Endocrinol Metab.  2022 Aug;37(4):575-586. 10.3803/EnM.2022.402.

Lipoprotein Lipase: Is It a Magic Target for the Treatment of Hypertriglyceridemia

Affiliations
  • 1Divison of Endocrinology & Metabolism, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
  • 2Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
  • 3Divison of Endocrinology & Metabolism, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea

Abstract

High levels of triglycerides (TG) and triglyceride-rich lipoproteins (TGRLs) confer a residual risk of cardiovascular disease after optimal low-density lipoprotein cholesterol (LDL-C)–lowering therapy. Consensus has been made that LDL-C is a non-arguable primary target for lipid lowering treatment, but the optimization of TGRL for reducing the remnant risk of cardiovascular diseases is urged. Omega-3 fatty acids and fibrates are used to reduce TG levels, but many patients still have high TG and TGRL levels combined with low high-density lipoprotein concentration that need to be ideally treated. Lipoprotein lipase (LPL) is a key regulator for TGs that hydrolyzes TGs to glycerol and free fatty acids in lipoprotein particles for lipid storage and consumption in peripheral organs. A deeper understanding of human genetics has enabled the identification of proteins regulating the LPL activity, which include the apolipoproteins and angiopoietin-like families. Novel therapeutic approach such as antisense oligonucleotides and monoclonal antibodies that regulate TGs have been developed in recent decades. In this article, we focus on the biology of LPL and its modulators and review recent clinical application, including genetic studies and clinical trials of novel therapeutics. Optimization of LPL activity to lower TG levels could eventually reduce incident atherosclerotic cardiovascular disease in conjunction with successful LDL-C reduction.

Keyword

Lipoprotein lipase; Triglycerides; Apolipoproteins; Cholesterol; Cardiovascular diseases; Oligonucleotides, antisense; Angiopoietin-like protein 3; PPAR alpha; Fatty acids, omega-3; Fibric acids

Figure

  • Fig. 1. Overview of the role of lipoprotein lipase in triglyceride metabolism. De novo lipogenesis from the liver results in the secretion of triglycerides in the form of very-low-density lipoprotein cholesterol (VLDL). Dietary fat is transported from intestine to circulation as part of chylomicrons. The triglycerides in chylomicrons and VLDL are hydrolyzed by lipoprotein lipase (LPL). Chylomicron remnants, intermediate-density lipoprotein (IDL) cholesterol, and low-density lipoprotein (LDL) cholesterol are produced as byproducts. LPL is produced from parenchymal cells, including adipose tissue and muscle. LPL is attached to the cell surface via heparan sulfate proteoglycans (HSPGs). Glycosylphosphatidylinositol-anchored high-density lipoprotein binding protein 1 (GPIHBP1) transports LPL from the cell surface to the vascular endothelium. LPL transported to the vascular endothelium hydrolyzes triacylglycerol (TAG) to diacylglycerol (DAG), DAG to monoacylglcerol (MAG), and MAG to glycerol with free fatty acids (FFAs) being released at each step. Parenchymal cells take up FFAs for storage or use FFAs as fuel.

  • Fig. 2. Novel therapeutics that modulate lipoprotein lipase activity. Apolipoproteins (APOs) and angiopoietin-like proteins (ANGPTLs) modulate lipoprotein lipase (LPL) activity: apolipoprotein C2 (APOC2) and APOC5 activate and APOC3, ANGPTL3, ANGPTL4, ANGPTL8 inhibit LPL activity. All of them are produced by the liver, while ANGPTL4 and ANGTPL8 are also produced by adipocytes. Volanesorsen and olezarsen are antisense oligonucleotides (ASOs) targeting APOC3. Evinacumab is a monoclonal antibody (Ab) inhibiting ANGPTL3. Vupanorsen is an ASO targeting ANGPTL3. Peroxisome proliferator-activated receptor alpha (PPARα) activates the transcription of LPL and stimulates LPL activity. Pemafibrate is a PPARα agonist. Omega-3 fatty acids activate both PPARα and LPL.


Cited by  1 articles

Role of Fenofibrate Use in Dyslipidemia and Related Comorbidities in the Asian Population: A Narrative Review
Chaicharn Deerochanawong, Sin Gon Kim, Yu-Cheng Chang
Diabetes Metab J. 2024;48(2):184-195.    doi: 10.4093/dmj.2023.0168.


Reference

1. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: executive summary: a report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines. J Am Coll Cardiol. 2019; 73:3168–209.
2. Rhee EJ, Kim HC, Kim JH, Lee EY, Kim BJ, Kim EM, et al. 2018 Guidelines for the management of dyslipidemia in Korea. J Lipid Atheroscler. 2019; 8:78–131.
Article
3. Sampson UK, Fazio S, Linton MF. Residual cardiovascular risk despite optimal LDL cholesterol reduction with statins: the evidence, etiology, and therapeutic challenges. Curr Atheroscler Rep. 2012; 14:1–10.
Article
4. Santamarina-Fojo S. The familial chylomicronemia syndrome. Endocrinol Metab Clin North Am. 1998; 27:551–67.
Article
5. Meade T, Zuhrie R, Cook C, Cooper J. Bezafibrate in men with lower extremity arterial disease: randomised controlled trial. BMJ. 2002; 325:1139.
Article
6. Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, et al. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet. 2005; 366:1849–61.
Article
7. ACCORD Study Group, Ginsberg HN, Elam MB, Lovato LC, Crouse JR 3rd, Leiter LA, et al. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med. 2010; 362:1563–74.
Article
8. Rubins HB, Robins SJ, Collins D, Fye CL, Anderson JW, Elam MB, et al. Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med. 1999; 341:410–8.
Article
9. Jun M, Foote C, Lv J, Neal B, Patel A, Nicholls SJ, et al. Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet. 2010; 375:1875–84.
Article
10. Kim NH, Kim SG. Fibrates revisited: potential role in cardiovascular risk reduction. Diabetes Metab J. 2020; 44:213–21.
Article
11. Ference BA, Kastelein JJ, Ray KK, Ginsberg HN, Chapman MJ, Packard CJ, et al. Association of triglyceride-lowering LPL variants and LDL-C-lowering LDLR variants with risk of coronary heart disease. JAMA. 2019; 321:364–73.
Article
12. Cohen JC, Boerwinkle E, Mosley TH Jr, Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med. 2006; 354:1264–72.
Article
13. Packard CJ, Boren J, Taskinen MR. Causes and consequences of hypertriglyceridemia. Front Endocrinol (Lausanne). 2020; 11:252.
Article
14. Santos-Baez LS, Ginsberg HN. Hypertriglyceridemia-causes, significance, and approaches to therapy. Front Endocrinol (Lausanne). 2020; 11:616.
Article
15. Choi SH, Ginsberg HN. Increased very low density lipoprotein (VLDL) secretion, hepatic steatosis, and insulin resistance. Trends Endocrinol Metab. 2011; 22:353–63.
Article
16. Nakano T, Nakajima K, Niimi M, Fujita MQ, Nakajima Y, Takeichi S, et al. Detection of apolipoproteins B-48 and B-100 carrying particles in lipoprotein fractions extracted from human aortic atherosclerotic plaques in sudden cardiac death cases. Clin Chim Acta. 2008; 390:38–43.
Article
17. Laufs U, Parhofer KG, Ginsberg HN, Hegele RA. Clinical review on triglycerides. Eur Heart J. 2020; 41:99–109c.
Article
18. Wen Y, Chen YQ, Konrad RJ. The regulation of triacylglycerol metabolism and lipoprotein lipase activity. Adv Biol (Weinh). 2022; Jun. 8. [Epub]. https://doi.org/10.1002/adbi.202200093.
Article
19. Ameis D, Kobayashi J, Davis RC, Ben-Zeev O, Malloy MJ, Kane JP, et al. Familial chylomicronemia (type I hyperlipoproteinemia) due to a single missense mutation in the lipoprotein lipase gene. J Clin Invest. 1991; 87:1165–70.
Article
20. Falko JM. Familial chylomicronemia syndrome: a clinical guide for endocrinologists. Endocr Pract. 2018; 24:756–63.
Article
21. Breckenridge WC, Little JA, Steiner G, Chow A, Poapst M. Hypertriglyceridemia associated with deficiency of apolipoprotein C-II. N Engl J Med. 1978; 298:1265–73.
Article
22. Pennacchio LA, Olivier M, Hubacek JA, Cohen JC, Cox DR, Fruchart JC, et al. An apolipoprotein influencing triglycerides in humans and mice revealed by comparative sequencing. Science. 2001; 294:169–73.
Article
23. Willer CJ, Schmidt EM, Sengupta S, Peloso GM, Gustafsson S, Kanoni S, et al. Discovery and refinement of loci associated with lipid levels. Nat Genet. 2013; 45:1274–83.
Article
24. Olkkonen VM, Sinisalo J, Jauhiainen M. New medications targeting triglyceride-rich lipoproteins: can inhibition of ANGPTL3 or apoC-III reduce the residual cardiovascular risk? Atherosclerosis. 2018; 272:27–32.
Article
25. TG and HDL Working Group of the Exome Sequencing Project, National Heart, Lung, and Blood Institute, Crosby J, Peloso GM, Auer PL, Crosslin DR, Stitziel NO, et al. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med. 2014; 371:22–31.
26. Jorgensen AB, Frikke-Schmidt R, Nordestgaard BG, Tybjaerg-Hansen A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med. 2014; 371:32–41.
27. Gaudet D, Brisson D, Tremblay K, Alexander VJ, Singleton W, Hughes SG, et al. Targeting APOC3 in the familial chylomicronemia syndrome. N Engl J Med. 2014; 371:2200–6.
Article
28. Graham MJ, Lee RG, Bell TA 3rd, Fu W, Mullick AE, Alexander VJ, et al. Antisense oligonucleotide inhibition of apolipoprotein C-III reduces plasma triglycerides in rodents, nonhuman primates, and humans. Circ Res. 2013; 112:1479–90.
Article
29. Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, et al. Volanesorsen and triglyceride levels in familial chylomicronemia syndrome. N Engl J Med. 2019; 381:531–42.
Article
30. Gouni-Berthold I, Alexander VJ, Yang Q, Hurh E, Steinhagen-Thiessen E, Moriarty PM, et al. Efficacy and safety of volanesorsen in patients with multifactorial chylomicronaemia (COMPASS): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Diabetes Endocrinol. 2021; 9:264–75.
31. Paik J, Duggan S. Volanesorsen: first global approval. Drugs. 2019; 79:1349–54.
Article
32. Alexander VJ, Xia S, Hurh E, Hughes SG, O’Dea L, Geary RS, et al. N-acetyl galactosamine-conjugated antisense drug to APOC3 mRNA, triglycerides and atherogenic lipoprotein levels. Eur Heart J. 2019; 40:2785–96.
Article
33. Karwatowska-Prokopczuk E, Tardif JC, Gaudet D, Ballantyne CM, Shapiro MD, Moriarty PM, et al. Effect of olezarsen targeting APOC-III on lipoprotein size and particle number measured by NMR in patients with hypertriglyceridemia. J Clin Lipidol. 2022; Jun. 23. [Epub]. https://doi.org/10.1016/j.jacl.2022.06.005.
Article
34. Li J, Li L, Guo D, Li S, Zeng Y, Liu C, et al. Triglyceride metabolism and angiopoietin-like proteins in lipoprotein lipase regulation. Clin Chim Acta. 2020; 503:19–34.
Article
35. Koishi R, Ando Y, Ono M, Shimamura M, Yasumo H, Fujiwara T, et al. Angptl3 regulates lipid metabolism in mice. Nat Genet. 2002; 30:151–7.
Article
36. Musunuru K, Pirruccello JP, Do R, Peloso GM, Guiducci C, Sougnez C, et al. Exome sequencing, ANGPTL3 mutations, and familial combined hypolipidemia. N Engl J Med. 2010; 363:2220–7.
Article
37. Raal FJ, Rosenson RS, Reeskamp LF, Hovingh GK, Kastelein JJ, Rubba P, et al. Evinacumab for homozygous familial hypercholesterolemia. N Engl J Med. 2020; 383:711–20.
Article
38. Dewey FE, Gusarova V, Dunbar RL, O’Dushlaine C, Schurmann C, Gottesman O, et al. Genetic and pharmacologic inactivation of ANGPTL3 and cardiovascular disease. N Engl J Med. 2017; 377:211–21.
Article
39. Reeskamp LF, Millar JS, Wu L, Jansen H, van Harskamp D, Schierbeek H, et al. ANGPTL3 inhibition with evinacumab results in faster clearance of IDL and LDL apoB in patients with homozygous familial hypercholesterolemia: brief report. Arterioscler Thromb Vasc Biol. 2021; 41:1753–9.
40. Graham MJ, Lee RG, Brandt TA, Tai LJ, Fu W, Peralta R, et al. Cardiovascular and metabolic effects of ANGPTL3 antisense oligonucleotides. N Engl J Med. 2017; 377:222–32.
Article
41. Bergmark BA, Marston NA, Bramson CR, Curto M, Ramos V, Jevne A, et al. Effect of vupanorsen on non-high-density lipoprotein cholesterol levels in statin-treated patients with elevated cholesterol: TRANSLATE-TIMI 70. Circulation. 2022; 145:1377–86.
Article
42. Kliewer SA, Sundseth SS, Jones SA, Brown PJ, Wisely GB, Koble CS, et al. Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma. Proc Natl Acad Sci U S A. 1997; 94:4318–23.
43. Schoonjans K, Peinado-Onsurbe J, Lefebvre AM, Heyman RA, Briggs M, Deeb S, et al. PPARalpha and PPARgamma activators direct a distinct tissue-specific transcriptional response via a PPRE in the lipoprotein lipase gene. EMBO J. 1996; 15:5336–48.
Article
44. Lee Y, Kim BR, Kang GH, Lee GJ, Park YJ, Kim H, et al. The effects of PPAR agonists on atherosclerosis and nonalcoholic fatty liver disease in ApoE-/-FXR-/- Mice. Endocrinol Metab (Seoul). 2021; 36:1243–53.
Article
45. Hertz R, Bishara-Shieban J, Bar-Tana J. Mode of action of peroxisome proliferators as hypolipidemic drugs: suppression of apolipoprotein C-III. J Biol Chem. 1995; 270:13470–5.
46. Pawlak M, Lefebvre P, Staels B. Molecular mechanism of PPARα action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J Hepatol. 2015; 62:720–33.
Article
47. Staels B, Maes M, Zambon A. Fibrates and future PPARalpha agonists in the treatment of cardiovascular disease. Nat Clin Pract Cardiovasc Med. 2008; 5:542–53.
48. Marx N, Sukhova GK, Collins T, Libby P, Plutzky J. PPARalpha activators inhibit cytokine-induced vascular cell adhesion molecule-1 expression in human endothelial cells. Circulation. 1999; 99:3125–31.
Article
49. Wang D, Liu B, Tao W, Hao Z, Liu M. Fibrates for secondary prevention of cardiovascular disease and stroke. Cochrane Database Syst Rev. 2015; 2015:CD009580.
Article
50. Jakob T, Nordmann AJ, Schandelmaier S, Ferreira-Gonzalez I, Briel M. Fibrates for primary prevention of cardiovascular disease events. Cochrane Database Syst Rev. 2016; 11:CD009753.
Article
51. Kim NH, Han KH, Choi J, Lee J, Kim SG. Use of fenofibrate on cardiovascular outcomes in statin users with metabolic syndrome: propensity matched cohort study. BMJ. 2019; 366:l5125.
Article
52. Araki E, Yamashita S, Arai H, Yokote K, Satoh J, Inoguchi T, et al. Effects of pemafibrate, a novel selective PPARα modulator, on lipid and glucose metabolism in patients with type 2 diabetes and hypertriglyceridemia: a randomized, double-blind, placebo-controlled, phase 3 trial. Diabetes Care. 2018; 41:538–46.
Article
53. Arai H, Yamashita S, Yokote K, Araki E, Suganami H, Ishibashi S, et al. Efficacy and safety of pemafibrate versus fenofibrate in patients with high triglyceride and low HDL cholesterol levels: a multicenter, placebo-controlled, double-blind, randomized trial. J Atheroscler Thromb. 2018; 25:521–38.
Article
54. Pradhan AD, Paynter NP, Everett BM, Glynn RJ, Amarenco P, Elam M, et al. Rationale and design of the Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes (PROMINENT) study. Am Heart J. 2018; 206:80–93.
Article
55. Calder PC. Mechanisms of action of (n-3) fatty acids. J Nutr. 2012; 142:592S–9S.
Article
56. Backes J, Anzalone D, Hilleman D, Catini J. The clinical relevance of omega-3 fatty acids in the management of hypertriglyceridemia. Lipids Health Dis. 2016; 15:118.
Article
57. Khan S, Minihane AM, Talmud PJ, Wright JW, Murphy MC, Williams CM, et al. Dietary long-chain n-3 PUFAs increase LPL gene expression in adipose tissue of subjects with an atherogenic lipoprotein phenotype. J Lipid Res. 2002; 43:979–85.
Article
58. Park Y, Harris WS. Omega-3 fatty acid supplementation accelerates chylomicron triglyceride clearance. J Lipid Res. 2003; 44:455–63.
Article
59. Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta. Proc Natl Acad Sci U S A. 1997; 94:4312–7.
60. Kastelein JJ, Maki KC, Susekov A, Ezhov M, Nordestgaard BG, Machielse BN, et al. Omega-3 free fatty acids for the treatment of severe hypertriglyceridemia: the EpanoVa fOr Lowering Very high triglyceridEs (EVOLVE) trial. J Clin Lipidol. 2014; 8:94–106.
Article
61. ASCEND Study Collaborative Group, Bowman L, Mafham M, Wallendszus K, Stevens W, Buck G, et al. Effects of n-3 fatty acid supplements in diabetes mellitus. N Engl J Med. 2018; 379:1540–50.
62. Manson JE, Cook NR, Lee IM, Christen W, Bassuk SS, Mora S, et al. Marine n-3 fatty acids and prevention of cardiovascular disease and cancer. N Engl J Med. 2019; 380:23–32.
Article
63. Nicholls SJ, Lincoff AM, Garcia M, Bash D, Ballantyne CM, Barter PJ, et al. Effect of high-dose omega-3 fatty acids vs corn oil on major adverse cardiovascular events in patients at high cardiovascular risk: the STRENGTH Randomized Clinical Trial. JAMA. 2020; 324:2268–80.
Article
64. Kalstad AA, Myhre PL, Laake K, Tveit SH, Schmidt EB, Smith P, et al. Effects of n-3 fatty acid supplements in elderly patients after myocardial infarction: a randomized, controlled trial. Circulation. 2021; 143:528–39.
Article
65. Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, et al. Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia. N Engl J Med. 2019; 380:11–22.
Article
66. Doi T, Langsted A, Nordestgaard BG. A possible explanation for the contrasting results of REDUCE-IT vs. STRENGTH: cohort study mimicking trial designs. Eur Heart J. 2021; 42:4807–17.
Article
67. Khan SU, Lone AN, Khan MS, Virani SS, Blumenthal RS, Nasir K, et al. Effect of omega-3 fatty acids on cardiovascular outcomes: a systematic review and meta-analysis. EClinicalMedicine. 2021; 38:100997.
Article
68. Endocrinologic & Metabolic Drugs Advisory Committee. FDA Briefing Document: EMDAC Meeting for Volanesorsen (Waylivra). Silver Spring: Food and Drug Administration;2018. [cited 2022 Aug 10]. Available from: https://pink.pharmaintelligence.informa.com/-/media/supporting-documents/pink-sheet/2018/05/waylivrafda_backgrounder.pdf?rev=b873388a6381495c9df19ee1e1895d3c&hash=E57C40AEF44E4C8839C9542AA533EE5B.
69. Gaudet D, Gonciarz M, Shen X. Late Breaker Session 1: A first in-human single ascending dose study of a monoclonal antibody against the ANGPTL3/8 complex in subjects with mixed hyperlipidaemia. In : In: 90th European Atherosclerosis Society Congress; 2022 May 22-25; Milan, IT.
70. Kim K, Ginsberg HN, Choi SH. New, novel lipid-lowering agents for reducing cardiovascular risk: beyond statins. Diabetes Metab J. 2022; 46:517–32.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr