Review Article

Role of Lipoprotein(a) in Cardiovascular Disease: A Review of Clinical Practice

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

Abstract

The association between elevated lipoprotein(a) (Lp(a)) and an increased risk of cardiovascular disease has been demonstrated. Although the impact of Lp(a) plasma level on the onset of disease depends on the type of disease and the patient’s comorbidities, an Lp(a) plasma level >1.29 mmol/l (50 mg/dl) has been proposed as a practical cut-off. Approximately 10% of the general Asian population may have Lp(a) >1.29 mmol/l, as do 15–30% of the global population. An alternative cut-off for Asian populations may be 0.78 mmol/l (30 mg/dl). Measurements may have to be considered at least once in each adult’s lifetime, particularly for people with a family or personal history of premature atherosclerotic cardiovascular disease. Although plasma level is mostly consistent throughout life, some therapies, such as proprotein convertase subtilisin/ kexin type 9 inhibitors and antisense oligonucleotides, may reduce the Lp(a) plasma level.

Disclosure:JA is a deputy editor of Journal of Asian Pacific Society of Cardiology; this did not influence peer review. All other authors have no conflicts of interest to declare.

Received:

Accepted:

Published online:

Correspondence Details:Yoshiyasu Minami, Department of Cardiovascular Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan. E: nrg12391@yahoo.co.jp

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

The lipoprotein(a) (Lp(a)) molecule consists of an apolipoprotein (Apo) B-containing LDL-like segment and a plasminogen-like glycoprotein Apo(a) segment. An increased risk of cardiovascular disease, including acute coronary syndrome, ischaemic stroke and aortic stenosis, in patients with high plasma Lp(a) has been demonstrated in clinical studies, genome-wide association studies and Mendelian randomisation studies.1–4 The increased risk is often explained by the enhanced progression of atherosclerosis in the arterial system through the proatherogenic and anti-fibrinolytic nature of Lp(a) in combination with other factors, including age, sex, ethnicity, comorbidities and LDL cholesterol level. The independent association between elevated Lp(a) and an increased risk of cardiovascular events has been demonstrated, therefore the role of residual risk in patients with statin-based conventional therapy has been emphasised.2 The level of Lp(a) is established during childhood and is mostly consistent throughout the lifetime because it is regulated primarily by the LPA gene locus. However, several therapies, including proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, inclisiran and antisense oligonucleotides, have been reported to reduce the plasma level.5–8 Approximately 20–30% of people have elevated Lp(a), so more clinical attention is required.9

In this review, we summarise the clinical impact of elevated Lp(a), the evaluation of Lp(a) in daily practice, and the potential therapeutic approaches for high Lp(a) (Figure 1).

Epidemiology of High Plasma Lp(a)

The distribution of plasma Lp(a) level is skewed and ranges widely between individuals, with no sex differences.3 Plasma concentration is determined primarily by the LPA gene locus, without significant acquired or environmental influences.10 Patients with established cardiovascular disease had higher levels of Lp(a) (Figure 2). There are ethnic differences in median Lp(a) level between countries caused by the difference in the prevalence of LPA single nucleotide polymorphisms and Apo(a) isoforms.11,12 The median Lp(a) level in the general Asian population is 0.21–0.36 mmol/l, which is lower than that in black (1.01 mmol/l), Hispanic (0.49 mmol/l) and Arab (0.39 mmol/l) populations.11,13–15

Cut-off of Lp(a) for Risk Stratification

Several practical cut-offs for Lp(a), including 0.78 mmol/l (30 mg/dl) and 1.29 mmol/l (50 mg/dl) for the risk stratification of coronary artery disease (CAD) and ischaemic stroke, have been proposed. The Copenhagen City Heart Study (n=7,524) found that Lp(a) >0.78 mmol/l is associated with an increased risk of MI in a dose-dependent manner, with an adjusted HR of 1.2 (95% CI [0.9–1.6]) for Lp(a) 0.13–0.75 mmol/l (5–29 mg/dl), 1.6 (95% CI [1.1–2.2]) for Lp(a) 0.78–1.97 mmol/l (30–76 mg/dl), 1.9 (95% CI [1.2–3.0]) for Lp(a) 1.99–3.03 mmol/l (77–117 mg/dl), and 2.6 (95% CI [1.6–4.1]) for Lp(a) >3.03 mmol/l (117 mg/dl) versus for Lp(a) <0.13 mmol/l (5 mg/dl).3

A meta-analysis of 126,634 participants showed that the risk of MI started to increase steeply at Lp(a) >0.62 mmol/l, and curvilinearly increased according to the increase in Lp(a) and became significant at around 1.24 mmol/l.2 A genome-wide association study also showed a steep increase in risk at >0.78 mmol/l and an added risk at >1.22 mmol/l.16 Current guidelines in the US, Canada and Europe define 1.29 mmol/l (50 mg/dl) as a risk-enhancing factor.17–19

Lipoprotein (a) and Cardiovascular Disease

Article image

Globally, the estimated number of people with elevated Lp(a) is >1 billion. Approximately 10% of the general Asian population may have Lp(a) >1.29 mmol/l, compared to 15–30% of the global population.20 A recent report investigated ethnic differences in the HR of high Lp(a) (>1.29 mmol/l) for the incidence of MI.15 The HR in Chinese, Southeast Asian and South Asian populations is reported as 1.62, 1.83, and 2.14, respectively, while it is 1.36 in European and 1.67 in Latin American populations. Although few reports have focused on the practical cut-off point of Lp(a) for Asian populations, Lp(a) ≥0.78 mmol/l (30 mg/dl) may be more suitable for the Chinese population.20,21 Several studies have suggested that elevated Lp(a) remains a risk factor for cardiovascular disease, even in patients with LDL cholesterol <1.81 mmol/l.22–24 Meanwhile, a recent study reported that the association of elevated Lp(a) and the risk of cardiovascular disease is attenuated in a primary prevention setting at LDL cholesterol <2.5 mmol/l.25

Impact of Elevated Lp(a) on the Incidence of Cardiovascular Disease

Elevated Lp(a) is associated with an increased risk of premature onset of CAD, ischaemic stroke, peripheral artery disease (PAD) and aortic valve degradation.2,26–28 The association of elevated Lp(a) and an increased incidence of recurrent cardiovascular disease in patients treated with statin and percutaneous coronary intervention (PCI) has been also reported.29–32

Elevated Lp(a) and Coronary Artery Disease

In the general population, a significant increase in the risk of coronary death and non-fatal MI has been observed in people with Lp(a) >1.29 mmol/l (50 mg/dl).2 In patients with established CAD, elevated Lp(a) increases the risk of recurrent clinical events, particularly in those with LDL cholesterol ≥3.37 mmol/l.33,34 The increased risk of recurrent cardiovascular events, according to the increase in Lp(a), has been shown even in patients treated with statins.29 The increased incidence of CAD in people with elevated Lp(a) is often explained by the higher prevalence of coronary plaque through the effect of the proatherogenic properties of Lp(a) particles.35,36 The association of elevated Lp(a) with a greater number and degree of coronary stenosis on angiography, a larger number of stenoses on CT, and a greater change in the plaque-plus-media area on serial intravascular ultrasound has also been reported.35–37 Chieng et al. investigated the impact of Lp(a) and LDL cholesterol levels on the angiographic disease severity assessed using the SYNergy between percutaneous coronary intervention with TAXus and cardiac surgery (SYNTAX) score in patients with premature CAD admitted to hospital in Australia.38 They showed that patients with both elevated Lp(a) and elevated LDL cholesterol comprised the majority of patients in the highest SYNTAX tertile, while patients with non-elevated Lp(a) and non-elevated LDL cholesterol were predominant in the lowest SYNTAX tertile.38

Muramatsu et al. investigated the impact of Lp(a) and LDL cholesterol level on the prevalence of vulnerable coronary plaque using optical coherence tomography in a Japanese population.39 They noted an increased prevalence of thin-cap fibroatheroma in the culprit coronary plaque with an increase in Lp(a) level, particularly in patients with LDL cholesterol ≥2.59 mmol/l.39 The increased incidence of recurrent cardiovascular events after PCI in patients with elevated Lp(a) has been shown in a Japanese cohort with chronic kidney disease and diabetes.30,31

A recent report from Korea has also shown the impact of elevated Lp(a) on the increased incidence of recurrent ischaemic cardiovascular events including repeated revascularisation and ischaemic stroke after PCI.32 The authors showed that the HR of recurrent events in patients with Lp(a) ≥0.78 mmol/l (30 mg/dl) during a median follow-up of 7.4 years was 1.17 (95% CI [1.05–1.30]; p=0.004). They noted that the enhanced proliferation of smooth muscle cells in addition to accelerated atherosclerosis via elevated Lp(a) might be a contributing factor.

Elevated Lp(a) and Peripheral Artery Disease

A modest but significant association between Lp(a) and PAD was noted in a study of Mendelian randomisation.26 The authors showed that increases in genetically predicted Lp(a) were associated with an increased risk of PAD (OR 1.04 per 0.26 mmol/l increase in Lp(a)). They also found that the association was not attenuated in multivariable analyses, accounting for the association of these genetic variants with ApoB. Further studies are needed to confirm the impact of elevated Lp(a) on the increased incidence of PAD.

Elevated Lp(a) and Stroke

A recent meta-analysis showed a significant association between elevated Lp(a) and an increased risk of ischaemic stroke compared with control subjects.27 The association between elevated Lp(a) and the increased risk of ischaemic stroke was also demonstrated in Asian patients.40–42 The meta-analysis further showed that elevated Lp(a) was significantly associated with an increased risk of the large artery atherosclerosis subtype as well as an increased risk of intracerebral haemorrhage. Most of the studies included in the meta-analysis used 0.52–1.04 mmol/l (20–40 mg/dl) as the cut-off for elevated Lp(a).

A recent prospective and observational study investigated the relationship between acute ischaemic stroke and serum Lp(a) level in the Chinese population.41 The authors reported a significant difference in median serum Lp(a) between patients with acute ischaemic stroke and control cases (0.85 versus 0.39 mmol/l [33 versus 15 mg/dl]; p=0.000). The authors also reported that elevated Lp(a) was an independent factor for stroke, and serum Lp(a) ≥0.78 mmol/l (30 mg/dl) was associated with a 2.23-fold increase in acute ischaemic stroke when adjusting for other possible risk factors. The pathogenesis of an increased incidence of ischaemic stroke in people with elevated Lp(a) could be explained by advanced carotid atherosclerosis.43,44 The association between increased Lp(a) and a higher prevalence of vulnerable plaque characteristics in the carotid artery in patients with symptomatic carotid artery stenosis has been demonstrated in a previous study.45

Lipoprotein(a) Levels in Patients with Acute Coronary Syndrome

Article image

Elevated Lp(a) and Aortic Valve Disease

The association between elevated Lp(a) and calcification and stenosis of the aortic valve has been reported in several studies.46,47 A study combining two prospective studies of the general population showed that elevated Lp(a) and the corresponding genotypes were associated with an increased risk of aortic valve stenosis in the general population, with Lp(a) >2.33 mmol/l (90 mg/dl) predicting a threefold higher risk.28 A recent study with serial observations using multiple modalities showed that patients in the highest Lp(a) tertile had a higher progression of valvular calcification, faster haemodynamic progression, and a higher risk of aortic valve replacement and death compared with patients in the lower tertiles.48 Increased alkaline phosphatase activity, hydroxyapatite, cell apoptosis and phosphorylation of signal transduction proteins have been proposed as a conceivable pathway of aortic valve degradation by elevated Lp(a).49 Although the ethnic difference in the effect of elevated Lp(a) on degradation of the aortic valve remains unclear, a recent study reported that there were no associations between Lp(a) level and the extent of aortic valve calcification in South Asian people, although the association was seen in white people and black people.50

Indications for Lp(a) Measurement

The potential of Lp(a) measurement as a tool for the stratification of risk of future cardiovascular events was shown in a study of 826 people in the general population.51 Elevated Lp(a) was associated with an increased risk of cardiovascular disease over a 15-year follow-up period: the adjusted HR for cardiovascular events was 1.37 per 1 SD higher level of Lp(a) (SD=0.83 mmol/l [32 mg/dl]) and 2.37 compared with the top fifth quintile. The evaluation of Lp(a) has been shown to improve the quality of risk stratification in the general population with borderline risk, defined using the Framingham risk score.25,52

Measurements of Lp(a) enable the reclassification of the risk of cardiovascular disease and facilitate shared decision-making for the initiation of treatment, especially in younger patients, particularly those with a family history of premature cardiovascular disease. Current guidelines from Europe and North America recommend measuring Lp(a) in patients with a family history of premature cardiovascular disease, with Lp(a) >1.29 mmol/l (50 mg/dl) considered a risk-enhancing factor.17–19 A Mendelian randomisation study showed that extremely elevated Lp(a) (>5.172 mmol/l [200 mg/dl]) is associated with a three- to fourfold increased risk of cardiovascular disease, representing a similar lifetime risk of cardiovascular disease in heterozygous familial hypercholesterolaemia.53

Although it remains unclear whether universal testing of Lp(a) is recommended for healthy people regardless of the presence of a family history, the potential causal association between Lp(a) and future cardiovascular events has been shown in several Mendelian randomisation studies, which could justify universal screening.1–4 Some of the current guidelines recommend Lp(a) measurement at least once in the lifetime of each adult person to identify people who may have inherited extremely high Lp(a) equivalent to the risk associated with familial hyperlipidaemia.18,19 In patients with established cardiovascular disease, elevated Lp(a) is associated with an increased incidence of recurrent cardiovascular events irrespective of LDL cholesterol.6,54 However, no randomised controlled trial has demonstrated a significant reduction in recurrent clinical events through lowering of Lp(a). Therefore, plasma Lp(a) cannot be a treatment goal in current clinical practice, and the serial measurement of Lp(a) is not recommended in guidelines.17–19 The possible indications for Lp(a) measurement in daily clinical practice are listed in Table 1.

Issues of Lp(a) Measurement

Lp(a) level may sometimes need to be interpreted with caution. Because of the wide range in the size of the Apo(a) segment and lipid content, the molecular mass of the Lp(a) particle also varies widely. The size of the Apo(a) segment depends on the number of kringle (KIV2) repeats, which is genetically determined.55 This may cause an ethnic difference in the size distribution of the Lp(a) molecule and raise difficulties in the application of results to patients of a different ethnicity.56 Although most currently available assays provide acceptable accuracy in the measurement of Lp(a) level to differentiate high-risk patients, the system of different assays still requires standardisation and appropriate calibration.1,56

Lp(a) Measurement in Daily Clinical Practice

Article image

Potential Therapies for Lp(a) Lowering

Article image

Potential Approaches for Lowering Lp(a)

There have been no dedicated randomised clinical trials on the impact of Lp(a) lowering on the reduction of cardiovascular disease. Although the pharmacological mechanisms are unclear, a potentially significant decrease in plasma Lp(a) by several drugs has been reported.7,8,57 For primary prevention in people with intermediate risk, defined as a cardiovascular disease risk of 7.5–20% calculated using the Framingham risk score, elevated Lp(a) may be considered a risk-enhancing factor to start statin therapy.17,58 In high-risk or very-high-risk patients with LDL cholesterol ≥6.98 mmol/l on maximally tolerated statin treatment, it might be reasonable to consider more intensive treatment based on elevated Lp(a).59 In the setting of secondary prevention, the presence of a high Lp(a) level is strongly predictive of recurrent cardiovascular events and suggests the need for more intensive therapies, including PCSK9 inhibitors. Table 2 summarises the current status of potential therapies for Lp(a) lowering and subsequent reduction of cardiovascular events.

Statins

The role of statins in the primary and secondary prevention of cardiovascular events has been established. However, several prospective statin studies have shown a significant increase in plasma Lp(a) after starting statin therapy.34,60–64 A meta-analysis showed that statin increased Lp(a) by 8.5–19.5% from baseline, although it remains to be elucidated whether the statin-mediated increase in Lp(a) contributes to the increased risk of clinical events.65 Another meta-analysis found that statins may modify the association between Lp(a) and the risk of cardiovascular disease.29 In the analysis, Lp(a) level was shown to be more strongly associated with an increased risk of cardiovascular disease in patients with statins than in those with placebo.29

Niacin

Niacin, also known as vitamin B3, is an essential vitamin in the human body. Niacin can decrease total cholesterol, LDL cholesterol and triglyceride levels, and increase HDL cholesterol in large doses. Its lipid-lowering function is associated with the inhibition of hormone-sensitive lipase activity in adipose tissue, reduction of free fatty acid entry into the liver, and decrease in very-low-density lipoprotein secretion.66 Niacin also reduces Lp(a) levels by 20–30% by inhibiting the LPA promoter.23 However, HPS2-4THRIVE, a double-blind randomised trial, demonstrated that niacin failed to reduce cardiovascular events in patients with CAD. The addition of niacin to statin treatment resulted in significant increases in adverse effects, including disturbances in diabetes control, dysplasia, diarrhoea, myopathy, serious infection and skin ulcerations.67 The combination of niacin and statins in lipid-lowering therapy is no longer recommended in most countries.18

PCSK9 Inhibitors

PCSK9 inhibitors have been shown to reduce Lp(a) by 7–36%.54,57,68 This effect is consistent and reproducible for both alirocumab and evolocumab, although the pharmacological mechanism of reduction of Lp(a) by PCSK9 inhibitors remains unclear.69,70 In the FOURIER trial that included patients with cardiovascular disease and maximum tolerated dose of statin, evolocumab significantly reduced Lp(a) by a median of 26.9% at 48 weeks.54 Evolocumab has been shown to reduce the risk of cardiovascular events by 23% in patients with high Lp(a) and by 7% in those with low Lp(a). This indicates that the higher the baseline Lp(a), the greater the potential benefit of PCSK9 inhibitors in reducing cardiovascular events.54 A subanalysis of the Odyssey Outcomes trial showed that alirocumab-induced reductions of Lp(a) independently predicted lower risk of recurrent cardiovascular events, after adjustment for baseline concentrations of both LDL cholesterol and Lp(a) and demographic and clinical characteristics (i.e. a 0.03 mmol/l [1 mg/dl] reduction in Lp(a) with alirocumab was associated with a HR of 0.994 for recurrent cardiovascular events).6

Inclisiran

A potential for Lp(a) lowering by inclisiran has been reported although the pharmacological mechanism remains unclear. Inclisiran is a small interfering RNA (siRNA) molecule that reduces LDL cholesterol through the inhibition of PCSK9 synthesis. The safety and efficacy of inclisiran given twice a year by subcutaneous injection have been demonstrated.7,71,72 The current ongoing randomised trial ORION-4 (NCT03705234) will provide information on the effect of inclisiran on the incidence of cardiovascular events. In addition to the reduction of LDL cholesterol levels by 52.3% without serious adverse effects, inclisiran also reduced Lp(a) by 14–26%, although this did not reach statistical significance.7,73

Antisense Oligonucleotides

Mipomersen is an antisense oligonucleotide (ASO) that is designed to inhibit the synthesis of the ApoB100 protein and which produces a modest reduction in Lp(a). The percentage change in LDL cholesterol and Lp(a) with mipomersen therapy was −24.0% and −31.6%, respectively.74 Apo(a)-LRx is also an ASO, and it is rapidly and specifically taken up by hepatocytes and inhibits the synthesis of Apo(a).75,76 Apo(a)-LRx has been shown to reduce Lp(a) by 66–91% in a dose-dependent manner in healthy participants with elevated Lp(a), as well as in patients with established cardiovascular disease and elevated Lp(a).8,77

In a randomised trial of patients with established cardiovascular disease who had Lp(a) ≥1.55 mmol/l (60 mg/dl) on screening, Apo(a)-LRx at a dose of 20 mg weekly for 6 months resulted in an 80% reduction in Lp(a) level, and 98% of the patients on this dose attained an Lp(a) level <1.29 mmol/l (50 mg/dl).8 Another randomised trial compared the Lp(a)-lowering and anti-inflammatory effects of Apo(a)-LRx with PCSK9 inhibitors.78 In that study, the percent change in Lp(a) in patients with Apo(a)-LRx and PCSK9 inhibitors was −46.6% and −16.1%, respectively. Furthermore, potent and highly selective Lp(a) lowering by Apo(a)-LRx resulted in the reduction of inflammatory gene expression in circulating monocytes in patients with cardiovascular disease. In contrast, PCSK9 inhibitors did not alter Lp(a)-induced proinflammatory profiles.78 These findings support the potential benefit of improving clinical outcomes through a greater reduction of Lp(a) in patients with established cardiovascular disease. The current Phase III study (HORIZON; NCT04023552) will clarify the treatment effect of Apo(a)-LRx on clinical outcomes in patients with cardiovascular disease.

Lipoprotein Apheresis

Lipoprotein apheresis (LA) is the final escalating option to reduce blood LDL cholesterol levels in patients with severe hypercholesterolaemia, such as familial hypercholesterolaemia or other forms of hypercholesterolaemia resistant to or intolerant to lipid-lowering medication. LA should be considered for patients with elevated Lp(a) and progressive cardiovascular disease and is the only FDA-approved therapy to lower Lp(a) in the US. LA reduces Lp(a) by 70% in patients with elevated Lp(a) and cardiovascular disease.79 Although no randomised controlled studies have been conducted to demonstrate the effectiveness of LA in reducing cardiovascular events, LA has been shown to be associated with a 94% reduction in cardiovascular events over a mean treatment period of 48 months.79 In the Pro(a)LiFe study, patients with high Lp(a) were prospectively followed, and the incidence rates of cardiovascular events between 2 years before and 2 years after LA treatment were compared. The mean rate of cardiovascular events declined from 0.41 for 2 years before LA to 0.09 for 2 years during LA.80 LA effectively lowered the incidence of cardiovascular events with a significant reduction in Lp(a). Both the German and UK apheresis guidelines use Lp(a) >1.55 mmol/l (60 mg/dl) to specifically allow reimbursement for patients with isolated Lp(a) elevation and recurrent cardiovascular events or in conjunction with uncontrolled elevated LDL cholesterol.81,82

Conclusion

Several scientific approaches, including observational studies, genetic studies, and Mendelian randomisation studies, have clarified the clinical impact of elevated Lp(a). A level of 1.29 mmol/l (50 mg/dl) has been suggested as a practical cut-off point for risk stratification for future cardiovascular events in daily practice, while a level of 0.78 mmol/l (30 mg/dl) may be an alternative cut-off for Asian populations. In addition to people with a high risk of atherosclerotic cardiovascular disease, the healthy general population may also be a potential candidate for Lp(a) measurement. Although the optimal pharmacological intervention for elevated Lp(a) has not been established, some ongoing trials may open the door to novel treatment strategies for people with elevated Lp(a). Healthcare providers must pay more attention to the risk stratification of cardiovascular disease according to Lp(a) level.

Clinical Perspective

  • Elevated lipoprotein (a) (Lp(a)) is associated with an increased risk of cardiovascular disease.
  • Plasma Lp(a) >1.29 mmol/l (50 mg/dl) has been proposed as a practical cut-off.
  • An alternative cut-off of Lp(a) for Asian populations may be 0.78 mmol/l (30 mg/dl).
  • Measurements may have to be considered at least once in the lifetime of each adult.
  • Proprotein convertase subtilisin kexin type 9 inhibitors and antisense oligonucleotides may reduce the Lp(a) plasma level.

References

  1. Tsimikas S. A test in context: lipoprotein(a): diagnosis, prognosis, controversies, and emerging therapies. J Am Coll Cardiol 2017;69:692–711.
    Crossref | PubMed
  2. Emerging Risk Factors Collaboration, Erqou S, Kaptoge S, et al. Lipoprotein(a) concentration and the risk of coronary heart disease, stroke, and nonvascular mortality. JAMA 2009;302:412–23.
    Crossref | PubMed
  3. Kamstrup PR, Tybjaerg-Hansen A, Steffensen R, Nordestgaard BG. Genetically elevated lipoprotein(a) and increased risk of myocardial infarction. JAMA 2009;301:2331–9.
    Crossref | PubMed
  4. 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.
    Crossref | PubMed
  5. Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of lipoprotein (a). J Lipid Res 2016;57:1339–59.
    Crossref | PubMed
  6. Bittner VA, Szarek M, Aylward PE, et al. Effect of alirocumab on lipoprotein(a) and cardiovascular risk after acute coronary syndrome. J Am Coll Cardiol 2020;75:133–44.
    Crossref | PubMed
  7. Ray KK, Wright RS, Kallend D, et al. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med 2020;382:1507–19.
    Crossref | PubMed
  8. 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.
    Crossref | PubMed
  9. Tsimikas S, Stroes ESG. The dedicated “Lp(a) clinic”: a concept whose time has arrived? Atherosclerosis 2020;300:1–9.
    Crossref | PubMed
  10. Kronenberg F. Human genetics and the causal role of lipoprotein(a) for various diseases. Cardiovasc Drugs Ther 2016;30:87–100.
    Crossref | PubMed
  11. Nordestgaard BG, Chapman MJ, Ray K, et al. Lipoprotein(a) as a cardiovascular risk factor: current status. Eur Heart J 2010;31:2844–53.
    Crossref | PubMed
  12. Lee SR, Prasad A, Choi YS, et al. LPA gene, ethnicity, and cardiovascular events. Circulation 2017;135:251–63.
    Crossref | PubMed
  13. Matthews KA, Sowers MF, Derby CA, et al. Ethnic differences in cardiovascular risk factor burden among middle-aged women: Study of Women’s Health Across the Nation (SWAN). Am Heart J 2005;149:1066–73.
    Crossref | PubMed
  14. Sandholzer C, Boerwinkle E, Saha N, et al. Apolipoprotein(a) phenotypes, Lp(a) concentration and plasma lipid levels in relation to coronary heart disease in a Chinese population: evidence for the role of the apo(a) gene in coronary heart disease. J Clin Invest 1992;89:1040–6.
    Crossref | PubMed
  15. Paré G, Çaku A, McQueen M, et al. Lipoprotein(a) levels and the risk of myocardial infarction among 7 ethnic groups. Circulation 2019;139:1472–82.
    Crossref | PubMed
  16. Kamstrup PR, Tybjærg-Hansen A, Nordestgaard BG. Extreme lipoprotein(a) levels and improved cardiovascular risk prediction. J Am Coll Cardiol 2013;61:1146–56.
    Crossref | PubMed
  17. 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.
    Crossref | PubMed
  18. 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.
    Crossref | PubMed
  19. 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.
    Crossref | PubMed
  20. 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.
    Crossref | PubMed
  21. 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.
    Crossref | PubMed
  22. Khera AV, Everett BM, Caulfield MP, et al. Lipoprotein(a) concentrations, rosuvastatin therapy, and residual vascular risk: an analysis from the Jupiter Trial (Justification for the Use of statins in Prevention: an Intervention Trial Evaluating Rosuvastatin). Circulation 2014;129:635–42.
    Crossref | PubMed
  23. Albers JJ, Slee A, O’Brien KD, et al. Relationship of apolipoproteins A-1 and B, and lipoprotein(a) to cardiovascular outcomes: the AIM-HIGH trial (Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglyceride and Impact on Global Health Outcomes). J Am Coll Cardiol 2013;62:1575–9.
    Crossref | PubMed
  24. Nestel PJ, Barnes EH, Tonkin AM, et al. Plasma lipoprotein(a) concentration predicts future coronary and cardiovascular events in patients with stable coronary heart disease. Arterioscler Thromb Vasc Biol 2013;33:2902–8.
    Crossref | PubMed
  25. Verbeek R, Hoogeveen RM, Langsted A, et al. Cardiovascular disease risk associated with elevated lipoprotein(a) attenuates at low low-density lipoprotein cholesterol levels in a primary prevention setting. Eur Heart J 2018;39:2589–96.
    Crossref | PubMed
  26. Levin MG, Zuber V, Walker VM, et al. Prioritizing the role of major lipoproteins and subfractions as risk factors for peripheral artery disease. Circulation 2021;144:353–64.
    Crossref | PubMed
  27. 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.
    Crossref | PubMed
  28. Kamstrup PR, Tybjærg-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.
    Crossref | PubMed
  29. Willeit P, Ridker PM, Nestel PJ, et al. Baseline and on-statin treatment lipoprotein(a) levels for prediction of cardiovascular events: individual patient-data meta-analysis of statin outcome trials. Lancet 2018;392:1311–20.
    Crossref | PubMed
  30. Konishi H, Miyauchi K, Tsuboi S, et al. Plasma lipoprotein(a) predicts major cardiovascular events in patients with chronic kidney disease who undergo percutaneous coronary intervention. Int J Cardiol 2016;205:50–3.
    Crossref | PubMed
  31. Konishi H, Miyauchi K, Shitara J, et al. Impact of lipoprotein(a) on long-term outcomes in patients with diabetes mellitus who underwent percutaneous coronary intervention. Am J Cardiol 2016;118:1781–5.
    Crossref | PubMed
  32. Yoon YH, Ahn JM, Kang DY, et al. Association of lipoprotein(a) with recurrent ischemic events following percutaneous coronary intervention. JACC Cardiovasc Interv 2021;14:2059–68.
    Crossref | PubMed
  33. Bucci M, Tana C, Giamberardino MA, Cipollone F. Lp(a) and cardiovascular risk: investigating the hidden side of the moon. Nutr Metab Cardiovasc Dis 2016;26:980–6.
    Crossref | PubMed
  34. O’Donoghue ML, Morrow DA, Tsimikas S, et al. Lipoprotein(a) for risk assessment in patients with established coronary artery disease. J Am Coll Cardiol 2014;63:520–7.
    Crossref | PubMed
  35. Dahlen GH, Guyton JR, Attar M, et al. Association of levels of lipoprotein Lp(a), plasma lipids, and other lipoproteins with coronary artery disease documented by angiography. Circulation 1986;74:758–65.
    Crossref | PubMed
  36. Kral BG, Kalyani RR, Yanek LR, et al. Relation of plasma lipoprotein(a) to subclinical coronary plaque volumes, three-vessel and left main coronary disease, and severe coronary stenoses in apparently healthy African-Americans with a family history of early-onset coronary artery disease. Am J Cardiol 2016;118:656–61.
    Crossref | PubMed
  37. Hartmann M, von Birgelen C, Mintz GS, et al. Relation between lipoprotein(a) and fibrinogen and serial intravascular ultrasound plaque progression in left main coronary arteries. J Am Coll Cardiol 2006;48:446–52.
    Crossref | PubMed
  38. Chieng D, Pang J, Ellis KL, et al. Elevated lipoprotein(a) and low-density lipoprotein cholesterol as predictors of the severity and complexity of angiographic lesions in patients with premature coronary artery disease. J Clin Lipidol 2018;12:1019–26.
    Crossref | PubMed
  39. Muramatsu Y, Minami Y, Kato A, et al. Lipoprotein (a) level is associated with plaque vulnerability in patients with coronary artery disease: an optical coherence tomography study. Int J Cardiol Heart Vasc 2019;24:100382.
    Crossref | PubMed
  40. Fu H, Zhang D, Zhu R, et al. Association between lipoprotein(a) concentration and the risk of stroke in the Chinese Han population: a retrospective case-control study. Ann Transl Med 2020;8:212.
    Crossref | PubMed
  41. Li SY, Gao Y, Ma WN, et al. The relationship between serum lipoprotein (a) levels and ischemic stroke risk: a cohort study in the Chinese population. Inflammation 2014;37:686–93.
    Crossref | PubMed
  42. Sun L, Li Z, Zhang H, et al. Pentanucleotide TTTTA repeat polymorphism of apolipoprotein(a) gene and plasma lipoprotein(a) are associated with ischemic and hemorrhagic stroke in Chinese: a multicenter case-control study in China. Stroke 2003;34:1617–22.
    Crossref | PubMed
  43. Gencer B, Kronenberg F, Stroes ES, Mach F. Lipoprotein(a): the revenant. Eur Heart J 2017;38:1553–60.
    Crossref | PubMed
  44. Kronenberg F, Kronenberg MF, Kiechl S, et al. Role of lipoprotein(a) and apolipoprotein(a) phenotype in atherogenesis: prospective results from the Bruneck study. Circulation 1999;100:1154–60.
    Crossref | PubMed
  45. van Dam-Nolen DHK, van Dijk AC, Crombag GAJC, et al. Lipoprotein(a) levels and atherosclerotic plaque characteristics in the carotid artery: the Plaque at RISK (PARISK) study. Atherosclerosis 2021;329:22–9.
    Crossref | PubMed
  46. Stewart BF, Siscovick D, Lind BK, et al. Clinical factors associated with calcific aortic valve disease. Cardiovascular Health Study. J Am Coll Cardiol 1997;29:630–4.
    Crossref | PubMed
  47. Bozbas H, Yildirir A, Atar I, et al. Effects of serum levels of novel atherosclerotic risk factors on aortic valve calcification. J Heart Valve Dis 2007;16:387–93.
    PubMed
  48. Zheng KH, Tsimikas S, Pawade T, et al. Lipoprotein(a) and oxidized phospholipids promote valve calcification in patients with aortic stenosis. J Am Coll Cardiol 2019;73:2150–62.
    Crossref | PubMed
  49. Yu B, Hafiane A, Thanassoulis G, et al. Lipoprotein(a) induces human aortic valve interstitial cell calcification. JACC Basic Transl Sci 2017;2:358–71.
    Crossref | PubMed
  50. Makshood M, Joshi PH, Kanaya AM, et al. Lipoprotein (a) and aortic valve calcium in South Asians compared to other race/ethnic groups. Atherosclerosis 2020;313:14–9.
    Crossref | PubMed
  51. Willeit P, Kiechl S, Kronenberg F, et al. Discrimination and net reclassification of cardiovascular risk with lipoprotein(a): prospective 15-year outcomes in the Bruneck Study. J Am Coll Cardiol 2014;64:851–60.
    Crossref | PubMed
  52. 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.
    Crossref | PubMed
  53. Burgess S, Ference BA, Staley JR, et al. Association of LPA variants with risk of coronary disease and the implications for lipoprotein(a)-lowering therapies: a Mendelian randomization analysis. JAMA Cardiol 2018;3:619–27.
    Crossref | PubMed
  54. O’Donoghue ML, Fazio S, Giugliano RP, et al. Lipoprotein(a), PCSK9 inhibition, and cardiovascular risk. Circulation 2019;139:1483–92.
    Crossref | PubMed
  55. Lackner C, Cohen JC, Hobbs HH. Molecular definition of the extreme size polymorphism in apolipoprotein(a). Hum Mol Genet 1993;2:933–40.
    Crossref | PubMed
  56. Rhainds D, Brodeur MR, Tardif JC. Lipoprotein (a): when to measure and how to treat? Curr Atheroscler Rep 2021;23:51.
    Crossref | PubMed
  57. Desai NR, Kohli P, Giugliano RP, et al. AMG145, a monoclonal antibody against proprotein convertase subtilisin kexin type 9, significantly reduces lipoprotein(a) in hypercholesterolemic patients receiving statin therapy: an analysis from the LDL-C assessment with proprotein convertase subtilisin kexin type 9 monoclonal antibody inhibition combined with statin therapy (LAPLACE)-Thrombolysis in Myocardial Infarction (TIMI) 57 trial. Circulation 2013;128:962–9.
    Crossref | PubMed
  58. Wilson DP, Jacobson TA, Jones PH, 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.
    Crossref | PubMed
  59. Cholesterol Treatment Trialists’ (CTT) Collaboration, Baigent C, Blackwell L, et al. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet 2010;376:1670–81.
    Crossref | PubMed
  60. Tsimikas S, Witztum JL, Miller ER, et al. High-dose atorvastatin reduces total plasma levels of oxidized phospholipids and immune complexes present on apolipoprotein B-100 in patients with acute coronary syndromes in the MIRACL trial. Circulation 2004;110:1406–12.
    Crossref | PubMed
  61. Rodenburg J, Vissers MN, Wiegman A, et al. Oxidized low-density lipoprotein in children with familial hypercholesterolemia and unaffected siblings: effect of pravastatin. J Am Coll Cardiol 2006;47:1803–10.
    Crossref | PubMed
  62. Capoulade R, Chan KL, Yeang C, et al. Oxidized phospholipids, lipoprotein(a), and progression of calcific aortic valve stenosis. J Am Coll Cardiol 2015;66:1236–46.
    Crossref | PubMed
  63. Choi SH, Chae A, Miller E, et al. Relationship between biomarkers of oxidized low-density lipoprotein, statin therapy, quantitative coronary angiography, and atheroma: volume observations from the REVERSAL (Reversal of Atherosclerosis with Aggressive Lipid Lowering) study. J Am Coll Cardiol 2008;52:24–32.
    Crossref | PubMed
  64. Yoshida H, Shoda T, Yanai H, et al. Effects of pitavastatin and atorvastatin on lipoprotein oxidation biomarkers in patients with dyslipidemia. Atherosclerosis 2013;226:161–4.
    Crossref | PubMed
  65. Tsimikas S, Gordts PLSM, Nora C, et al. Statin therapy increases lipoprotein(a) levels. Eur Heart J 2020;41:2275–84.
    Crossref | PubMed
  66. Kamanna VS, Kashyap ML. Mechanism of action of niacin on lipoprotein metabolism. Curr Atheroscler Rep 2000;2:36–46.
    Crossref | PubMed
  67. HPS2-THRIVE Collaborative Group, Landray MJ, Haynes R, et al. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med 2014;371:203–12.
    Crossref | PubMed
  68. Raal FJ, Hovingh GK, Blom D, et al. Long-term treatment with evolocumab added to conventional drug therapy, with or without apheresis, in patients with homozygous familial hypercholesterolaemia: an interim subset analysis of the open-label TAUSSIG study. Lancet Diabetes Endocrinol 2017;5:280–90.
    Crossref | PubMed
  69. Raal FJ, Giugliano RP, Sabatine MS, et al. Reduction in lipoprotein(a) with PCSK9 monoclonal antibody evolocumab (AMG 145): a pooled analysis of more than 1,300 patients in 4 phase II trials. J Am Coll Cardiol 2014;63:1278–88.
    Crossref | PubMed
  70. Gaudet D, Kereiakes DJ, McKenney JM, et al. Effect of alirocumab, a monoclonal proprotein convertase subtilisin/kexin 9 antibody, on lipoprotein(a) concentrations (a pooled analysis of 150 mg every two weeks dosing from phase 2 trials). Am J Cardiol 2014;114:711–5.
    Crossref | PubMed
  71. Fitzgerald K, White S, Borodovsky A, et al. A highly durable RNAi therapeutic inhibitor of PCSK9. N Engl J Med 2017;376:41–51.
    Crossref | PubMed
  72. Ray KK, Landmesser U, Leiter LA, et al. Inclisiran in patients at high cardiovascular risk with elevated LDL cholesterol. N Engl J Med 2017;376:1430–40.
    Crossref | PubMed
  73. Ray KK, Stoekenbroek RM, Kallend D, et al. Effect of an siRNA therapeutic targeting PCSK9 on atherogenic lipoproteins: prespecified secondary end points in ORION 1. Circulation 2018;138:1304–16.
    Crossref | PubMed
  74. Raal FJ, Santos RD, Blom DJ, et al. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial. Lancet 2010;375:998–1006.
    Crossref | PubMed
  75. Merki E, Graham M, Taleb A, et al. Antisense oligonucleotide lowers plasma levels of apolipoprotein (a) and lipoprotein (a) in transgenic mice. J Am Coll Cardiol 2011;57:1611–21.
    Crossref | PubMed
  76. Graham MJ, Viney N, Crooke RM, Tsimikas S. Antisense inhibition of apolipoprotein (a) to lower plasma lipoprotein (a) levels in humans. J Lipid Res 2016;57:340–51.
    Crossref | PubMed
  77. Viney NJ, van Capelleveen JC, Geary RS, et al. Antisense oligonucleotides targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind, placebo-controlled, dose-ranging trials. Lancet 2016;388:2239–53.
    Crossref | PubMed
  78. Stiekema LCA, Prange KHM, Hoogeveen RM, et al. Potent lipoprotein(a) lowering following apolipoprotein(a) antisense treatment reduces the pro-inflammatory activation of circulating monocytes in patients with elevated lipoprotein(a). Eur Heart J 2020;41:2262–71.
    Crossref | PubMed
  79. Moriarty PM, Gray JV, Gorby LK. Lipoprotein apheresis for lipoprotein(a) and cardiovascular disease. J Clin Lipidol 2019;13:894–900.
    Crossref | PubMed
  80. Leebmann J, Roeseler E, Julius U, et al. Lipoprotein apheresis in patients with maximally tolerated lipid-lowering therapy, lipoprotein(a)-hyperlipoproteinemia, and progressive cardiovascular disease: prospective observational multicenter study. Circulation 2013;128:2567–76.
    Crossref | PubMed
  81. Klingel R, Heibges A, Fassbender C, Pro(a)LiFe-Study Group. Prevention of cardiovascular complications in patients with Lp(a)-hyperlipoproteinemia and progressive cardiovascular disease by long-term lipoprotein apheresis according to German national guidelines. Clin Res Cardiol Suppl 2017;12(Suppl 1):38–43.
    Crossref | PubMed
  82. Howell C, Douglas K, Cho G, et al. Guideline on the clinical use of apheresis procedures for the treatment of patients and collection of cellular therapy products. British Committee for Standards in Haematology. Transfus Med 2015;25:57–78.
    Crossref | PubMed