Abstract
High levels of lipoprotein(a) [Lp(a)] are causal for development of atherosclerotic cardiovascular disease and highly regulated by genetics. Levels are higher in Blacks compared to Whites, and in women compared to men. Lp(a)’s main protein components are apolipoprotein (apo) (a) and apoB100, the latter being the main component of Low-Density Lipoprotein (LDL) particles. Studies have identified Lp(a) to be associated with inflammatory, coagulation and wound healing pathways. Lack of validated and accepted assays to measure Lp(a), risk cutoff values, guidelines for diagnosis, and targeted therapies have added challenges to the field. Scientific efforts are ongoing to address these, including studies evaluating the cardiovascular benefits of decreasing Lp(a) levels with targeted apo(a) lowering treatments. This review will provide a synopsis of evidence-based effects of high Lp(a) on disease presentation, highlight available guidelines and discuss promising therapies in development. We will conclude with current clinical information and future research needs in the field.
Keywords: Lipoprotein(a), atherosclerotic cardiovascular disease (ASCVD), aortic valve stenosis, inflammation, lipid lowering therapies, Lp(a) antisense
What is lipoprotein (a)?
Lipoprotein (a) [Lp(a)] is a plasma lipid particle containing two main apolipoproteins: apolipoprotein B100 [apoB] and apolipoprotein(a) [apo(a)] [1]. It was first discovered by Kare Berg in 1963 [2]. Current evidence supports that the assembly of the Lp(a) particle occurs within the liver, including the binding of apo(a) to apoB via a disulfide bond [3]. Most individuals produce two isoforms of apo(a), as determined by the number of Kringle-IV type 2 (KIV-2) repeats, which are highly regulated by the LPA gene [4,5]. The location of the LPA gene is in close proximity to the plasminogen gene and the latter has a high degree of homology with apo(a) [6]. The proteome of isolated Lp(a) particles has been described [7] and pathway analysis of the expressed proteins link Lp(a) to wound healing, lipoprotein metabolism and complement activation. The specific role of these pathways in cardiovascular disease development and progression due to high Lp(a) levels is yet to be determined (Fig. 1).
Fig. 1.

Lipoprotein(a) current risk (A) and clinical considerations (B).
Metabolic regulation of circulating lipoprotein(a) levels: synthesis, production, and clearance
The level of circulating plasma lipoproteins is regulated by their production and clearance and, the specific pathways responsible for Lp(a) processing are still being investigated [8]. The production of Lp(a) is highly affected by the number of KIV-2 repeats and self-reported race/ethnicity [9,10]. It has been shown that levels of Lp(a) are higher in Blacks and Hispanics compared to Whites. Additionally, results from studies using low density lipoprotein cholesterol (LDL-C) and apoB lowering therapies, show that Lp(a) plasma levels decrease due combination of decreases in production and increases in the clearance of Lp(a). [11–13]. A study, where subjects received Anacetrapib, a Cholesteryl Ester Transfer Protein (CETP) inhibitor, showed that the drug lowered plasma Lp(a) levels by decreasing production [11]. Similar results were observed after apoB antisense treatment [12]. However, studies with proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor treatment, which works by upregulating the LDL receptor (LDL-R), lowered Lp(a) level in patients by increasing both fractional clearance rate and reducing production rate [13,14]. This might suggest that the LDL-R also participates in the catabolism of Lp (a) under certain conditions. There have been reports of Lp(a) being found in urine, suggesting that kidneys play a role in its excretion [15,16].
Lipoprotein(a): genetics and clinical perspective
Lp(a) levels are primarily genetically regulated [17]. The size of the apo(a) is determined by the KIV-2 copy number and is inversely related to the Lp(a) plasma levels. This copy number variance may account for the large variability (19–69%) of interindividual Lp(a) levels [18]. Genetic relationships with APOE ε2 allele have been linked to a small decreases in levels of Lp(a) [19]. Lastly, the work of various investigators [20–22], and most recent publications from Dr. Kronenberg’ s group [23], have described a large amount of single nucleotide polymorphisms (SNPs) in the LPA locus that are strongly associated with Lp(a) concentrations. The relationships of other gene associations are being investigated as reviewed in the recent Lp(a) consensus statement from the European Atherosclerosis Society (EAS) [24]. The clinical relevance of these associations is still to be confirmed. In the era of increase personalized medicine efforts, it is important to note the relationship of these common SNPs with specific patient populations. We highlight these in Table 1.
Table 1.
Genetic variations of the LPA gene by race/ethnicity.
| SNP | Most prevalent populations |
|---|---|
| rs3798220 | Hispanics (42.38%) |
| rs9457951 | Blacks (32.92%) |
| rs10455872 | Whites (14.27%) |
There are associations between Lp(a) levels and ASCVD risk among different ethnicities. These racial differences affect the heritability of apo(a), with African-Americans having a lower heritability when compared to Caucasian populations [25]. In 2021, a study utilizing data from the UK Biobank highlighted higher levels of Lp(a) in Blacks compare to other racial groups [26]. Although Hispanics have not been included in these reports, Lp(a) levels have been examined in the following studies: Atherosclerosis Risk in Communities (ARIC) [27–29], Multi-Ethnic Study of Atherosclerosis (MESA) [30], the Dallas Heart Study (DHS) [10] and South Asians [31] living in the US. In all studies and racial/ethnic groups, high Lp(a) levels were linked to an increase in ASCVD. Currently, there are no guidelines for risk stratification based on race and/or sex.
A review of literature from the last decade shows that only about 25% of the studies were done in diverse populations compared to only White cohorts. Of the studies that do examine Lp(a) concentrations in underrepresented groups, the population sample sizes are much lower than sample sizes available in Whites. While larger sample sizes will not eliminate disparity, greater precision for serum levels per race/ethnicity may be achieved. Clinicians often use race as a valuable predictor for cardiovascular risk. More robust analyses of Lp(a) levels by race may yield a more quantifiable basis for cardiovascular risk stratification [32,33].
Lp(a) levels have been found to be affected by non-genetic influences such as diet, hormonal regulation, kidney disease, and chronic inflammation as mentioned in Table 2. These factors may be related to high Lp(a) levels and increased risk of disease and have been recently reviewed [24]. Briefly, Lp(a) levels have been found to be lower in individuals consuming high saturated diets as recently reviewed by Enkhmaa B et al. [34]. These diet effects have also been seen in past studies (the Delta study) [35] and in a more recent randomized diet study [36]. Mechanisms regulating levels of apoB containing particles upon consumption of a high saturated fat diet need to be further investigated given that Lp(a) contains this protein. Possibly, there could be a change in the available pool of liver apoB that binds to apo(a) during consumption of high saturated fat content, causing lower Lp(a) particles to be assembled and secreted into the circulation.
Table 2.
Non-genetic influences on Lp(a) concentration.
| Influence | Increases Lp (a) | Decreases Lp (a) |
|---|---|---|
| Diet [34,36] | High carbohydrate and unsaturated fat diet | Low carbohydrate and high saturated fat diet |
| Hormonal changes [87–90] | Hypothyroidism Growth hormone Pregnancy |
Hyperthyroidism Post-menopausal hormone replacement therapy |
| Renal disease [91] | Dialysis dependency Nephrotic syndrome |
|
| Chronic inflammation [92] | Rheumatoid arthritis Systemic lupus erythematosus (SLE) Antiphospholipid syndrome (APS) |
|
Early reports by Mora et al. found an increase in the incidence of Type 2 Diabetes Mellitus in individuals with low Lp(a) levels [37]. These reports have been duplicated, and a meta-analysis within the recent EAS statement of all studies showed a 38% (95% CI 29–48%, P < 0.0001) higher risk for the bottom quintile (thresholds < 3–5 mg/dL) vs. top quintile of Lp(a) (thresholds > 27–55 mg/dL) with no significant heterogeneity across studies. Ongoing treatment development programs using targeted apo(a) lowering therapies to improve cardiovascular outcomes have not reported signals for increases in insulin resistance or glucose levels [38,39]. Importantly, Lp(a) assembly is linked to apoB availability in the liver, and apoB is highly regulated by insulin signaling. Further investigation into these mechanisms may provide some insight into the increase in insulin resistance with lower Lp(a) levels observed in some individuals.
Lp(a) and ASCVD and aortic valve stenosis
Lp(a) particles have been identified in human atheroma dissections [40,41], indicating it’s involvement in atherogenic mechanisms. In addition, genetic studies have provided convincing evidence that LPA is associated causally with coronary heart disease (CHD) and the development of aortic stenosis [22,42–45]. There is data supporting an association between high Lp(a) levels with myocardial infarction [46–48], aortic valve disease [49,50], stroke [51], peripheral arterial disease (PAD) and increased cardiovascular mortality [26,52,53].
Recent studies from the UK Biobank showed a linear rise in ASCVD risk with increasing Lp(a) concentrations, and specifically, these observations were seen across all racial groups [26]. More recent findings from MESA and DHS cohorts found that Lp(a) and coronary artery calcium (CAC) score were independently associated with ASCVD risk [30].
High levels of Lp(a) have also been associated with increased incidence, progression, and post-treatment recurrence of PAD [54,55]. This relationship may be driven by migration of small Lp(a) particles into the subendothelial space [56]. Lp(a) has also been shown to compete for binding of plasminogen and plasmin, generating a prothrombotic state [57]. A study published in COVID-19 patients, noted that thrombotic activity marked by elevated D-dimer was associated with high Lp(a) levels [58]. To this effort, a recent genomic analysis of the Aspirin in Reducing Events in the Elderly (ASPREE) trial revealed data which supports a link between high Lp(a) and a prothrombotic state [59]. This study used previously described Lp(a) genotypes [20,21] linked to high Lp(a) levels (rs3798220-C) and a polygenic Lp(a) risk, rather than a phenotype, as a predictor to examine the benefits of aspirin treatment. They provided data that support beneficial effects of low-dose aspirin in this genotyped high Lp(a) group [59]. However, the authors state that in clinical practice, the benefits of aspirin must be balanced with bleeding risks, which increase with age and other factors. Additional data using randomized and well-pheno-typed patient populations are needed to confirm these findings.
Lipoprotein(a) links to inflammatory pathways
Inflammatory pathways have been linked to the development and outcomes of ASCVD. Importantly, the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) for the first time provided conclusive evidence that inflammation enhances cardiovascular disease (CVD) in humans [60]. Inter-leukins (IL-6 and IL-18), markers of the inflammatory state, are associated with CVD risk in humans [61–63], and use of Tocilizumab, an IL-6 antagonist, resulted in decreased serum Lp(a) concentrations [64]. Moreover, a post-hoc analysis of the Assessment of Clinical Effects of Cholesteryl Ester Transfer Protein Inhibition with Evacetrapib in Patients at a High Risk for Vascular Outcomes (ACCELERATE) trial, in patients with established vascular disease who were optimally treated, found that increased Lp(a) levels during treatment were significantly associated with cardiovascular death, myocardial infarction, and stroke only in individuals with high-sensitivity C-reactive protein (hsCRP) levels of 2 mg/L or higher during treatment, but not in those with levels less than 2 mg/L [65]. A more recent, large study using the MESA cohort assessed whether Lp(a)-associated ASCVD risk was modified by hsCRP in a primary prevention setting [66]. The investigators found a significant interaction between Lp(a) and hsCRP (p = 0.04). With hsCRP < 2 mg/L, no significant CVD risk was observed at any level of Lp(a) from < 50 mg/dL to > 100 mg/dL. However, with hsCRP ≥ 2 mg/L, a significant CVD risk was observed with Lp(a) of 50–99.9 mg/dL (HR: 1.36; 95% CI: 1.02–1.81) and Lp(a) ≥ 100 mg/dL (HR: 2.09; 95% CI: 1.40–3.13). Importantly, in this primary prevention cohort, isolated elevations of Lp(a) or hsCRP were not associated with increased CVD risk. However, the combination of elevated Lp(a) (≥50 mg/dL) and hsCRP (≥2 mg/L) is associated with significant CVD risk and all-cause mortality. The authors concluded that individuals with both high Lp(a) and CRP levels have greater ASCVD risk and all-cause mortality, and thus may merit closer surveillance and more aggressive ASCVD risk management. Reports from a retrospective study in COVID-19 patients, found that elevated inflammatory biomarkers, including IL-6, CRP, and procalcitonin were associated with lower Lp(a). However, elevated Lp(a) was not associated with rate of hospital death or discharge [58].
Lastly, there is strong evidence classifying oxidized phospholipids (OxPL) as a pro-atherogenic biomarkers, and Lp(a) is identified as the preferential carrier of OxPL [67,68]. These pathways of inflammation and Lp(a) have been recently reviewed [68]. Importantly, mice and humans have been used to describe the role of cholesterol and OxPL within the isolated Lp(a) particles, establishing that binding of OxPL and Lp(a) promotes atherosclerosis [69].
Measuring Lp(a) in patients: how and when?
Despite the lack of standardized assays, the data on Lp(a)’s causal link to ASCVD and multiple statements and guidelines have encouraged clinicians to measure Lp(a) levels at least once in a lifetime [53]. Although temporal variabilities in Lp(a) levels have been reported during a lifetime [70], these are not significant for individuals to be classified as having high levels in one measurement, and later in life finding these levels to be low with a second measurement. However, multiple measurements may help with risk assessment. Importantly, there are no established cut-offs that define low vs. high levels. The lack of standardization of the available assays prevents comparison of studies and a true measure of the population distribution and risk assessment based on Lp(a) levels [71–73].
Measurements of Lp(a) should be obtained in nmol/L via an assay that is isoform independent and is traceable to calibrators that are internationally accepted [53].
Various organizations have provided statements and guidelines to assist with raising awareness of Lp (a) as a causal risk for ASCVD, Table 3. These efforts have assisted clinicians with understanding the available evidence to use in clinical practice. In 2018, the NHLBI gathered a group of experts and published a statement to help the Lp(a) field move forward [74]. The experts outlined the top areas for research in the field: to define the mechanisms of Lp(a) synthesis, assembly, and clearance; to understand the mechanisms linking Lp(a) to oxidized phospholipids and inflammation that are driving ASCVD; to standardize Lp(a) measurements; to define population risk levels; to understand racial and ethnic differences in levels; and to understand the effects of lowering Lp(a) levels on cardiovascular disease outcomes. Soon after various organizations followed with their own statements [24,53,75] and guideline reports [76–78].
Table 3.
Highlights of global statements and guidelines.
| Year | Publication Team | Highlight |
|---|---|---|
| 2018 | National Heart, Blood and Lung Institute (NHLBI) Workgroup on Lipoprotein(a) [93] | Outline of top areas for research and future directions to optimize Lp(a) risk assessment. |
| 2019 | National Lipid Association (NLA) Lipid Association Statement [94] | Use of Lipoprotein(a) in clinical practice. |
| 2019 | AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/ APhA/ASPC/NLA/PCNA. 2018 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 [75] | Guidance for and use of Lp(a) as a risk enhancer. |
| 2019 | European Society of Cardiology (ESC)/EAS Guidelines for the management of dyslipidemias: Lipid modification to reduce cardiovascular risk Atherosclerosis [95] | Guidelines suggesting once in a lifetime measurement of Lp(a). |
| 2020 | ESC Scientific Document Group. 2019 ESC/EAS Guidelines for the management of dyslipidemias: lipid modification to reduce cardiovascular risk [96] | Guidelines suggesting once in a lifetime measurement of Lp(a). |
| 2020 | Endocrine Society Clinical Practice Guidelines [97] | Guidelines suggesting once in a lifetime measurement of Lp(a) without repeating it later in life. |
| 2021 | Canadian Cardiovascular Society Guidelines for the Management of Dyslipidemia for the Prevention of Cardiovascular Disease in the Adult [76] | Guidelines suggesting measuring Lp(a) two times in a lifetime. |
| 2022 | AHA Lp(a) statement [53] | Rationale for targeted research efforts that can provide clinical direction for risk reduction. Encouragement for screening strategies and the need for further mechanistic and biological studies of Lp(a). |
| 2022 | EAS Scientific Statement on Lp(a) [24] | Updates on evidence for the role of Lp(a) in atherosclerotic cardiovascular disease (ASCVD) and Aortic Valve Stenosis. Clinical guidance for testing and treating elevated Lp(a) levels, and consideration of its inclusion in global risk estimation. |
The latest guidelines from the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines suggest using Lp(a) levels as a risk enhancer [75]. They also indicate that relative indications for Lp(a) measurement are a family history of premature ASCVD (< 55 years of age in men; < 65 years of age in women), personal history of ASCVD not explained by major risk factors, primary severe hypercholesterolemia (LDL-C ≥190 mg/dL), or suspected familial hypercholesterolemia. It is important to recognize that patients who have a family history of elevated Lp(a) or premature ASCVD may not themselves have clinical signs of cardiovascular disease. Since 2018, various reports have aided in further interpretation of the findings and suggestions, with the most recent statement from the European Atherosclerosis Society [24], including new analysis of existing data (meta-analysis) and updates on the effects of race/ethnicity on Lp(a) levels. The authors agree with other efforts that measurement of Lp(a) should be done at least once in adults, and that a high Lp(a) concentration should be interpreted in the context of other risk factors, including absolute global cardiovascular risk, and addressed through intensified lifestyle and risk factor management. Table 4 provides an overview for using Lp(a) levels in clinical practice.
Table 4.
Lp(a) in Clinical Practice.
| Relative AHA indications | ASCVD presentation | Clinical considerations |
|---|---|---|
|
|
|
What to do when Lp(a) is high?
The current guidelines on management of Lp(a) levels suggest lowering all other risk factors [53,76]. In the published statement on Lp(a) from the American Heart Association (AHA), authors have provided user friendly tables to assess risk, these can aid clinicians to assess additional cardiovascular risk factors, including Lp(a) levels when/if available [53]. Some therapies that lower apoB and LDL-C also decrease Lp (a) modestly, such as niacin, CETP inhibitors, ApoB antisense. PSCK9 inhibitor showed a 25% reduction in Lp(a), and after one year of treatment reduced cardiovascular event rate. However, that could be due to the combined effect of lowering LDL-C with Lp(a). Although plasma LDL-C lowering agents, such as statins, are widely used, and have been proven to decrease ASCVD [79] in many populations, statins do not lower Lp(a) levels. In fact, they may increase Lp(a) levels [80]. Importantly, in patients with elevated LDL-C despite statin therapy, a substantial fraction of cholesterol may be carried by Lp(a) rather than LDL particles. However, the role of high or low cholesterol carried by Lp(a) particles and its link to cardiovascular disease and/or benefit has not been established.
The FDA approved lipoprotein apheresis for specific cases at significantly increased risk [81,82]. In some countries, apheresis is standard of care for individuals with high Lp(a) levels [81,83]. A list of current and new programs to lower Lp(a) is listed on Table 5. Various publications on the effects of apheresis in individuals show a benefit [84]. However, more rigorous randomized control studies are needed in this area. Diets rich in saturated fats have been linked to lower Lp(a) levels. However, the current data does not support the use of high saturated fats to lower Lp(a) plasma levels. Moreover, current data suggests that all individuals with high levels of Lp(a) should follow a heart healthy diet and lifestyle modification to lower all other cardiovascular risk factors [24,53,75].
Table 5.
Current management of high Lp(a).
| A. Lowering all other risk factors with available therapies including lifestyle modifications |
| B. Lp(a) apheresis FDA approved for four categories: |
| 1. Functional Hypercholesterolemic Homozygotes with LDL-C > 500 mg/dL |
| 2. Functional Hypercholesterolemic Heterozygotes with LDL-C ≥ 300 mg/dL |
| 3. Functional Hypercholesterolemic Heterozygotes with LDL-C ≥ 100 mg/dL and either documented coronary artery disease or documented peripheral artery disease |
| 4. Lp(a) > 60 mg/dL and LDL-C > 100 mg/dL and with either documented coronary artery disease or documented peripheral artery disease. |
| C. Small interfering RNA (siRNA) and antisense technology are in Phase III of development |
Drugs in development that lower Lp(a)
Over the last 5 years, there has been an increased interest in understanding Lp(a) disease presentation and its link to development of ASCVD. This has been driven by the investments being made in novel therapies that target apo(a) synthesis, using small interfering RNA (siRNA) and antisense technology in the liver, thereby lowering the production of Lp(a) [53]. Completed studies in these programs observed reductions in Lp(a) plasma levels between 71% and 97% with olpasiran, a N-acetylgalactosamine (GalNAc)-conjugated siRNA; and similar changes with the GalNAc-conjugated siRNA (SLN360, Silence Therapeutics) [85] (NCT05581303). Pelacarsen, an antisense oligonucleotide, reduced Lp(a) levels from 72% to 80% [86] (NCT04023552). The cardiovascular outcome trial with Pelacarsen is anticipated to conclude in 2025, providing long awaited results in patients with established cardiovascular disease (NCT04023552). The safety of these programs continues to be evaluated and additional therapies are also in development (NCT05565742).
Summary
This review described Lp(a), discussed the role of genetics and other factors that affect Lp(a) levels, and its role in cardiovascular disease with the emphasis on clinical implications. There is strong scientific support for measuring Lp(a) in every individual at least once and some suggest measuring it twice. Obtaining a thorough medical history in every patient will ensure correct follow-up and personalized advise from treating physicians. It is clear that in individuals with high Lp(a), current efforts should target lowering all other risk factors that increase ASCVD. Future studies and efforts should focus on understanding the racial/ethnic differences in Lp(a) levels and whether these higher levels in certain populations are linked to higher cardiovascular risk and/or additional mortality. Factors that determine disease presentation in individuals with high Lp(a) should be better defined.
Practice points.
High Lp(a) levels are causal for ASCVD.
Measurements of Lp(a) should be obtained in nmol/L via an assay that is isoform independent and is traceable to calibrators that are internationally accepted. However, physicians should measure Lp(a) with available assays (i.e.when levels are high all assays will report this).
Various National and International Institutions suggest measuring Lp(a) once in a lifetime, Table 4.
Current data suggests that all individuals with high levels of Lp(a) should follow a heart healthy diet and lifestyle modification to lower all other cardiovascular risk factors.
Treatment for individuals with high Lp(a) levels should focus on decreasing all other risk. Current evidence supports the use of Lp(a) levels to assess ASCVD risk.
Statins do not lower Lp(a), other apoB lowering therapies provide Lp(a) lowering.
Research agenda.
Clinical studies are needed to help identify Lp(a) levels cut-offs that can help with risk assessments.
Lp(a) levels are higher in Hispanics, South Asians and Blacks when compared to Whites, the mechanism underlying this is not clear. Whether these populations are at higher risk for development of ASCVD due to high Lp(a) levels is unclear.
Outcome studies from ongoing development programs targeting apo(a), and hence lowering plasma Lp(a) levels, will provide data on cardiovascular benefits.
Acknowledgment
All presented text and figures have been developed by the study authors.
Funding and conflicts of interest
There are no conflicts from the authors for the work presented. The authors receive funding from NIH-NHLBI (R01HL139759, T32HL007343) and industry studies (Amgen, Inc; Kaneka) examining the role of lipid-lowering therapies on lipid metabolic factors.
References
- *[1].Utermann G, Weber W. Protein composition of Lp(a) lipoprotein from human plasma. FEBS Lett 1983;154:357–61. [DOI] [PubMed] [Google Scholar]
- [2].Berg K. A new serum type system in man-the LP system. Acta Pathol Microbiol Scand 1963;59:369–82. [DOI] [PubMed] [Google Scholar]
- [3].Youssef A, Clark JR, Marcovina SM, et al. Apo(a) and ApoB interact noncovalently within hepatocytes: implications for regulation of Lp(a) levels by modulation of ApoB secretion. Arterioscler Thromb Vasc Biol 2022;42:289–304. [DOI] [PubMed] [Google Scholar]
- *[4].Marcovina SM, Albers JJ, Wijsman E, et al. Differences in Lp[a] concentrations and apo[a] polymorphs between black and white Americans. J Lipid Res 1996;37:2569–85. [PubMed] [Google Scholar]
- [5].Gencer B, Kronenberg F, Stroes ES, et al. Lipoprotein(a): the revenant. Eur Heart J 2017;38:1553–60. [DOI] [PubMed] [Google Scholar]
- [6].Frank SL, Klisak I, Sparkes RS, et al. The apolipoprotein(a) gene resides on human chromosome 6q26–27, in close proximity to the homologous gene for plasminogen. Hum Genet 1988;79:352–6. [DOI] [PubMed] [Google Scholar]
- [7].von Zychlinski A, Kleffmann T, Williams MJ, et al. Proteomics of lipoprotein(a) identifies a protein complement associated with response to wounding. J Proteom 2011;74:2881–91. [DOI] [PubMed] [Google Scholar]
- [8].Reyes-Soffer G, Ginsberg HN, Ramakrishnan R. The metabolism of lipoprotein (a): an ever-evolving story. J Lipid Res 2017;58:1756–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Kraft HG, Sandholzer C, Menzel HJ, et al. Apolipoprotein (a) alleles determine lipoprotein (a) particle density and concentration in plasma. Arterioscler Thromb 1992;12:302–6. [DOI] [PubMed] [Google Scholar]
- [10].Pare G, Caku A, McQueen M, et al. Lipoprotein(a) levels and the risk of myocardial infarction among 7 ethnic groups. Circulation 2019;139:1472–82. [DOI] [PubMed] [Google Scholar]
- [11].Thomas T, Zhou H, Karmally W, et al. CETP (Cholesteryl Ester Transfer Protein) inhibition with anacetrapib decreases production of lipoprotein(a) in mildly hypercholesterolemic subjects. Arterioscler Thromb Vasc Biol 2017;37:1770–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Nandakumar R, Matveyenko A, Thomas T, et al. Effects of mipomersen, an apolipoprotein B100 antisense, on lipoprotein (a) metabolism in healthy subjects. J Lipid Res 2018;59:2397–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Reyes-Soffer G, Pavlyha M, Ngai C, et al. Effects of PCSK9 inhibition with alirocumab on lipoprotein metabolism in healthy humans. Circulation 2017;135:352–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Watts GF, Chan DC, Pang J, et al. PCSK9 Inhibition with alirocumab increases the catabolism of lipoprotein(a) particles in statin-treated patients with elevated lipoprotein(a). Metabolism 2020;107:154221. [DOI] [PubMed] [Google Scholar]
- [15].Oida K, Takai H, Maeda H, et al. Apolipoprotein(a) is present in urine and its excretion is decreased in patients with renal failure. Clin Chem 1992;38:2244–8. [PubMed] [Google Scholar]
- [16].Kronenberg F, Trenkwalder E, Lingenhel A, et al. Renovascular arteriovenous differences in Lp[a] plasma concentrations suggest removal of Lp[a] from the renal circulation. J Lipid Res 1997;38:1755–63. [PubMed] [Google Scholar]
- *[17].Kronenberg F, Utermann G. Lipoprotein(a): resurrected by genetics. J Intern Med 2013;273:6–30. [DOI] [PubMed] [Google Scholar]
- [18].Schmidt K, Noureen A, Kronenberg F, et al. Structure, function, and genetics of lipoprotein (a). J Lipid Res 2016;57:1339–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Mack S, Coassin S, Rueedi R, et al. A genome-wide association meta-analysis on lipoprotein (a) concentrations adjusted for apolipoprotein (a) isoforms. J Lipid Res 2017;58:1834–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Shiffman D, O’Meara ES, Bare LA, et al. Association of gene variants with incident myocardial infarction in the Cardiovascular Health Study. Arterioscler Thromb Vasc Biol 2008;28:173–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Luke MM, Kane JP, Liu DM, et al. A polymorphism in the protease-like domain of apolipoprotein(a) is associated with severe coronary artery disease. Arterioscler Thromb Vasc Biol 2007;27:2030–6. [DOI] [PubMed] [Google Scholar]
- *[22].Thanassoulis G, Campbell CY, Owens DS, et al. Genetic associations with valvular calcification and aortic stenosis. N Engl J Med 2013;368:503–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].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. [DOI] [PubMed] [Google Scholar]
- *[24].Kronenberg F, Mora S, Stroes ESG, et al. Lipoprotein(a) in atherosclerotic cardiovascular disease and aortic stenosis: a European Atherosclerosis Society consensus statement. Eur Heart J 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Enkhmaa B, Anuurad E, Zhang W, et al. Heritability of apolipoprotein (a) traits in two-generational African-American and Caucasian families. J Lipid Res 2019;60:1603–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Patel AP, Wang M, Pirruccello JP, et al. Lp(a) (lipoprotein[a]) concentrations and incident atherosclerotic cardiovascular disease: new insights from a large national biobank. Arterioscler Thromb Vasc Biol 2021;41:465–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Schreiner PJ, Heiss G, Tyroler HA, et al. 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;16:471–8. [DOI] [PubMed] [Google Scholar]
- [28].Mehta A, Virani SS, Ayers CR, et al. Lipoprotein(a) and family history predict cardiovascular disease risk. J Am Coll Cardiol 2020;76:781–93. [DOI] [PubMed] [Google Scholar]
- [29].Obisesan OH, Kou M, Wang FM, et al. Lipoprotein(a) and subclinical vascular and valvular calcification on cardiac computed tomography: the atherosclerosis risk in communities study. J Am Heart Assoc 2022;11:e024870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Mehta A, Vasquez N, Ayers CR, et al. Independent association of lipoprotein(a) and coronary artery calcification with atherosclerotic cardiovascular risk. J Am Coll Cardiol 2022;79:757–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Anand SS, Enas EA, Pogue J, et al. Elevated lipoprotein(a) levels in South Asians in North America. Metabolism 1998;47:182–4. [DOI] [PubMed] [Google Scholar]
- [32].Pearson K, Rodriguez F. Lipoprotein(a) and cardiovascular disease prevention across diverse populations. Cardiol Ther 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Guan W, Cao J, Steffen BT, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- *[34].Enkhmaa B, Petersen KS, Kris-Etherton PM, et al. Diet and Lp(a): does dietary change modify residual cardiovascular risk conferred by Lp(a)? Nutrients 2020;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Ginsberg HN, Kris-Etherton P, Dennis B, et al. Effects of reducing dietary saturated fatty acids on plasma lipids and lipoproteins in healthy subjects: the DELTA Study, protocol 1. Arterioscler Thromb Vasc Biol 1998;18:441–9. [DOI] [PubMed] [Google Scholar]
- [36].Ebbeling CB, Knapp A, Johnson A, et al. Effects of a low-carbohydrate diet on insulin-resistant dyslipoproteinemia-a randomized controlled feeding trial. Am J Clin Nutr 2022;115:154–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Mora S, Kamstrup PR, Rifai N, et al. Lipoprotein(a) and risk of type 2 diabetes. Clin Chem 2010;56:1252–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Tsimikas S, Viney NJ, Hughes SG, et al. Antisense therapy targeting apolipoprotein(a): a randomised, double-blind, placebo-controlled phase 1 study. Lancet 2015;386:1472–83. [DOI] [PubMed] [Google Scholar]
- [39].Koren MJ, Moriarty PM, Baum SJ, et al. Preclinical development and phase 1 trial of a novel siRNA targeting lipoprotein(a). Nat Med 2022;28:96–103. [DOI] [PubMed] [Google Scholar]
- [40].Pepin JM, O’Neil JA, Hoff HF. Quantification of apo[a] and apoB in human atherosclerotic lesions. J Lipid Res 1991;32:317–27. [PubMed] [Google Scholar]
- [41].Hoff HF, O’Neil J, Yashiro A. Partial characterization of lipoproteins containing apo[a] in human atherosclerotic lesions. J Lipid Res 1993;34:789–98. [PubMed] [Google Scholar]
- [42].Clarke R, Peden JF, Hopewell JC, et al. Genetic variants associated with Lp(a) lipoprotein level and coronary disease. N Engl J Med 2009;361:2518–28. [DOI] [PubMed] [Google Scholar]
- [43].Kamstrup PR, Tybjaerg-Hansen A, Nordestgaard BG. Genetic evidence that lipoprotein(a) associates with atherosclerotic stenosis rather than venous thrombosis. Arterioscler Thromb Vasc Biol 2012;32:1732–41. [DOI] [PubMed] [Google Scholar]
- [44].Kamstrup PR, Tybjaerg-Hansen A, Nordestgaard BG. Elevated lipoprotein(a) and risk of aortic valve stenosis in the general population. J Am Coll Cardiol 2014;63:470–7. [DOI] [PubMed] [Google Scholar]
- [45].Saleheen D, Haycock PC, Zhao W, et al. Apolipoprotein(a) isoform size, lipoprotein(a) concentration, and coronary artery disease: a Mendelian randomisation analysis. Lancet Diabetes Endocrinol 2017;5:524–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Kamstrup PR, Benn M, Tybjaerg-Hansen A, et al. Extreme lipoprotein(a) levels and risk of myocardial infarction in the general population: the Copenhagen City Heart Study. Circulation 2008;117:176–84. [DOI] [PubMed] [Google Scholar]
- [47].Erqou S, Kaptoge S, Perry PL, et al. Lipoprotein(a) concentration and the risk of coronary heart disease, stroke, and non-vascular mortality. JAMA 2009;302:412–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Langsted A, Varbo A, Kamstrup PR, et al. Elevated lipoprotein(a) does not cause low-grade inflammation despite causal association with aortic valve stenosis and myocardial infarction: a study of 100,578 individuals from the general population. J Clin Endocrinol Metab 2015;100:2690–9. [DOI] [PubMed] [Google Scholar]
- [49].Mathieu P, Arsenault BJ, Boulanger MC, et al. Pathobiology of Lp(a) in calcific aortic valve disease. Expert Rev Cardiovasc Ther 2017;15:797–807. [DOI] [PubMed] [Google Scholar]
- [50].Towler DA. Lp(a) oxyphospholipids: markers and mediators of vascular mineral metabolism in calcific aortic valve disease. J Am Coll Cardiol 2019;73:2163–5. [DOI] [PubMed] [Google Scholar]
- [51].Langsted A, Nordestgaard BG, Kamstrup PR. Elevated lipoprotein(a) and risk of ischemic stroke. J Am Coll Cardiol 2019;74:54–66. [DOI] [PubMed] [Google Scholar]
- [52].Langsted A, Kamstrup PR, Nordestgaard BG. High lipoprotein(a) and high risk of mortality. Eur Heart J 2019;40:2760–70. [DOI] [PubMed] [Google Scholar]
- *[53].Reyes-Soffer G, Ginsberg HN, Berglund L, et al. Lipoprotein(a): a genetically determined, causal, and prevalent risk factor for atherosclerotic cardiovascular disease: a scientific statement from the American Heart Association. Arterioscler Thromb Vasc Biol 2022;42:e48–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Tmoyan NA, Ezhov MV, Afanasieva OI, et al. The association of lipoprotein(a) and apolipoprotein(a) phenotypes with peripheral artery disease. Ter Arkh 2018;90:31–6. [DOI] [PubMed] [Google Scholar]
- [55].Price JF, Lee AJ, Rumley A, et al. Lipoprotein (a) and development of intermittent claudication and major cardiovascular events in men and women: the Edinburgh Artery Study. Atherosclerosis 2001;157:241–9. [DOI] [PubMed] [Google Scholar]
- [56].Kraaijenhof JM, Hovingh GK, Stroes ESG, et al. The iterative lipid impact on inflammation in atherosclerosis. Curr Opin Lipidol 2021;32:286–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Schnitzler JG, Hoogeveen RM, Ali L, et al. Atherogenic lipoprotein(a) increases vascular glycolysis, thereby facilitating inflammation and leukocyte extravasation. Circ Res 2020;126:1346–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Kaltoft M, Glavind KS, Nielsen SF, et al. Lipoprotein(a) during COVID-19 hospitalization: Thrombosis, inflammation, and mortality. Atherosclerosis 2022;357:33–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Lacaze P, Bakshi A, Riaz M, et al. Aspirin for primary prevention of cardiovascular events in relation to lipoprotein(a) genotypes. J Am Coll Cardiol 2022;80:1287–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med 2017;377:1119–31. [DOI] [PubMed] [Google Scholar]
- [61].Ridker PM, MacFadyen JG, Thuren T, et al. Residual inflammatory risk associated with interleukin-18 and interleukin-6 after successful interleukin-1beta inhibition with canakinumab: further rationale for the development of targeted anti-cytokine therapies for the treatment of atherothrombosis. Eur Heart J 2019. [DOI] [PubMed] [Google Scholar]
- [62].Ridker PM, Libby P, MacFadyen JG, et al. Modulation of the interleukin-6 signalling pathway and incidence rates of atherosclerotic events and all-cause mortality: analyses from the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS). Eur Heart J 2018;39:3499–507. [DOI] [PubMed] [Google Scholar]
- [63].Blankenberg S, Tiret L, Bickel C, et al. Interleukin-18 is a strong predictor of cardiovascular death in stable and unstable angina. Circulation 2002;106:24–30. [DOI] [PubMed] [Google Scholar]
- [64].Muller N, Schulte DM, Turk K, et al. IL-6 blockade by monoclonal antibodies inhibits apolipoprotein (a) expression and lipoprotein (a) synthesis in humans. J Lipid Res 2015;56:1034–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Puri R, Nissen SE, Arsenault St BJ, et al. Effect of C-reactive protein on lipoprotein(a)-associated cardiovascular risk in optimally treated patients with high-risk vascular disease: a prespecified secondary analysis of the ACCELERATE trial. JAMA Cardiol 2020;5:1136–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Zhang W, Speiser JL, Ye F, et al. High-sensitivity C-reactive protein modifies the cardiovascular risk of lipoprotein(a): multi-ethnic study of atherosclerosis. J Am Coll Cardiol 2021;78:1083–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Leibundgut G, Scipione C, Yin H, et al. Determinants of binding of oxidized phospholipids on apolipoprotein (a) and lipoprotein (a). J Lipid Res 2013;54:2815–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Reyes-Soffer G, Westerterp M. Beyond lipoprotein(a) plasma measurements: lipoprotein(a) and inflammation. Pharmacol Res 2021;169:105689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Yeang C, Wilkinson MJ, Tsimikas S. Lipoprotein(a) and oxidized phospholipids in calcific aortic valve stenosis. Curr Opin Cardiol 2016;31:440–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Marcovina SM, Moriarty PM, Koschinsky ML, et al. JCL roundtable-Lipoprotein(a): the emerging risk factor. J Clin Lipidol 2018;12:1335–45. [DOI] [PubMed] [Google Scholar]
- [71].Tsimikas S, Fazio S, Viney NJ, et al. Relationship of lipoprotein(a) molar concentrations and mass according to lipoprotein(a) thresholds and apolipoprotein(a) isoform size. J Clin Lipidol 2018. [DOI] [PubMed] [Google Scholar]
- [72].Wieringa G, Toogood AA, Ryder WD, et al. Changes in lipoprotein(a) levels measured by six kit methods during growth hormone treatment of growth hormone-deficient adults. Growth Hormone IGF Res 2000;10:14–9. [DOI] [PubMed] [Google Scholar]
- [73].Dembinski T, Nixon P, Shen G, et al. Evaluation of a new apolipoprotein(a) isoform-independent assay for serum lipoprotein (a). Mol Cell Biochem 2000;207:149–55. [DOI] [PubMed] [Google Scholar]
- [74].Tsimikas S, Fazio S, Ferdinand KC, et al. NHLBI Working Group Recommendations to reduce lipoprotein(a)-mediated risk of cardiovascular disease and aortic stenosis. J Am Coll Cardiol 2018;71:177–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;139:e1082–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Pearson GJ, Thanassoulis G, Anderson TJ, et al. 2021 Canadian Cardiovascular Society Guidelines for the management of dyslipidemia for the prevention of cardiovascular disease in adults. Can J Cardiol 2021;37:1129–50. [DOI] [PubMed] [Google Scholar]
- [77].Puri R, Mehta V, Iyengar SS, et al. Lipid association of India expert consensus statement on management of dyslipidemia in Indians 2020: Part III. J Assoc Phys India 2020;68:8–9. [PubMed] [Google Scholar]
- [78].Durlach V, Bonnefont-Rousselot D, Boccara F, et al. Lipoprotein(a): pathophysiology, measurement, indication and treatment in cardiovascular disease. A consensus statement from the Nouvelle Societe Francophone d’Atherosclerose (NSFA). Arch Cardiovasc Dis 2021;114:828–47. [DOI] [PubMed] [Google Scholar]
- [79].Makover ME, Shapiro MD, Toth PP. There is urgent need to treat atherosclerotic cardiovascular disease risk earlier, more intensively, and with greater precision: A review of current practice and recommendations for improved effectiveness. Am J Prev Cardiol 2022;12:100371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Tsimikas S, Gordts P, Nora C, et al. Statin therapy increases lipoprotein(a) levels. Eur Heart J 2020;41:2275–84. [DOI] [PubMed] [Google Scholar]
- [81].Pokrovsky SN, Afanasieva OI, Ezhov MV. Therapeutic apheresis for management of Lp(a) hyperlipoproteinemia. Curr Atheroscler Rep 2020;22:68. [DOI] [PubMed] [Google Scholar]
- [82].Hohenstein B, Julius U, Lansberg P, et al. Rationale and design of MultiSELECt: a European multicenter study on the effect of lipoprotein(a) elimination by lipoprotein apheresis on Cardiovascular outcomes. Atheroscler Suppl 2017;30:180–6. [DOI] [PubMed] [Google Scholar]
- [83].Khan TZ, Pottle A, Pennell DJ, et al. The expanding role of lipoprotein apheresis in the treatment of raised lipoprotein(a) in ischaemic heart disease and refractory angina. Glob Cardiol Sci Pract 2014;2014:3–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Khan TZ, Hsu LY, Arai AE, et al. Apheresis as novel treatment for refractory angina with raised lipoprotein(a): a randomized controlled cross-over trial. Eur Heart J 2017;38:1561–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Nissen SE, Wolski K, Balog C, 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;327:1679–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- *[86].Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, et al. Lipoprotein(a) reduction in persons with cardiovascular disease. N Engl J Med 2020;382:244–55. [DOI] [PubMed] [Google Scholar]
- [87].Kotwal A, Cortes T, Genere N, et al. Treatment of thyroid dysfunction and serum lipids: a systematic review and meta-analysis. J Clin Endocrinol Metab 2020;105. [DOI] [PubMed] [Google Scholar]
- [88].Eden S, Wiklund O, Oscarsson J, et al. Growth hormone treatment of growth hormone-deficient adults results in a marked increase in Lp(a) and HDL cholesterol concentrations. Arterioscler Thromb 1993;13:296–301. [DOI] [PubMed] [Google Scholar]
- [89].Zechner R, Desoye G, Pfeiffer KP, et al. Fluctuations of plasma lipoprotein-A concentrations during pregnancy and post partum. Metabolism 1986;35:333–6. [DOI] [PubMed] [Google Scholar]
- [90].Salpeter SR, Buckley NS, Salpeter EE, et al. Meta-analysis: effect of hormone-replacement therapy on components of the metabolic syndrome in postmenopausal women. Diabetes Obes Metab 2006;8:538–54. [DOI] [PubMed] [Google Scholar]
- [91].Kronenberg F. Causes and consequences of lipoprotein(a) abnormalities in kidney disease. Clin Exp Nephrol 2014;18:234–7. [DOI] [PubMed] [Google Scholar]
- [92].Missala I, Kassner U, Steinhagen-Thiessen E. A systematic literature review of the association of lipoprotein(a) and auto-immune diseases and atherosclerosis. Int J Rheumatol 2012;2012:480784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Tsimikas SFS, Santos RD, Thanassoulis G, et al. NHLBI Working Group Recommendations to reduce lipoprotein(a)-mediated risk of cardiovascular disease and aortic stenosis. J Am Coll Cardiol 2018;71:177–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- *[94].Wilson DP, Nordestgaard BG, Orringer CE, et al. Use of lipoprotein(a) in clinical practice: a biomarker whose time has come. A scientific statement from the National Lipid Association. J Clin Lipidol 2019;13:374–92. [DOI] [PubMed] [Google Scholar]
- [95].Authors/Task Force M, Guidelines ESCCfP and Societies ESCNC. 2019 ESC/EAS guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Atherosclerosis 2019;290:140–205. [DOI] [PubMed] [Google Scholar]
- *[96].Mach F, Baigent C, Catapano AL, et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020(41):111–88. [DOI] [PubMed] [Google Scholar]
- [97].Newman CB, Subramanian S, Tannock LR, et al. Lipid management in patients with endocrine disorders: an endocrine society clinical practice guideline. J Clin Endocrinol Metab 2020;105. [DOI] [PubMed] [Google Scholar]
