu, P.; Bosse, Y.; Dumesnil, J.G.; Tam, J.W.; Teo, K.K.; Mahmut, A.; Yang, X.; et al. Oxidized Phospholipids, Lipoprotein(a), and Progression of Calcific Aortic Valve Stenosis. J. Am. Coll. Cardiol. 2015, 66, 1236–1246. [Google Scholar] [CrossRef]
  100. Mahmut, A.; Boulanger, M.C.; El Husseini, D.; Fournier, D.; Bouchareb, R.; Despres, J.P.; Pibarot, P.; Bosse, Y.; Mathieu, P. Elevated expression of lipoprotein-associated phospholipase A2 in calcific aortic valve disease: Implications for valve mineralization. J. Am. Coll. Cardiol. 2014, 63, 460–469. [Google Scholar] [CrossRef]
  101. O’Brien, K.D.; Reichenbach, D.D.; Marcovina, S.M.; Kuusisto, J.; Alpers, C.E.; Otto, C.M. Apolipoproteins B, (a), and E accumulate in the morphologically early lesion of ‘degenerative’ valvular aortic stenosis. Arterioscler. Thromb. Vasc. Biol. 1996, 16, 523–532. [Google Scholar] [CrossRef] [PubMed]
  102. Guan, W.; Cao, J.; Steffen, B.T.; Post, W.S.; Stein, J.H.; Tattersall, M.C.; Kaufman, J.D.; McConnell, J.P.; Hoefner, D.M.; Warnick, R.; 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, 996–1001. [Google Scholar] [CrossRef]
  103. Dhaliwal, G.; Mukherjee, D. Peripheral arterial disease: Epidemiology, natural history, diagnosis and treatment. Int. J. Angiol. 2007, 16, 36–44. [Google Scholar] [CrossRef] [PubMed]
  104. Bartholomew, J.R.; Olin, J.W. Pathophysiology of peripheral arterial disease and risk factors for its development. Cleve Clin. J. Med. 2006, 73 (Suppl. 4), S8–S14. [Google Scholar] [CrossRef]
  105. Kosmas, C.E.; Silverio, D.; Sourlas, A.; Peralta, R.; Montan, P.D.; Guzman, E.; Garcia, M.J. Role of lipoprotein (a) in peripheral arterial disease. Ann. Transl. Med. 2019, 7, S242. [Google Scholar] [CrossRef]
  106. Ridker, P.M.; Stampfer, M.J.; Rifai, N. Novel risk factors for systemic atherosclerosis: A comparison of C-reactive protein, fibrinogen, homocysteine, lipoprotein(a), and standard cholesterol screening as predictors of peripheral arterial disease. JAMA 2001, 285, 2481–2485. [Google Scholar] [CrossRef] [PubMed]
  107. Kamstrup, P.R.; Tybjaerg-Hansen, A.; Nordestgaard, B.G. Genetic evidence that lipoprotein(a) associates with atherosclerotic stenosis rather than venous thrombosis. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 1732–1741. [Google Scholar] [CrossRef]
  108. Klarin, D.; Lynch, J.; Aragam, K.; Chaffin, M.; Assimes, T.L.; Huang, J.; Lee, K.M.; Shao, Q.; Huffman, J.E.; Natarajan, P.; et al. Genome-wide association study of peripheral artery disease in the Million Veteran Program. Nat. Med. 2019, 25, 1274–1279. [Google Scholar] [CrossRef]
  109. Laschkolnig, A.; Kollerits, B.; Lamina, C.; Meisinger, C.; Rantner, B.; Stadler, M.; Peters, A.; Koenig, W.; Stockl, A.; Dahnhardt, D.; et al. Lipoprotein (a) concentrations, apolipoprotein (a) phenotypes, and peripheral arterial disease in three independent cohorts. Cardiovasc. Res. 2014, 103, 28–36. [Google Scholar] [CrossRef]
  110. Forbang, N.I.; Criqui, M.H.; Allison, M.A.; Ix, J.H.; Steffen, B.T.; Cushman, M.; Tsai, M.Y. 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, 453–458. [Google Scholar] [CrossRef]
  111. Volpato, S.; Vigna, G.B.; McDermott, M.M.; Cavalieri, M.; Maraldi, C.; Lauretani, F.; Bandinelli, S.; Zuliani, G.; Guralnik, J.M.; Fellin, R.; et al. Lipoprotein(a), inflammation, and peripheral arterial disease in a community-based sample of older men and women (the InCHIANTI study). Am. J. Cardiol. 2010, 105, 1825–1830. [Google Scholar] [CrossRef] [PubMed]
  112. Gurdasani, D.; Sjouke, B.; Tsimikas, S.; Hovingh, G.K.; Luben, R.N.; Wainwright, N.W.; Pomilla, C.; Wareham, N.J.; Khaw, K.T.; Boekholdt, S.M.; et al. Lipoprotein(a) and risk of coronary, cerebrovascular, and peripheral artery disease: The EPIC-Norfolk prospective population study. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 3058–3065. [Google Scholar] [CrossRef] [PubMed]
  113. Catapano, A.L.; Graham, I.; De Backer, G.; Wiklund, O.; Chapman, M.J.; Drexel, H.; Hoes, A.W.; Jennings, C.S.; Landmesser, U.; Pedersen, T.R.; et al. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias. Rev. Esp. Cardiol. 2017, 70, 115. [Google Scholar] [CrossRef] [PubMed]
  114. Grundy, S.M.; Stone, N.J.; Bailey, A.L.; Beam, C.; Birtcher, K.K.; Blumenthal, R.S.; Braun, L.T.; de Ferranti, S.; Faiella-Tommasino, J.; Forman, D.E.; et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 2019, 73, 3168–3209. [Google Scholar] [CrossRef]
  115. Valentine, R.J.; Grayburn, P.A.; Vega, G.L.; Grundy, S.M. Lp(a) lipoprotein is an independent, discriminating risk factor for premature peripheral atherosclerosis among white men. Arch. Intern. Med. 1994, 154, 801–806. [Google Scholar] [CrossRef]
  116. Tseng, C.H. Lipoprotein(a) is an independent risk factor for peripheral arterial disease in Chinese type 2 diabetic patients in Taiwan. Diabetes Care 2004, 27, 517–521. [Google Scholar] [CrossRef]
  117. Cheng, S.W.; Ting, A.C.; Wong, J. Lipoprotein (a) and its relationship to risk factors and severity of atherosclerotic peripheral vascular disease. Eur. J. Vasc. Endovasc. Surg. 1997, 14, 17–23. [Google Scholar] [CrossRef]
  118. Sanchez Munoz-Torrero, J.F.; Rico-Martin, S.; Alvarez, L.R.; Aguilar, E.; Alcala, J.N.; Monreal, M.; Investigators, F. Lipoprotein (a) levels and outcomes in stable outpatients with symptomatic artery disease. Atherosclerosis 2018, 276, 10–14. [Google Scholar] [CrossRef]
  119. Golledge, J.; Rowbotham, S.; Velu, R.; Quigley, F.; Jenkins, J.; Bourke, M.; Bourke, B.; Thanigaimani, S.; Chan, D.C.; Watts, G.F. Association of Serum Lipoprotein (a) With the Requirement for a Peripheral Artery Operation and the Incidence of Major Adverse Cardiovascular Events in People With Peripheral Artery Disease. J. Am. Heart Assoc. 2020, 9, e015355. [Google Scholar] [CrossRef]
  120. Yanaka, K.; Akahori, H.; Imanaka, T.; Miki, K.; Yoshihara, N.; Kimura, T.; Tanaka, T.; Asakura, M.; Ishihara, M. Impact of lipoprotein(a) levels on primary patency after endovascular therapy for femoropopliteal lesions. Heart Vessels 2023, 38, 171–176. [Google Scholar] [CrossRef]
  121. Masson, W.; Lobo, M.; Barbagelata, L.; Molinero, G.; Bluro, I.; Nogueira, J.P. Elevated lipoprotein (a) levels and risk of peripheral artery disease outcomes: A systematic review. Vasc. Med. 2022, 27, 385–391. [Google Scholar] [CrossRef] [PubMed]
  122. Song, P.; Fang, Z.; Wang, H.; Cai, Y.; Rahimi, K.; Zhu, Y.; Fowkes, F.G.R.; Fowkes, F.J.I.; Rudan, I. Global and regional prevalence, burden, and risk factors for carotid atherosclerosis: A systematic review, meta-analysis, and modelling study. Lancet Glob. Health 2020, 8, e721–e729. [Google Scholar] [CrossRef]
  123. Lampsas, S.; Xenou, M.; Oikonomou, E.; Pantelidis, P.; Lysandrou, A.; Sarantos, S.; Goliopoulou, A.; Kalogeras, K.; Tsigkou, V.; Kalpis, A.; et al. Lipoprotein(a) in Atherosclerotic Diseases: From Pathophysiology to Diagnosis and Treatment. Molecules 2023, 28, 969. [Google Scholar] [CrossRef]
  124. Klein, J.H.; Hegele, R.A.; Hackam, D.G.; Koschinsky, M.L.; Huff, M.W.; Spence, J.D. Lipoprotein(a) is associated differentially with carotid stenosis, occlusion, and total plaque area. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 1851–1856. [Google Scholar] [CrossRef] [PubMed]
  125. Steffen, B.T.; Thanassoulis, G.; Duprez, D.; Stein, J.H.; Karger, A.B.; Tattersall, M.C.; Kaufman, J.D.; Guan, W.; Tsai, M.Y. Race-Based Differences in Lipoprotein(a)-Associated Risk of Carotid Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 523–529. [Google Scholar] [CrossRef] [PubMed]
  126. Nasr, N.; Ruidavets, J.B.; Farghali, A.; Guidolin, B.; Perret, B.; Larrue, V. Lipoprotein (a) and carotid atherosclerosis in young patients with stroke. Stroke 2011, 42, 3616–3618. [Google Scholar] [CrossRef]
  127. Zhao, X.Q.; Hatsukami, T.S.; Hippe, D.S.; Sun, J.; Balu, N.; Isquith, D.A.; Crouse, J.R., 3rd; Anderson, T.; Huston, J., 3rd; Polissar, N.; et al. Clinical factors associated with high-risk carotid plaque features as assessed by magnetic resonance imaging in patients with established vascular disease (from the AIM-HIGH Study). Am. J. Cardiol. 2014, 114, 1412–1419. [Google Scholar] [CrossRef]
  128. Hippe, D.S.; Phan, B.A.P.; Sun, J.; Isquith, D.A.; O’Brien, K.D.; Crouse, J.R.; Anderson, T.; Huston, J.; Marcovina, S.M.; Hatsukami, T.S.; et al. Lp(a) (Lipoprotein(a)) Levels Predict Progression of Carotid Atherosclerosis in Subjects With Atherosclerotic Cardiovascular Disease on Intensive Lipid Therapy: An Analysis of the AIM-HIGH (Atherothrombosis Intervention in Metabolic Syndrome With Low HDL/High Triglycerides: Impact on Global Health Outcomes) Carotid Magnetic Resonance Imaging Substudy-Brief Report. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 673–678. [Google Scholar] [CrossRef]
  129. Ooi, E.M.; Ellis, K.L.; Barrett, P.H.R.; Watts, G.F.; Hung, J.; Beilby, J.P.; Thompson, P.L.; Stobie, P.; McQuillan, B.M. Lipoprotein(a) and apolipoprotein(a) isoform size: Associations with angiographic extent and severity of coronary artery disease, and carotid artery plaque. Atherosclerosis 2018, 275, 232–238. [Google Scholar] [CrossRef]
  130. Schreiner, P.J.; Heiss, G.; Tyroler, H.A.; Morrisett, J.D.; Davis, C.E.; Smith, R. Race and gender differences in the association of Lp(a) with carotid artery wall thickness. The Atherosclerosis Risk in Communities (ARIC) Study. Arterioscler. Thromb. Vasc. Biol. 1996, 16, 471–478. [Google Scholar] [CrossRef]
  131. Van Dam-Nolen, D.H.K.; van Dijk, A.C.; Crombag, G.; Lucci, C.; Kooi, M.E.; Hendrikse, J.; Nederkoorn, P.J.; Daemen, M.; van der Steen, A.F.W.; Koudstaal, P.J.; et al. Lipoprotein(a) levels and atherosclerotic plaque characteristics in the carotid artery: The Plaque at RISK (PARISK) study. Atherosclerosis 2021, 329, 22–29. [Google Scholar] [CrossRef] [PubMed]
  132. Zenker, G.; Koltringer, P.; Bone, G.; Niederkorn, K.; Pfeiffer, K.; Jurgens, G. Lipoprotein(a) as a strong indicator for cerebrovascular disease. Stroke 1986, 17, 942–945. [Google Scholar] [CrossRef] [PubMed]
  133. Langsted, A.; Nordestgaard, B.G.; Kamstrup, P.R. Elevated Lipoprotein(a) and Risk of Ischemic Stroke. J. Am. Coll. Cardiol. 2019, 74, 54–66. [Google Scholar] [CrossRef]
  134. van Dijk, R.A.; Kolodgie, F.; Ravandi, A.; Leibundgut, G.; Hu, P.P.; Prasad, A.; Mahmud, E.; Dennis, E.; Curtiss, L.K.; Witztum, J.L.; 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, 2773–2790. [Google Scholar] [CrossRef] [PubMed]
  135. Kumar, P.; Swarnkar, P.; Misra, S.; Nath, M. Lipoprotein (a) level as a risk factor for stroke and its subtype: A systematic review and meta-analysis. Sci. Rep. 2021, 11, 15660. [Google Scholar] [CrossRef] [PubMed]
  136. Ding, W.Y.; Protty, M.B.; Davies, I.G.; Lip, G.Y.H. Relationship between lipoproteins, thrombosis, and atrial fibrillation. Cardiovasc. Res. 2022, 118, 716–731. [Google Scholar] [CrossRef]
  137. Igarashi, Y.; Yamaura, M.; Ito, M.; Inuzuka, H.; Ojima, K.; Aizawa, Y. Elevated serum lipoprotein(a) is a risk factor for left atrial thrombus in patients with chronic atrial fibrillation: A transesophageal echocardiographic study. Am. Heart J. 1998, 136, 965–971. [Google Scholar] [CrossRef] [PubMed]
  138. Yan, S.; Li, Q.; Xia, Z.; Yan, S.; Wei, Y.; Hong, K.; Wu, Y.; Li, J.; Cheng, X. Risk factors of thromboembolism in nonvalvular atrial fibrillation patients with low CHA2DS2-VASc score. Medicine 2019, 98, e14549. [Google Scholar] [CrossRef]
  139. Aronis, K.N.; Zhao, D.; Hoogeveen, R.C.; Alonso, A.; Ballantyne, C.M.; Guallar, E.; Jones, S.R.; Martin, S.S.; Nazarian, S.; Steffen, B.T.; et al. Associations of Lipoprotein(a) Levels With Incident Atrial Fibrillation and Ischemic Stroke: The ARIC (Atherosclerosis Risk in Communities) Study. J. Am. Heart Assoc. 2017, 6, e007372. [Google Scholar] [CrossRef]
  140. Enkhmaa, B.; Anuurad, E.; Zhang, W.; Tran, T.; Berglund, L. Lipoprotein(a): Genotype-phenotype relationship and impact on atherogenic risk. Metab. Syndr. Relat. Disord. 2011, 9, 411–418. [Google Scholar] [CrossRef]
  141. Kario, K.; Matsuo, T.; Kobayashi, H.; Asada, R.; Matsuo, M. ‘Silent’ cerebral infarction is associated with hypercoagulability, endothelial cell damage, and high Lp(a) levels in elderly Japanese. Arterioscler. Thromb. Vasc. Biol. 1996, 16, 734–741. [Google Scholar] [CrossRef] [PubMed]
  142. Nagayama, M.; Shinohara, Y.; Nagayama, T. Lipoprotein(a) and ischemic cerebrovascular disease in young adults. Stroke 1994, 25, 74–78. [Google Scholar] [CrossRef] [PubMed]
  143. Kamstrup, P.R.; Nordestgaard, B.G. Elevated Lipoprotein(a) Levels, LPA Risk Genotypes, and Increased Risk of Heart Failure in the General Population. JACC Heart Fail. 2016, 4, 78–87. [Google Scholar] [CrossRef] [PubMed]
  144. Agarwala, A.; Pokharel, Y.; Saeed, A.; Sun, W.; Virani, S.S.; Nambi, V.; Ndumele, C.; Shahar, E.; Heiss, G.; Boerwinkle, E.; et al. The association of lipoprotein(a) with incident heart failure hospitalization: Atherosclerosis Risk in Communities study. Atherosclerosis 2017, 262, 131–137. [Google Scholar] [CrossRef]
  145. Kamstrup, P.R.; Benn, M.; Tybjaerg-Hansen, A.; Nordestgaard, B.G. Extreme lipoprotein(a) levels and risk of myocardial infarction in the general population: The Copenhagen City Heart Study. Circulation 2008, 117, 176–184. [Google Scholar] [CrossRef]
  146. Steffen, B.T.; Duprez, D.; Bertoni, A.G.; Guan, W.; Tsai, M.Y. 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, 2498–2504. [Google Scholar] [CrossRef] [PubMed]
  147. Wu, B.; Zhang, Z.; Long, J.; Zhao, H.; Zeng, F. Association between lipoprotein (a) and heart failure with reduced ejection fraction development. J. Clin. Lab. Anal. 2022, 36, e24083. [Google Scholar] [CrossRef] [PubMed]
  148. Gudbjartsson, D.F.; Thorgeirsson, G.; Sulem, P.; Helgadottir, A.; Gylfason, A.; Saemundsdottir, J.; Bjornsson, E.; Norddahl, G.L.; Jonasdottir, A.; Jonasdottir, A.; et al. Lipoprotein(a) Concentration and Risks of Cardiovascular Disease and Diabetes. J. Am. Coll. Cardiol. 2019, 74, 2982–2994. [Google Scholar] [CrossRef] [PubMed]
  149. Rehberger Likozar, A.; Zavrtanik, M.; Sebestjen, M. Lipoprotein(a) in atherosclerosis: From pathophysiology to clinical relevance and treatment options. Ann. Med. 2020, 52, 162–177. [Google Scholar] [CrossRef]
  150. Khera, A.V.; Everett, B.M.; Caulfield, M.P.; Hantash, F.M.; Wohlgemuth, J.; Ridker, P.M.; Mora, S. 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, 635–642. [Google Scholar] [CrossRef]
  151. Yeang, C.; Hung, M.Y.; Byun, Y.S.; Clopton, P.; Yang, X.; Witztum, J.L.; Tsimikas, S. Effect of therapeutic interventions on oxidized phospholipids on apolipoprotein B100 and lipoprotein(a). J. Clin. Lipidol. 2016, 10, 594–603. [Google Scholar] [CrossRef]
  152. Tsimikas, S.; Gordts, P.; Nora, C.; Yeang, C.; Witztum, J.L. Statin therapy increases lipoprotein(a) levels. Eur. Heart J. 2020, 41, 2275–2284. [Google Scholar] [CrossRef]
  153. Sahebkar, A.; Simental-Mendia, L.E.; Watts, G.F.; Serban, M.C.; Banach, M.; Lipid and Blood Pressure Meta-analysis Collaboration (LBPMC) Group. Comparison of the effects of fibrates versus statins on plasma lipoprotein(a) concentrations: A systematic review and meta-analysis of head-to-head randomized controlled trials. BMC Med. 2017, 15, 22. [Google Scholar] [CrossRef]
  154. Fras, Z. Increased cardiovascular risk associated with hyperlipoproteinemia (a) and the challenges of current and future therapeutic possibilities. Anatol. J. Cardiol. 2020, 23, 60–69. [Google Scholar] [CrossRef] [PubMed]
  155. Guyton, J.R. Niacin in cardiovascular prevention: Mechanisms, efficacy, and safety. Curr. Opin. Lipidol. 2007, 18, 415–420. [Google Scholar] [CrossRef]
  156. Carlson, L.A.; Hamsten, A.; Asplund, A. Pronounced lowering of serum levels of lipoprotein Lp(a) in hyperlipidaemic subjects treated with nicotinic acid. J. Intern. Med. 1989, 226, 271–276. [Google Scholar] [CrossRef]
  157. Guyton, J.R.; Slee, A.E.; Anderson, T.; Fleg, J.L.; Goldberg, R.B.; Kashyap, M.L.; Marcovina, S.M.; Nash, S.D.; O’Brien, K.D.; Weintraub, W.S.; et al. Relationship of lipoproteins to cardiovascular events: The AIM-HIGH Trial (Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglycerides and Impact on Global Health Outcomes). J. Am. Coll. Cardiol. 2013, 62, 1580–1584. [Google Scholar] [CrossRef]
  158. Group, H.T.C.; Landray, M.J.; Haynes, R.; Hopewell, J.C.; Parish, S.; Aung, T.; Tomson, J.; Wallendszus, K.; Craig, M.; Jiang, L.; et al. Effects of extended-release niacin with laropiprant in high-risk patients. N. Engl. J. Med. 2014, 371, 203–212. [Google Scholar] [CrossRef]
  159. Barter, P.J.; Rye, K.A. Cholesteryl ester transfer protein inhibition as a strategy to reduce cardiovascular risk. J. Lipid Res. 2012, 53, 1755–1766. [Google Scholar] [CrossRef] [PubMed]
  160. Barter, P.J.; Rye, K.A. Cholesteryl Ester Transfer Protein Inhibition Is Not Yet Dead-Pro. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 439–441. [Google Scholar] [CrossRef] [PubMed]
  161. Handhle, A.; Viljoen, A.; Wierzbicki, A.S. Elevated Lipoprotein(a): Background, Current Insights and Future Potential Therapies. Vasc. Health Risk Manag. 2021, 17, 527–542. [Google Scholar] [CrossRef] [PubMed]
  162. Arsenault, B.J.; Petrides, F.; Tabet, F.; Bao, W.; Hovingh, G.K.; Boekholdt, S.M.; Ramin-Mangata, S.; Meilhac, O.; DeMicco, D.; Rye, K.A.; 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, 130–136. [Google Scholar] [CrossRef] [PubMed]
  163. Ortega Martinez de Victoria, E. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors. A new drug class for the treatment of hypercholesterolemia. Endocrinol. Diabetes Nutr. 2017, 64, 341–344. [Google Scholar] [CrossRef] [PubMed]
  164. Kronenberg, F. Lipoprotein(a). Handb. Exp. Pharmacol. 2022, 270, 201–232. [Google Scholar] [CrossRef] [PubMed]
  165. Gaudet, D.; Watts, G.F.; Robinson, J.G.; Minini, P.; Sasiela, W.J.; Edelberg, J.; Louie, M.J.; Raal, F.J. Effect of Alirocumab on Lipoprotein(a) Over >/=1.5 Years (from the Phase 3 ODYSSEY Program). Am. J. Cardiol. 2017, 119, 40–46. [Google Scholar] [CrossRef]
  166. Raal, F.J.; Giugliano, R.P.; Sabatine, M.S.; Koren, M.J.; Blom, D.; Seidah, N.G.; Honarpour, N.; Lira, A.; Xue, A.; Chiruvolu, P.; et al. 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, 1086–1096. [Google Scholar] [CrossRef]
  167. Sabatine, M.S.; Giugliano, R.P.; Keech, A.C.; Honarpour, N.; Wiviott, S.D.; Murphy, S.A.; Kuder, J.F.; Wang, H.; Liu, T.; Wasserman, S.M.; et al. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N. Engl. J. Med. 2017, 376, 1713–1722. [Google Scholar] [CrossRef]
  168. Schwartz, G.G.; Steg, P.G.; Szarek, M.; Bhatt, D.L.; Bittner, V.A.; Diaz, R.; Edelberg, J.M.; Goodman, S.G.; Hanotin, C.; Harrington, R.A.; et al. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N. Engl. J. Med. 2018, 379, 2097–2107. [Google Scholar] [CrossRef]
  169. Ray, K.K.; Wright, R.S.; Kallend, D.; Koenig, W.; Leiter, L.A.; Raal, F.J.; Bisch, J.A.; Richardson, T.; Jaros, M.; Wijngaard, P.L.J.; et al. Two Phase 3 Trials of Inclisiran in Patients with Elevated LDL Cholesterol. N. Engl. J. Med. 2020, 382, 1507–1519. [Google Scholar] [CrossRef]
  170. Raal, F.J.; Kallend, D.; Ray, K.K.; Turner, T.; Koenig, W.; Wright, R.S.; Wijngaard, P.L.J.; Curcio, D.; Jaros, M.J.; Leiter, L.A.; et al. Inclisiran for the Treatment of Heterozygous Familial Hypercholesterolemia. N. Engl. J. Med. 2020, 382, 1520–1530. [Google Scholar] [CrossRef]
  171. Sahebkar, A.; Watts, G.F. New LDL-cholesterol lowering therapies: Pharmacology, clinical trials, and relevance to acute coronary syndromes. Clin. Ther. 2013, 35, 1082–1098. [Google Scholar] [CrossRef] [PubMed]
  172. Cuchel, M.; Meagher, E.A.; du Toit Theron, H.; Blom, D.J.; Marais, A.D.; Hegele, R.A.; Averna, M.R.; Sirtori, C.R.; Shah, P.K.; Gaudet, D.; et al. Efficacy and safety of a microsomal triglyceride transfer protein inhibitor in patients with homozygous familial hypercholesterolaemia: A single-arm, open-label, phase 3 study. Lancet 2013, 381, 40–46. [Google Scholar] [CrossRef] [PubMed]
  173. Franchini, M.; Capuzzo, E.; Liumbruno, G.M. Lipoprotein apheresis for the treatment of elevated circulating levels of lipoprotein(a): A critical literature review. Blood Transfus. 2016, 14, 413–418. [Google Scholar] [CrossRef]
  174. Thompson, G.R.; Group, H.-U.L.A.W. Recommendations for the use of LDL apheresis. Atherosclerosis 2008, 198, 247–255. [Google Scholar] [CrossRef] [PubMed]
  175. Padmanabhan, A.; Connelly-Smith, L.; Aqui, N.; Balogun, R.A.; Klingel, R.; Meyer, E.; Pham, H.P.; Schneiderman, J.; Witt, V.; Wu, Y.; et al. Guidelines on the Use of Therapeutic Apheresis in Clinical Practice—Evidence-Based Approach from the Writing Committee of the American Society for Apheresis: The Eighth Special Issue. J. Clin. Apher. 2019, 34, 171–354. [Google Scholar] [CrossRef] [PubMed]
  176. Cegla, J.; Neely, R.D.G.; France, M.; Ferns, G.; Byrne, C.D.; Halcox, J.; Datta, D.; Capps, N.; Shoulders, C.; Qureshi, N.; et al. HEART UK consensus statement on Lipoprotein(a): A call to action. Atherosclerosis 2019, 291, 62–70. [Google Scholar] [CrossRef]
  177. Schettler, V.J.J.; Neumann, C.L.; Peter, C.; Zimmermann, T.; Julius, U.; Hohenstein, B.; Roeseler, E.; Heigl, F.; Grutzmacher, P.; Blume, H.; et al. Lipoprotein apheresis is an optimal therapeutic option to reduce increased Lp(a) levels. Clin. Res. Cardiol. Suppl. 2019, 14, 33–38. [Google Scholar] [CrossRef]
  178. Julius, U.; Tselmin, S.; Schatz, U.; Fischer, S.; Birkenfeld, A.L.; Bornstein, S.R. Actual situation of lipoprotein apheresis in patients with elevated lipoprotein(a) levels. Atheroscler. Suppl. 2019, 40, 1–7. [Google Scholar] [CrossRef]
  179. Jaeger, B.R.; Richter, Y.; Nagel, D.; Heigl, F.; Vogt, A.; Roeseler, E.; Parhofer, K.; Ramlow, W.; Koch, M.; Utermann, G.; et al. 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, 229–239. [Google Scholar] [CrossRef]
  180. Roeseler, E.; Julius, U.; Heigl, F.; Spitthoever, R.; Heutling, D.; Breitenberger, P.; Leebmann, J.; Lehmacher, W.; Kamstrup, P.R.; Nordestgaard, B.G.; et al. Lipoprotein Apheresis for Lipoprotein(a)-Associated Cardiovascular Disease: Prospective 5 Years of Follow-Up and Apolipoprotein(a) Characterization. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 2019–2027. [Google Scholar] [CrossRef]
  181. Stefanutti, C.; D’Alessandri, G.; Russi, G.; De Silvestro, G.; Zenti, M.G.; Marson, P.; Belotherkovsky, D.; Vivenzio, A.; Di Giacomo, S. Treatment of symptomatic HyperLp(a)lipoproteinemia with LDL-apheresis: A multicentre study. Atheroscler. Suppl. 2009, 10, 89–94. [Google Scholar] [CrossRef] [PubMed]
  182. Bigazzi, F.; Sbrana, F.; Berretti, D.; Maria Grazia, Z.; Zambon, S.; Fabris, A.; Fonda, M.; Vigna, G.B.; D’Alessandri, G.; Passalacqua, S.; et al. Reduced incidence of cardiovascular events in hyper-Lp(a) patients on lipoprotein apheresis. The G.I.L.A. (Gruppo Interdisciplinare Aferesi Lipoproteica) pilot study. Transfus. Apher. Sci. 2018, 57, 661–664. [Google Scholar] [CrossRef] [PubMed]
  183. Tsimikas, S. A Test in Context: Lipoprotein(a): Diagnosis, Prognosis, Controversies, and Emerging Therapies. J. Am. Coll. Cardiol. 2017, 69, 692–711. [Google Scholar] [CrossRef] [PubMed]
  184. Tsimikas, S.; Viney, N.J.; Hughes, S.G.; Singleton, W.; Graham, M.J.; Baker, B.F.; Burkey, J.L.; Yang, Q.; Marcovina, S.M.; Geary, R.S.; et al. Antisense therapy targeting apolipoprotein(a): A randomised, double-blind, placebo-controlled phase 1 study. Lancet 2015, 386, 1472–1483. [Google Scholar] [CrossRef]
  185. Jang, A.Y.; Lim, S.; Jo, S.H.; Han, S.H.; Koh, K.K. New Trends in Dyslipidemia Treatment. Circ. J. 2021, 85, 759–768. [Google Scholar] [CrossRef]
  186. Graham, M.J.; Viney, N.; Crooke, R.M.; Tsimikas, S. Antisense inhibition of apolipoprotein (a) to lower plasma lipoprotein (a) levels in humans. J. Lipid Res. 2016, 57, 340–351. [Google Scholar] [CrossRef]
  187. Viney, N.J.; van Capelleveen, J.C.; Geary, R.S.; Xia, S.; Tami, J.A.; Yu, R.Z.; Marcovina, S.M.; Hughes, S.G.; Graham, M.J.; Crooke, R.M.; 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, 2239–2253. [Google Scholar] [CrossRef]
  188. Tsimikas, S.; Karwatowska-Prokopczuk, E.; Gouni-Berthold, I.; Tardif, J.C.; Baum, S.J.; Steinhagen-Thiessen, E.; Shapiro, M.D.; Stroes, E.S.; Moriarty, P.M.; Nordestgaard, B.G.; et al. Lipoprotein(a) Reduction in Persons with Cardiovascular Disease. N. Engl. J. Med. 2020, 382, 244–255. [Google Scholar] [CrossRef]
  189. Yeang, C.; Karwatowska-Prokopczuk, E.; Su, F.; Dinh, B.; Xia, S.; Witztum, J.L.; Tsimikas, S. Effect of Pelacarsen on Lipoprotein(a) Cholesterol and Corrected Low-Density Lipoprotein Cholesterol. J. Am. Coll. Cardiol. 2022, 79, 1035–1046. [Google Scholar] [CrossRef]
  190. Korneva, V.A.; Kuznetsova, T.Y.; Julius, U. Modern Approaches to Lower Lipoprotein(a) Concentrations and Consequences for Cardiovascular Diseases. Biomedicines 2021, 9, 1271. [Google Scholar] [CrossRef]
  191. Alshaer, W.; Zureigat, H.; Al Karaki, A.; Al-Kadash, A.; Gharaibeh, L.; Hatmal, M.M.; Aljabali, A.A.A.; Awidi, A. siRNA: Mechanism of action, challenges, and therapeutic approaches. Eur. J. Pharmacol. 2021, 905, 174178. [Google Scholar] [CrossRef] [PubMed]
  192. Nurmohamed, N.S.; Kraaijenhof, J.M.; Stroes, E.S.G. Lp(a): A New Pathway to Target? Curr. Atheroscler. Rep. 2022, 24, 831–838. [Google Scholar] [CrossRef] [PubMed]
  193. Koren, M.J.; Moriarty, P.M.; Baum, S.J.; Neutel, J.; Hernandez-Illas, M.; Weintraub, H.S.; Florio, M.; Kassahun, H.; Melquist, S.; Varrieur, T.; et al. Preclinical development and phase 1 trial of a novel siRNA targeting lipoprotein(a). Nat. Med. 2022, 28, 96–103. [Google Scholar] [CrossRef] [PubMed]
  194. O’Donoghue, M.L.; Rosenson, R.S.; Gencer, B.; Lopez, J.A.G.; Lepor, N.E.; Baum, S.J.; Stout, E.; Gaudet, D.; Knusel, B.; Kuder, J.F.; et al. Small Interfering RNA to Reduce Lipoprotein(a) in Cardiovascular Disease. N. Engl. J. Med. 2022, 387, 1855–1864. [Google Scholar] [CrossRef] [PubMed]
  195. O’Donoghue, M.L.; JA, G.L.; Knusel, B.; Gencer, B.; Wang, H.; Wu, Y.; Kassahun, H.; Sabatine, M.S. Study design and rationale for the Olpasiran trials of Cardiovascular Events And lipoproteiN(a) reduction-DOSE finding study (OCEAN(a)-DOSE). Am. Heart J. 2022, 251, 61–69. [Google Scholar] [CrossRef] [PubMed]
  196. Sohn, W.; Winkle, P.; Neutel, J.; Wu, Y.; Jabari, F.; Terrio, C.; Varrieur, T.; Wang, J.; Hellawell, J. Pharmacokinetics, Pharmacodynamics, and Tolerability of Olpasiran in Healthy Japanese and Non-Japanese Participants: Results from a Phase I, Single-dose, Open-label Study. Clin. Ther. 2022, 44, 1237–1247. [Google Scholar] [CrossRef]
  197. Rider, D.A.; Eisermann, M.; Loffler, K.; Aleku, M.; Swerdlow, D.I.; Dames, S.; Hauptmann, J.; Morrison, E.; Lindholm, M.W.; Schubert, S.; et al. Pre-clinical assessment of SLN360, a novel siRNA targeting LPA, developed to address elevated lipoprotein (a) in cardiovascular disease. Atherosclerosis 2022, 349, 240–247. [Google Scholar] [CrossRef]
  198. Nissen, S.E.; Wolski, K.; Balog, C.; Swerdlow, D.I.; Scrimgeour, A.C.; Rambaran, C.; Wilson, R.J.; Boyce, M.; Ray, K.K.; Cho, L.; et al. Single Ascending Dose Study of a Short Interfering RNA Targeting Lipoprotein(a) Production in Individuals With Elevated Plasma Lipoprotein(a) Levels. JAMA 2022, 327, 1679–1687. [Google Scholar] [CrossRef]
Figure 1. Structure and composition of Lp(a). Lp(a) consists of a low-density lipoprotein to which Apo(a) is added, forming a disulfide bridge between Apo B100 and Apo(a). The protein structure of Apo(a) consists of numerous functional domains. Taken as a whole, it closely resembles the structure of plasminogen, a proenzyme whose conversion into plasmin determines the activation of thrombolytic processes. Plasminogen also exhibits a series of kringle (K) structures, numbered I through V, followed by a serine protease-like catalytic domain that can be cleaved via tissue plasminogen activator and urokinase, thus generating plasmin. In apo(a), some domains closely resemble the analogous domains of plasminogen, including the carboxy-terminal catalytic protein domain which, however, cannot be activated as in plasminogen. The peculiar element of apo(a) is the high number of repetitions of kringle IV; however, these repetitions do not give rise to identical structures, but to subtypes of kringle IV, from type 1 to type 10. All of the subtypes are present in a single copy except for kringle IV type 2, whose repetitions are responsible for the length polymorphism of Apo(a).
Figure 1. Structure and composition of Lp(a). Lp(a) consists of a low-density lipoprotein to which Apo(a) is added, forming a disulfide bridge between Apo B100 and Apo(a). The protein structure of Apo(a) consists of numerous functional domains. Taken as a whole, it closely resembles the structure of plasminogen, a proenzyme whose conversion into plasmin determines the activation of thrombolytic processes. Plasminogen also exhibits a series of kringle (K) structures, numbered I through V, followed by a serine protease-like catalytic domain that can be cleaved via tissue plasminogen activator and urokinase, thus generating plasmin. In apo(a), some domains closely resemble the analogous domains of plasminogen, including the carboxy-terminal catalytic protein domain which, however, cannot be activated as in plasminogen. The peculiar element of apo(a) is the high number of repetitions of kringle IV; however, these repetitions do not give rise to identical structures, but to subtypes of kringle IV, from type 1 to type 10. All of the subtypes are present in a single copy except for kringle IV type 2, whose repetitions are responsible for the length polymorphism of Apo(a).
Ijerph 20 06721 g001
Figure 2. Association between cardiovascular diseases, mortality, and high lipoprotein(a) plasma levels from the Copenhagen studies.
Figure 2. Association between cardiovascular diseases, mortality, and high lipoprotein(a) plasma levels from the Copenhagen studies.
Ijerph 20 06721 g002
Table 1. Appraised population presenting a lipoprotein(a) plasma concentration >50 mg/dL or >125 nmol/L.
Table 1. Appraised population presenting a lipoprotein(a) plasma concentration >50 mg/dL or >125 nmol/L.
World AreaPrevalence (%)Prevalence
Asia/China10%261 million
Latin America13%97 million
Europe20%148 million
Australia20%8 million
North America20%73 million
South Asia25%469 million
Africa30%376 million
Global10 to 30%1.43 billion
Table 3. Clinical trials of RNA targeted therapies to reduce Lp(a).
Table 3. Clinical trials of RNA targeted therapies to reduce Lp(a).
Clinical Trial NumberNCT03070782NCT03626662NCT04270760NCT04606602
Phase of studyPhase 2Phase 1 Phase 2Phase 1
Patient population2866428132
Tested therapyPelacarsen
(AKCEA-APO(a)-LRx)
OLp(a)siranOLp(a)siranSLN360
Partecipant (drug/placebo)239/4748/16 (cohort 1-7)227/5424/8
Baseline therapy80 to 90% of the patients received statin therapy, 50% received ezetimibe, and 20% received a PCSK9 inhibitorIn cohorts 1–5, no participants were on statins; in cohorts 6 and 7, 67% were on statins88% took statin therapy (including 61% taking high-intensity statin therapy), 52% ezetimibe, and 23% (PCSK9) inhibitorConcomitant satin use: P: 63%, 30 mg SLN360: 0, 100 mg SLN360: 33%, 300 mg SLN360: 50%, 600 mg SLN360: 60%
OutcomesThe percent change in the lipoprotein(a) level from the baseline to the primary analysis time point at 6 months of exposure (week 25 or week 27)Safety and tolerability/ change in Lp(a) concentrationPercent change in the lipoprotein(a) concentration from the baseline to week 36 and at week 48Safety and tolerability/
change in Lp(a) concentration
Dose20 mg every 4 weeks, 40 mg every 4 weeks, 60 mg every 4 weeks, 20 mg every 2 weeks, or 20 mg every week, or a physiologic saline placeboCohort 1: 3 mg sd (n = 6), cohort 2: 9 mg sd (n = 6), cohort 3: 30 mg sd (n = 6), cohort 4: 75 mg sd (n = 6), cohort 5: 225 mg sd (n = 6), cohort 6: 9 mg sd (n = 9), and cohort 7: 75 mg sd (n = 9)10 mg every 12 weeks, 75 mg every 12 weeks, 225 mg every 12 weeks, or 225 mg every 24 weeks30 mg sd (n = 6), 100 mg sd(n = 6), 300 mg sd (n = 6), and 600 mg sd (n = 6)
Baseline concentration of Lp(a)Median levels ranged from 205 to 247 nmol/L70–199 nM (cohorts 1–5), ≥200 nM (cohorts 6 and 7)Median (nmol/L): 260.3
(interquartile range of 198.1 to 352.4)
Median (nmol/L): P 238, 30 mg 171, 100 mg 217, 300 mg 285, 600 mg 231
Percent change from baseline Lp(a)Decreases of 35% at a dose of 20 mg every 4 weeks, 56% at 40 mg every 4 weeks, 58% at 20 mg every 2 weeks, 72% at 60 mg every 4 weeks, and 80% at 20 mg every week, as compared with 6% for a pooled placebo groupFrom −71% to −97% in cohorts 1–5 and from −76% to −91% in cohorts 6 and 7At 36 weeks: + 3.6% in the placebo group, −70.5% with the 10 mg dose every 12 weeks, −97.4% with the 75 mg dose every 12 weeks, −101.1% with the 225 mg dose every 12 weeks, and −100.5% with the 225 mg dose every 24 weeks. At 48 weeks: −68.5% with the 10 mg dose every 12 weeks, −96.1% with the 75 mg dose every 12 weeks, −100.9% with the 225 mg dose every 12 weeks, and −85.9% with the 225 mg dose every 24 weeksP: −10%, 30 mg: −46%, 100 mg: −86%, 300 mg: −96%, 600 mg: −98%
P, placebo; Sd, single dose.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vinci, P.; Di Girolamo, F.G.; Panizon, E.; Tosoni, L.M.; Cerrato, C.; Pellicori, F.; Altamura, N.; Pirulli, A.; Zaccari, M.; Biasinutto, C.; et al. Lipoprotein(a) as a Risk Factor for Cardiovascular Diseases: Pathophysiology and Treatment Perspectives. Int. J. Environ. Res. Public Health 2023, 20, 6721. https://doi.org/10.3390/ijerph20186721

AMA Style

Vinci P, Di Girolamo FG, Panizon E, Tosoni LM, Cerrato C, Pellicori F, Altamura N, Pirulli A, Zaccari M, Biasinutto C, et al. Lipoprotein(a) as a Risk Factor for Cardiovascular Diseases: Pathophysiology and Treatment Perspectives. International Journal of Environmental Research and Public Health. 2023; 20(18):6721. https://doi.org/10.3390/ijerph20186721

Chicago/Turabian Style

Vinci, Pierandrea, Filippo Giorgio Di Girolamo, Emiliano Panizon, Letizia Maria Tosoni, Carla Cerrato, Federica Pellicori, Nicola Altamura, Alessia Pirulli, Michele Zaccari, Chiara Biasinutto, and et al. 2023. "Lipoprotein(a) as a Risk Factor for Cardiovascular Diseases: Pathophysiology and Treatment Perspectives" International Journal of Environmental Research and Public Health 20, no. 18: 6721. https://doi.org/10.3390/ijerph20186721

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop