open access

Vol 9, No 1 (2024)
Review article
Published online: 2024-02-14
Get Citation

Pharmacological methods to lower lipoprotein(a) levels

Natalia Wierzbowska1, Joanna Olejnik-Wojciechowska1, Aleksandra Dąbrowska1, Karolina Żurawska1
·
Medical Research Journal 2024;9(1):96-102.
Affiliations
  1. Department of Physiology, Pomeranian Medical University, Szczecin, Poland

open access

Vol 9, No 1 (2024)
REVIEW ARTICLES
Published online: 2024-02-14

Abstract

Lipoprotein(a) exhibits proatherogenic properties, thus promoting the development of atherosclerotic
cardiovascular disease. Lp(a) levels are genetically determined and relatively constant at the turn of a
patient’s life. Even a single measurement could be an important screening test to distinguish a group of
patients with increased cardiovascular risk. Despite the lack of specific therapies, drugs with potential
effects on reducing Lp(a) levels include ezetimibe, PCSK-9 inhibitors, fibrates, inclisiran, olparisan, aspirin,
tocilizumab or mipomersen.

Although ezetimibe has shown a moderate effect on lowering Lp(a) in monotherapy, its combination with
statins does not provide a significant additional benefit in reducing Lp(a). PCSK-9 inhibitors contribute
to a significant reduction in cardiovascular risk in patients in whom maximum-dose statin therapy fails to
achieve lipoprotein targets. Patients with baseline higher Lp(a) levels receive greater benefit from PCSK9
inhibitor therapy. The use of aspirin to reduce Lp(a) levels could be most significant in rs3798220 carriers,
but the European Atherosclerosis Society does not support the advisability of such a drug. Studies involving
tocilizumab are promising, but data on non-RA groups are lacking. Mipomersen, on the other hand, has
shown significant lipoprotein(a)-lowering effects, but is only used to treat familial hypercholesterolemia
due to the risk of side effects.

The aim of this systematic review was to discuss lipoprotein(a)’s potential as an independent cardiovascular
risk factor and summarize pharmacological approaches available to lower its levels according to
currently available knowledge based on the main findings of randomized clinical studies, review studies
and meta-analyses.

Abstract

Lipoprotein(a) exhibits proatherogenic properties, thus promoting the development of atherosclerotic
cardiovascular disease. Lp(a) levels are genetically determined and relatively constant at the turn of a
patient’s life. Even a single measurement could be an important screening test to distinguish a group of
patients with increased cardiovascular risk. Despite the lack of specific therapies, drugs with potential
effects on reducing Lp(a) levels include ezetimibe, PCSK-9 inhibitors, fibrates, inclisiran, olparisan, aspirin,
tocilizumab or mipomersen.

Although ezetimibe has shown a moderate effect on lowering Lp(a) in monotherapy, its combination with
statins does not provide a significant additional benefit in reducing Lp(a). PCSK-9 inhibitors contribute
to a significant reduction in cardiovascular risk in patients in whom maximum-dose statin therapy fails to
achieve lipoprotein targets. Patients with baseline higher Lp(a) levels receive greater benefit from PCSK9
inhibitor therapy. The use of aspirin to reduce Lp(a) levels could be most significant in rs3798220 carriers,
but the European Atherosclerosis Society does not support the advisability of such a drug. Studies involving
tocilizumab are promising, but data on non-RA groups are lacking. Mipomersen, on the other hand, has
shown significant lipoprotein(a)-lowering effects, but is only used to treat familial hypercholesterolemia
due to the risk of side effects.

The aim of this systematic review was to discuss lipoprotein(a)’s potential as an independent cardiovascular
risk factor and summarize pharmacological approaches available to lower its levels according to
currently available knowledge based on the main findings of randomized clinical studies, review studies
and meta-analyses.

Get Citation

Keywords

atherosclerotic cardiovascular disease; lipoprotein(a); olparisan; tocilizumab; mipomersen

About this article
Title

Pharmacological methods to lower lipoprotein(a) levels

Journal

Medical Research Journal

Issue

Vol 9, No 1 (2024)

Article type

Review article

Pages

96-102

Published online

2024-02-14

Page views

202

Article views/downloads

93

DOI

10.5603/mrj.99088

Bibliographic record

Medical Research Journal 2024;9(1):96-102.

Keywords

atherosclerotic cardiovascular disease
lipoprotein(a)
olparisan
tocilizumab
mipomersen

Authors

Natalia Wierzbowska
Joanna Olejnik-Wojciechowska
Aleksandra Dąbrowska
Karolina Żurawska

References (50)
  1. Jawi MM, Frohlich J, Chan SY. Lipoprotein(a) the insurgent: a new insight into the structure, function, metabolism, pathogenicity, and medications affecting lipoprotein(a) molecule. J Lipids. 2020; 2020: 3491764.
  2. Maranhão RC, Carvalho PO, Strunz CC, et al. Lipoprotein (a): structure, pathophysiology and clinical implications. Arq Bras Cardiol. 2014; 103(1): 76–84.
  3. Riches K, Porter KE. Lipoprotein(a): cellular effects and molecular mechanisms. Cholesterol. 2012; 2012: 923289.
  4. Orfanos P, Fonseca AF, Hu X, et al. Burden of elevated lipoprotein(a) among patients with atherosclerotic cardiovascular disease: Evidence from a systematic literature review and feasibility assessment of meta-analysis. PLoS One. 2023; 18(11): e0294250.
  5. Willeit P, Kiechl S, Kronenberg F, et al. Discrimination and net reclassification of cardiovascular risk with lipoprotein(a). J Am Coll Cardiol. 2014; 64(9): 851–860.
  6. Verbeek R, Sandhu MS, Hovingh GK, et al. Lipoprotein(a) improves cardiovascular risk prediction based on established risk algorithms. J Am Coll Cardiol. 2017; 69(11): 1513–1515.
  7. Moher D, Liberati A, Tetzlaff J, et al. PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med. 2009; 151(4): 264–9, W64.
  8. 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; 43(39): 3925–3946.
  9. Burgess S, Ference BA, Staley JR, et al. European Prospective Investigation Into Cancer and Nutrition–Cardiovascular Disease (EPIC-CVD) Consortium. 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(7): 619–627.
  10. Kronenberg F, Mora S, Stroes ESG, et al. Frequent questions and responses on the 2022 lipoprotein(a) consensus statement of the European Atherosclerosis Society. Atherosclerosis. 2023; 374: 107–120.
  11. Strandkjær N, Hansen MK, Nielsen ST, et al. Lipoprotein(a) Levels at Birth and in Early Childhood: The COMPARE Study. J Clin Endocrinol Metab. 2022; 107(2): 324–335.
  12. Marcovina SM, Albers JJ. Lipoprotein (a) measurements for clinical application. J Lipid Res. 2016; 57(4): 526–537.
  13. Trinder M, Uddin MdM, Finneran P, et al. Clinical utility of lipoprotein(a) and LPA genetic risk score in risk prediction of incident atherosclerotic cardiovascular disease. JAMA Cardiol. 2020 [Epub ahead of print]; 6(3): 1–9.
  14. Nozue T, Michishita I, Mizuguchi I. Effects of ezetimibe on remnant-like particle cholesterol, lipoprotein (a), and oxidized low-density lipoprotein in patients with dyslipidemia. J Atheroscler Thromb. 2010; 17(1): 37–44.
  15. Awad K, Mikhailidis DP, Katsiki N, et al. Lipid and Blood Pressure Meta-Analysis Collaboration (LBPMC) Group. Effect of ezetimibe monotherapy on plasma lipoprotein(a) concentrations in patients with primary hypercholesterolemia: a systematic review and meta-analysis of randomized controlled trials. Drugs. 2018; 78(4): 453–462.
  16. Sahebkar A, Simental-Mendía LE, Pirro M, et al. Impact of ezetimibe on plasma lipoprotein(a) concentrations as monotherapy or in combination with statins: a systematic review and meta-analysis of randomized controlled trials. Sci Rep. 2018; 8(1): 17887.
  17. Watts GF, Chan DC, Somaratne R, et al. Controlled study of the effect of proprotein convertase subtilisin-kexin type 9 inhibition with evolocumab on lipoprotein(a) particle kinetics. Eur Heart J. 2018; 39(27): 2577–2585.
  18. Sabatine MS, Giugliano RP, Keech AC, et al. FOURIER Steering Committee and Investigators. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017; 376(18): 1713–1722.
  19. Schwartz GG, Steg PG, Szarek M, et al. ODYSSEY OUTCOMES Committees and Investigators. Alirocumab and cardiovascular outcomes after acute coronary syndrome. N Engl J Med. 2018; 379(22): 2097–2107.
  20. O'Donoghue ML, Fazio S, Giugliano RP, et al. Lipoprotein(a), PCSK9 inhibition, and cardiovascular risk. Circulation. 2019; 139(12): 1483–1492.
  21. Szarek M, Bittner VA, Aylward P, et al. ODYSSEY OUTCOMES Investigators. Lipoprotein(a) lowering by alirocumab reduces the total burden of cardiovascular events independent of low-density lipoprotein cholesterol lowering: ODYSSEY OUTCOMES trial. Eur Heart J. 2020; 41(44): 4245–4255.
  22. Dai H, Zhu Y, Chen Z, et al. Impact of alirocumab/evolocumab on lipoprotein (a) concentrations in patients with familial hypercholesterolaemia: a systematic review and meta-analysis of randomized controlled trials. Endokrynol Pol. 2023; 74(3): 234–242.
  23. Chennamsetty I, Claudel T, Kostner KM, et al. Farnesoid X receptor represses hepatic human APOA gene expression. J Clin Invest. 2011; 121(9): 3724–3734.
  24. May HT, Anderson JL, Pearson RR, et al. Comparison of effects of simvastatin alone versus fenofibrate alone versus simvastatin plus fenofibrate on lipoprotein subparticle profiles in diabetic patients with mixed dyslipidemia (from the Diabetes and Combined Lipid Therapy Regimen study). Am J Cardiol. 2008; 101(4): 486–489.
  25. Davidson MH, Rooney MW, Drucker J, et al. LCP-AtorFen Investigators. Efficacy and tolerability of atorvastatin/fenofibrate fixed-dose combination tablet compared with atorvastatin and fenofibrate monotherapies in patients with dyslipidemia: a 12-week, multicenter, double-blind, randomized, parallel-group study. Clin Ther. 2009; 31(12): 2824–2838.
  26. Rizos E, Bairaktari E, Ganotakis E, et al. Effect of ciprofibrate on lipoproteins, fibrinogen, renal function, and hepatic enzymes. J Cardiovasc Pharmacol Ther. 2002; 7(4): 219–226.
  27. Sahebkar A, Simental-Mendía LE, Watts GF, et al. Lipid and Blood Pressure Meta-analysis Collaboration (LBPMC) Group. Comparison of the effects of fibrates versus statins on plasma lipoprotein(a) concentrations: a systematic review and meta-analysis of head-to-head randomized controlled trials. BMC Med. 2017; 15(1): 22.
  28. Nishikido T, Ray KK. Inclisiran for the treatment of dyslipidemia. Expert Opin Investig Drugs. 2018; 27(3): 287–294.
  29. Raal FJ, Kallend D, Ray KK, et al. ORION-9 Investigators. Inclisiran for the Treatment of Heterozygous Familial Hypercholesterolemia. N Engl J Med. 2020; 382(16): 1520–1530.
  30. Ray KK, Wright RS, Kallend D, et al. ORION-10 and ORION-11 Investigators. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med. 2020; 382(16): 1507–1519.
  31. Kosmas CE, Muñoz Estrella A, Skavdis A, et al. Inclisiran for the treatment of cardiovascular disease: a short review on the emerging data and therapeutic potential. Ther Clin Risk Manag. 2020; 16: 1031–1037.
  32. Sosnowska B, Surma S, Banach M. Targeted treatment against lipoprotein (a): the coming breakthrough in lipid lowering therapy. Pharmaceuticals (Basel). 2022; 15(12).
  33. Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, et al. AKCEA-APO(a)-LRx Study Investigators. Lipoprotein(a) Reduction in Persons with Cardiovascular Disease. N Engl J Med. 2020; 382(3): 244–255.
  34. Catapano AL, Graham I, De Backer G, et al. ESC Scientific Document Group. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias. Eur Heart J. 2016; 37(39): 2999–3058.
  35. 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: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2019; 73(24): 3168–3209.
  36. O'Donoghue ML, Rosenson RS, Gencer B, et al. OCEAN(a)-DOSE Trial Investigators. Small interfering RNA to reduce lipoprotein(a) in cardiovascular disease. N Engl J Med. 2022; 387(20): 1855–1864.
  37. Sukkari MH, Al-Bast B, Al Tamimi R, et al. Is there a benefit of aspirin therapy for primary prevention to reduce the risk of atherosclerotic cardiovascular disease in patients with elevated Lipoprotein (a)-A review of the evidence. Am J Prev Cardiol. 2023; 15: 100579.
  38. Akaike M, Azuma H, Kagawa A, et al. Effect of aspirin treatment on serum concentrations of lipoprotein(a) in patients with atherosclerotic diseases. Clinical Chemistry. 2002; 48(9): 1454–1459.
  39. Ridker PM, Cook NR, Lee IM, et al. A randomized trial of low-dose aspirin in the primary prevention of cardiovascular disease in women. N Engl J Med. 2005; 352(13): 1293–1304.
  40. Chasman DI, Shiffman D, Zee RYL, et al. Polymorphism in the apolipoprotein(a) gene, plasma lipoprotein(a), cardiovascular disease, and low-dose aspirin therapy. Atherosclerosis. 2009; 203(2): 371–376.
  41. 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(14): 1287–1298.
  42. 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(3): 374–392.
  43. Preuss CV, Anjum F. Tocilizumab. In: StatPearls. Treasure Island (FL): StatPearls Publishing; September 21, 2022. .
  44. Müller N, Schulte DM, Türk K, et al. IL-6 blockade by monoclonal antibodies inhibits apolipoprotein (a) expression and lipoprotein (a) synthesis in humans. J Lipid Res. 2015; 56(5): 1034–1042.
  45. Berthold HK, Laudes M, Krone W, et al. Association between the interleukin-6 promoter polymorphism -174G/C and serum lipoprotein(a) concentrations in humans. PLoS One. 2011; 6(9): e24719.
  46. McInnes IB, Thompson L, Giles JT, et al. Effect of interleukin-6 receptor blockade on surrogates of vascular risk in rheumatoid arthritis: MEASURE, a randomised, placebo-controlled study. Ann Rheum Dis. 2015; 74(4): 694–702.
  47. Yu RZ, Gunawan R, Li Z, et al. No effect on QT intervals of mipomersen, a 2'-O-methoxyethyl modified antisense oligonucleotide targeting ApoB-100 mRNA, in a phase I dose escalation placebo-controlled study, and confirmed by a thorough QT (tQT) study, in healthy subjects. Eur J Clin Pharmacol. 2016; 72(3): 267–275.
  48. 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(12): 2397–2402.
  49. Santos RD, Raal FJ, Catapano AL, et al. Mipomersen, an antisense oligonucleotide to apolipoprotein B-100, reduces lipoprotein(a) in various populations with hypercholesterolemia: results of 4 phase III trials. Arterioscler Thromb Vasc Biol. 2015; 35(3): 689–699.
  50. Fogacci F, Ferri N, Toth PP, et al. Efficacy and Safety of Mipomersen: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. Drugs. 2019; 79(7): 751–766.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk, Poland
tel.:+48 58 320 94 94, fax:+48 58 320 94 60, e-mail: viamedica@viamedica.pl