Skip to main content
Log in

Targeting ApoC-III to Reduce Coronary Disease Risk

  • Nonstatin Drugs (E. deGoma, Section Editor)
  • Published:
Current Atherosclerosis Reports Aims and scope Submit manuscript

Abstract

Triglyceride-rich lipoproteins (TRLs) are causal contributors to the risk of developing coronary artery disease (CAD). Apolipoprotein C-III (apoC-III) is a component of TRLs that elevates plasma triglycerides (TGs) through delaying the lipolysis of TGs and the catabolism of TRL remnants. Recent human genetics approaches have shown that heterozygous loss-of-function mutations in APOC3, the gene encoding apoC-III, lower plasma TGs and protect from CAD. This observation has spawned new interest in therapeutic efforts to target apoC-III. Here, we briefly review both currently available as well as developing therapies for reducing apoC-III levels and function to lower TGs and cardiovascular risk. These therapies include existing options including statins, fibrates, thiazolidinediones, omega-3-fatty acids, and niacin, as well as an antisense oligonucleotide targeting APOC3 currently in clinical development. We review the mechanisms of action by which these drugs reduce apoC-III and the current understanding of how reduction in apoC-III may impact CAD risk.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Kastelein JJ. Decade in review—dyslipidaemia: resurgence of targets and compounds to treat dyslipidaemia. Nat Rev Cardiol. 2014;11:629–31.

    Article  CAS  PubMed  Google Scholar 

  2. Baigent C, Keech A, Kearney PM, Blackwell L, Buck G, Pollicino C, et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet. 2005;366:1267–78.

    Article  CAS  Google Scholar 

  3. Cholesterol Treatment Trialists C, Baigent C, Blackwell L, Emberson J, Holland LE, Reith C, 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.

    Article  Google Scholar 

  4. Boekholdt SM, Arsenault BJ, Mora S, Pedersen TR, LaRosa JC, Nestel PJ, et al. Association of LDL cholesterol, non-HDL cholesterol, and apolipoprotein B levels with risk of cardiovascular events among patients treated with statins: a meta-analysis. JAMA. 2012;307:1302–9.

    Article  CAS  PubMed  Google Scholar 

  5. Nordestgaard BG, Benn M, Schnohr P, Tybjaerg-Hansen A. Nonfasting triglycerides and risk of myocardial infarction, ischemic heart disease, and death in men and women. JAMA. 2007;298:299–308. This large prospective study in almost 14,000 subjects demonstrated the significant association of nonfasted TGs and remnant cholesterol with risk of cardiovascular morbidity and mortality.

    Article  CAS  PubMed  Google Scholar 

  6. Sarwar N, Danesh J, Eiriksdottir G, Sigurdsson G, Wareham N, Bingham S, et al. Triglycerides and the risk of coronary heart disease: 10,158 incident cases among 262,525 participants in 29 Western prospective studies. Circulation. 2007;115:450–8. This meta-analysis of data from more than 10,000 participants demonstrated the significant association of triglycerides with coronary heart disease risk.

    Article  CAS  PubMed  Google Scholar 

  7. Thorin E. Vascular disease risk in patients with hypertriglyceridemia: endothelial progenitor cells, oxidative stress, accelerated senescence, and impaired vascular repair. Can J Cardiol. 2011;27:538–40.

    Article  PubMed  Google Scholar 

  8. Yu KC, Cooper AD. Postprandial lipoproteins and atherosclerosis. Front Biosci. 2001;6:D332–354.

    Article  CAS  PubMed  Google Scholar 

  9. Schwartz EA, Reaven PD. Lipolysis of triglyceride-rich lipoproteins, vascular inflammation, and atherosclerosis. Biochim Biophys Acta. 2012;1821:858–66.

    Article  CAS  PubMed  Google Scholar 

  10. Carey VJ, Bishop L, Laranjo N, Harshfield BJ, Kwiat C, Sacks FM. Contribution of high plasma triglycerides and low high-density lipoprotein cholesterol to residual risk of coronary heart disease after establishment of low-density lipoprotein cholesterol control. Am J Cardiol. 2010;106:757–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Faergeman O, Holme I, Fayyad R, Bhatia S, Grundy SM, Kastelein JJ, et al. Plasma triglycerides and cardiovascular events in the Treating to New Targets and Incremental Decrease in End-Points through Aggressive Lipid Lowering trials of statins in patients with coronary artery disease. Am J Cardiol. 2009;104:459–63.

    Article  CAS  PubMed  Google Scholar 

  12. Miller M, Cannon CP, Murphy SA, Qin J, Ray KK, Braunwald E, et al. Impact of triglyceride levels beyond low-density lipoprotein cholesterol after acute coronary syndrome in the PROVE IT-TIMI 22 trial. J Am Coll Cardiol. 2008;51:724–30.

    Article  CAS  PubMed  Google Scholar 

  13. Global Lipids Genetics Consortium, Willer CJ, Schmidt EM, Sengupta S, Peloso GM, Gustafsson S, et al. Discovery and refinement of loci associated with lipid levels. Nat Genet. 2013;45:1274–83.

    Article  Google Scholar 

  14. Do R, Willer CJ, Schmidt EM, Sengupta S, Gao C, Peloso GM, et al. Common variants associated with plasma triglycerides and risk for coronary artery disease. Nat Genet. 2013;45:1345–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Rip J, Nierman MC, Ross CJ, Jukema JW, Hayden MR, Kastelein JJ, et al. Lipoprotein lipase S447X: a naturally occurring gain-of-function mutation. Arterioscler Thromb Vasc Biol. 2006;26:1236–45.

    Article  CAS  PubMed  Google Scholar 

  16. Varbo A, Benn M, Tybjaerg-Hansen A, Jorgensen AB, Frikke-Schmidt R, Nordestgaard BG. Remnant cholesterol as a causal risk factor for ischemic heart disease. J Am Coll Cardiol. 2013;61:427–36.

    Article  CAS  PubMed  Google Scholar 

  17. Wittrup HH, Tybjaerg-Hansen A, Nordestgaard BG. Lipoprotein lipase mutations, plasma lipids and lipoproteins, and risk of ischemic heart disease. A meta-analysis Circulation. 1999;99:2901–7.

    Article  CAS  PubMed  Google Scholar 

  18. Gagne SE, Larson MG, Pimstone SN, Schaefer EJ, Kastelein JJ, Wilson PW, et al. A common truncation variant of lipoprotein lipase (Ser447X) confers protection against coronary heart disease: the Framingham Offspring Study. Clin Genet. 1999;55:450–4.

    Article  CAS  PubMed  Google Scholar 

  19. Do R, Stitziel NO, Won HH, Jorgensen AB, Duga S, Angelica Merlini P, et al. Exome sequencing identifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction. Nature. 2015;518:102–6.

    Article  CAS  PubMed  Google Scholar 

  20. Myocardial Infarction, G., and Investigators, C.A.E.C. 2016. Coding Variation in ANGPTL4, LPL, and SVEP1 and the Risk of Coronary Disease. N Engl J Med

  21. Dewey, F.E., Gusarova, V., O’Dushlaine, C., Gottesman, O., Trejos, J., Hunt, C., Van Hout, C.V., Habegger, L., Buckler, D., Lai, K.V., et al. 2016. Inactivating Variants in ANGPTL4 and Risk of Coronary Artery Disease. N Engl J Med.

  22. Kersten S. Physiological regulation of lipoprotein lipase. Biochim Biophys Acta. 2014;1841:919–33.

    Article  CAS  PubMed  Google Scholar 

  23. Pollin TI, Damcott CM, Shen H, Ott SH, Shelton J, Horenstein RB, et al. A null mutation in human APOC3 confers a favorable plasma lipid profile and apparent cardioprotection. Science. 2008;322:1702–5. This is the initial study of genetic loss-of-function in APOC3 showing that heterozygous APOC3 deficiency in humans may correlate not only with reduced TGs but also reduced atherosclerosis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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. This is one of two large sequencing studies from 2014 showing that coding variants in APOC3 that confer loss-of-function cause lower TGs, higher HDL-C, minimal impact on LDL-C but protection from ischemic vascular diseases.

    Article  PubMed  Google Scholar 

  25. Tg, Hdl Working Group of the Exome Sequencing Project, N.H.L., Blood, I., Crosby, J., Peloso, G.M., Auer, P.L., Crosslin, D.R., Stitziel, N.O., Lange, L.A., Lu, Y., et al. 2014. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med 371:22–31. This study is the second of two large sequencing efforts in 2014 that showed that genetic inactivation of APOC3 through loss-of-function coding variants protects from coronary disease risk despite a minimal impact on LDL-C but a lifelong reduction in TGs.

  26. Aalto-Setala K, Fisher EA, Chen X, Chajek-Shaul T, Hayek T, Zechner R, et al. Mechanism of hypertriglyceridemia in human apolipoprotein (apo) CIII transgenic mice. Diminished very low density lipoprotein fractional catabolic rate associated with increased apo CIII and reduced apo E on the particles. J Clin Invest. 1992;90:1889–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Aalto-Setala K, Weinstock PH, Bisgaier CL, Wu L, Smith JD, Breslow JL. Further characterization of the metabolic properties of triglyceride-rich lipoproteins from human and mouse apoC-III transgenic mice. J Lipid Res. 1996;37:1802–11.

    CAS  PubMed  Google Scholar 

  28. Qin W, Sundaram M, Wang Y, Zhou H, Zhong S, Chang CC, et al. Missense mutation in APOC3 within the C-terminal lipid binding domain of human ApoC-III results in impaired assembly and secretion of triacylglycerol-rich very low density lipoproteins: evidence that ApoC-III plays a major role in the formation of lipid precursors within the microsomal lumen. J Biol Chem. 2011;286:27769–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Goldstein JL, Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell. 2015;161:161–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Dallinga-Thie, G.M., Berk, P., II, Bootsma, A.H., Jansen, H., and Diabetes Atorvastatin Lipid intervention Study, G. Atorvastatin decreases apolipoprotein C-III in apolipoprotein B-containing lipoprotein and HDL in type 2 diabetes: a potential mechanism to lower plasma triglycerides. Diabetes Care. 2004;27:1358–64.

    Article  Google Scholar 

  31. Ooi EM, Watts GF, Chan DC, Chen MM, Nestel PJ, Sviridov D, et al. Dose-dependent effect of rosuvastatin on VLDL-apolipoprotein C-III kinetics in the metabolic syndrome. Diabetes Care. 2008;31:1656–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Chan DC, Watts GF, Ooi EM, Ji J, Johnson AG, Barrett PH. Atorvastatin and fenofibrate have comparable effects on VLDL-apolipoprotein C-III kinetics in men with the metabolic syndrome. Arterioscler Thromb Vasc Biol. 2008;28:1831–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ginsberg HN, Ramakrishnan R. Kinetic studies of the metabolism of rapidly exchangeable apolipoproteins may leave investigators and readers with exchangeable results. Arterioscler Thromb Vasc Biol. 2008;28:1685–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chan DC, Nguyen MN, Watts GF, Ooi EM, Barrett PH. Effects of atorvastatin and n-3 fatty acid supplementation on VLDL apolipoprotein C-III kinetics in men with abdominal obesity. Am J Clin Nutr. 2010;91:900–6.

    Article  CAS  PubMed  Google Scholar 

  35. Sando KR, Knight M. Nonstatin therapies for management of dyslipidemia: a review. Clin Ther. 2015;37:2153–79.

    Article  CAS  PubMed  Google Scholar 

  36. Sahebkar A, Chew GT, Watts GF. Recent advances in pharmacotherapy for hypertriglyceridemia. Prog Lipid Res. 2014;56:47–66.

    Article  CAS  PubMed  Google Scholar 

  37. Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Saito Y, Ishikawa Y, et al. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet. 2007;369:1090–8.

    Article  CAS  PubMed  Google Scholar 

  38. Saito Y, Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Ishikawa Y, et al. Effects of EPA on coronary artery disease in hypercholesterolemic patients with multiple risk factors: sub-analysis of primary prevention cases from the Japan EPA Lipid Intervention Study (JELIS). Atherosclerosis. 2008;200:135–40.

    Article  CAS  PubMed  Google Scholar 

  39. Kwak, S.M., Myung, S.K., Lee, Y.J., Seo, H.G., and Korean Meta-analysis Study, G. Efficacy of omega-3 fatty acid supplements (eicosapentaenoic acid and docosahexaenoic acid) in the secondary prevention of cardiovascular disease: a meta-analysis of randomized, double-blind, placebo-controlled trials. Arch Intern Med. 2012;172:686–94.

    Article  Google Scholar 

  40. Rizos EC, Ntzani EE, Bika E, Kostapanos MS, Elisaf MS. Association between omega-3 fatty acid supplementation and risk of major cardiovascular disease events: a systematic review and meta-analysis. JAMA. 2012;308:1024–33.

    Article  CAS  PubMed  Google Scholar 

  41. 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  PubMed  Google Scholar 

  42. Maki KC, Bays HE, Dicklin MR, Johnson SL, Shabbout M. Effects of prescription omega-3-acid ethyl esters, coadministered with atorvastatin, on circulating levels of lipoprotein particles, apolipoprotein CIII, and lipoprotein-associated phospholipase A2 mass in men and women with mixed dyslipidemia. J Clin Lipidol. 2011;5:483–92.

    Article  PubMed  Google Scholar 

  43. Kersten S. Peroxisome proliferator activated receptors and lipoprotein metabolism. PPAR Res. 2008;2008:132960.

    Article  PubMed  Google Scholar 

  44. Haubenwallner S, Essenburg AD, Barnett BC, Pape ME, DeMattos RB, Krause BR, et al. Hypolipidemic activity of select fibrates correlates to changes in hepatic apolipoprotein C-III expression: a potential physiologic basis for their mode of action. J Lipid Res. 1995;36:2541–51.

    CAS  PubMed  Google Scholar 

  45. Staels B, Vu-Dac N, Kosykh VA, Saladin R, Fruchart JC, Dallongeville J, et al. Fibrates downregulate apolipoprotein C-III expression independent of induction of peroxisomal acyl coenzyme A oxidase. A potential mechanism for the hypolipidemic action of fibrates. J Clin Invest. 1995;95:705–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. 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.

    Article  CAS  PubMed  Google Scholar 

  47. Malmendier CL, Lontie JF, Delcroix C, Dubois DY, Magot T, De Roy L. Apolipoproteins C-II and C-III metabolism in hypertriglyceridemic patients. Effect of a drastic triglyceride reduction by combined diet restriction and fenofibrate administration. Atherosclerosis. 1989;77:139–49.

    Article  CAS  PubMed  Google Scholar 

  48. Frick MH, Elo O, Haapa K, Heinonen OP, Heinsalmi P, Helo P, et al. Helsinki Heart Study: primary-prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety of treatment, changes in risk factors, and incidence of coronary heart disease. N Engl J Med. 1987;317:1237–45.

    Article  CAS  PubMed  Google Scholar 

  49. 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  CAS  PubMed  Google Scholar 

  50. 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  CAS  PubMed  Google Scholar 

  51. Group, A.S., Ginsberg, H.N., Elam, M.B., Lovato, L.C., Crouse, J.R., 3rd, Leiter, L.A., Linz, P., Friedewald, W.T., Buse, J.B., Gerstein, H.C., et al. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med. 2010;362:1563–74.

    Article  Google Scholar 

  52. Reyes-Soffer G, Ngai CI, Lovato L, Karmally W, Ramakrishnan R, Holleran S, et al. Effect of combination therapy with fenofibrate and simvastatin on postprandial lipemia in the ACCORD lipid trial. Diabetes Care. 2013;36:422–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Soccio RE, Chen ER, Lazar MA. Thiazolidinediones and the promise of insulin sensitization in type 2 diabetes. Cell Metab. 2014;20:573–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Soccio RE, Chen ER, Rajapurkar SR, Safabakhsh P, Marinis JM, Dispirito JR, et al. Genetic variation determines PPARγ function and anti-diabetic drug response in vivo. Cell. 2015;162:33–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Yki-Jarvinen H. Thiazolidinediones and the liver in humans. Curr Opin Lipidol. 2009;20:477–83.

    Article  PubMed  Google Scholar 

  56. Samaha FF, Szapary PO, Iqbal N, Williams MM, Bloedon LT, Kochar A, et al. Effects of rosiglitazone on lipids, adipokines, and inflammatory markers in nondiabetic patients with low high-density lipoprotein cholesterol and metabolic syndrome. Arterioscler Thromb Vasc Biol. 2006;26:624–30.

    Article  CAS  PubMed  Google Scholar 

  57. Szapary PO, Bloedon LT, Samaha FF, Duffy D, Wolfe ML, Soffer D, et al. Effects of pioglitazone on lipoproteins, inflammatory markers, and adipokines in nondiabetic patients with metabolic syndrome. Arterioscler Thromb Vasc Biol. 2006;26:182–8.

    Article  CAS  PubMed  Google Scholar 

  58. Nagashima K, Lopez C, Donovan D, Ngai C, Fontanez N, Bensadoun A, et al. Effects of the PPARγ agonist pioglitazone on lipoprotein metabolism in patients with type 2 diabetes mellitus. J Clin Invest. 2005;115:1323–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Schuster H, Fagerberg B, Edwards S, Halmos T, Lopatynski J, Stender S, et al. Tesaglitazar, a dual peroxisome proliferator-activated receptor α/γ agonist, improves apolipoprotein levels in non-diabetic subjects with insulin resistance. Atherosclerosis. 2008;197:355–62.

    Article  CAS  PubMed  Google Scholar 

  60. Wise A, Foord SM, Fraser NJ, Barnes AA, Elshourbagy N, Eilert M, et al. Molecular identification of high and low affinity receptors for nicotinic acid. J Biol Chem. 2003;278:9869–74.

    Article  CAS  PubMed  Google Scholar 

  61. Soga T, Kamohara M, Takasaki J, Matsumoto S, Saito T, Ohishi T, et al. Molecular identification of nicotinic acid receptor. Biochem Biophys Res Commun. 2003;303:364–9.

    Article  CAS  PubMed  Google Scholar 

  62. Tunaru S, Kero J, Schaub A, Wufka C, Blaukat A, Pfeffer K, et al. PUMA-G and HM74 are receptors for nicotinic acid and mediate its anti-lipolytic effect. Nat Med. 2003;9:352–5.

    Article  CAS  PubMed  Google Scholar 

  63. Carlson LA. Nicotinic acid: the broad-spectrum lipid drug. A 50th anniversary review. J Intern Med. 2005;258:94–114.

    Article  CAS  PubMed  Google Scholar 

  64. Carlson LA, Havel RJ, Ekelund LG, Holmgren A. Effect of nicotinic acid on the turnover rate and oxidation of the free fatty acids of plasma in man during exercise. Metabolism. 1963;12:837–45.

    CAS  PubMed  Google Scholar 

  65. Carlson LA, Oro L. The effect of nicotinic acid on the plasma free fatty acid; demonstration of a metabolic type of sympathicolysis. Acta Med Scand. 1962;172:641–5.

    Article  CAS  PubMed  Google Scholar 

  66. Carlson LA, Ostman J. Inhibition of the mobilization of free fatty acids from adipose tissue in diabetes. II. Effect of nicotinic acid and acetylsalicylate on blood glucose in human diabetics. Acta Med Scand. 1965;178:71–9.

    Article  CAS  PubMed  Google Scholar 

  67. Carlson LA, Ostman J. Inhibition of the mobilization of free fatty acids from adipose tissue in diabetes. I. Effect of nicotinic acid on the alloxan-diabetic state in rats. Acta Med Scand. 1965;177:631–7.

    Article  CAS  PubMed  Google Scholar 

  68. Duncan CH, Best MM, Robertson GL. A comparison of the effects of ethyl chlorophenoxyisobutyrate and nicotinic acid on plasma free fatty acids. Lancet. 1965;1:191–3.

    Article  CAS  PubMed  Google Scholar 

  69. Froeberg S, Oroe L. The effects of nicotinic acid, phentolamine and nethalide on the plasma free fatty acids and the blood pressure in the dog. A comparative study. Acta Med Scand. 1963;174:635–41.

    Article  CAS  PubMed  Google Scholar 

  70. Havel RJ, Carlson LA, Ekelund LG, Holmgren A. Studies on the relation between mobilization of free fatty acids and energy metabolism in man: effects of norepinephrine and nicotinic acid. Metabolism. 1964;13:1402–12.

    Article  CAS  PubMed  Google Scholar 

  71. Nestel PJ. Plasma triglyceride concentration and plasma free fatty acid changes in response to norepinephrine in man. J Clin Invest. 1964;43:77–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Hernandez C, Molusky M, Li Y, Li S, Lin JD. Regulation of hepatic APOC3 expression by PGC-1beta mediates hypolipidemic effect of nicotinic acid. Cell Metab. 2010;12:411–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Lauring B, Taggart AK, Tata JR, Dunbar R, Caro L, Cheng K, et al. Niacin lipid efficacy is independent of both the niacin receptor GPR109A and free fatty acid suppression. Sci Transl Med. 2012;4:148ra115.

    Article  PubMed  Google Scholar 

  74. Yovos JG, Patel ST, Falko JM, Newman HA, Hill DS. Effects of nicotinic acid therapy on plasma lipoproteins and very low density lipoprotein apoprotein C subspecies in hyperlipoproteinemia. J Clin Endocrinol Metab. 1982;54:1210–5.

    Article  CAS  PubMed  Google Scholar 

  75. Investigators A-H, Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med. 2011;365:2255–67.

    Article  Google Scholar 

  76. Group, H.T.C, Landray MJ, Haynes R, Hopewell JC, Parish S, Aung T, et al. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med. 2014;371:203–12.

    Article  Google Scholar 

  77. Anderson TJ, Boden WE, Desvigne-Nickens P, Fleg JL, Kashyap ML, McBride R, et al. Safety profile of extended-release niacin in the AIM-HIGH trial. N Engl J Med. 2014;371:288–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kurreck J. Antisense technologies. Improvement through novel chemical modifications. Eur J Biochem. 2003;270:1628–44.

    Article  CAS  PubMed  Google Scholar 

  79. Crooke ST. Progress in antisense technology: the end of the beginning. Methods Enzymol. 2000;313:3–45.

    Article  CAS  PubMed  Google Scholar 

  80. Geary RS, Norris D, Yu R, Bennett CF. Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides. Adv Drug Deliv Rev. 2015;87:46–51.

    Article  CAS  PubMed  Google Scholar 

  81. Brown DA, Kang SH, Gryaznov SM, DeDionisio L, Heidenreich O, Sullivan S, et al. Effect of phosphorothioate modification of oligodeoxynucleotides on specific protein binding. J Biol Chem. 1994;269:26801–5.

    CAS  PubMed  Google Scholar 

  82. Yu RZ, Kim TW, Hong A, Watanabe TA, Gaus HJ, Geary RS. Cross-species pharmacokinetic comparison from mouse to man of a second-generation antisense oligonucleotide, ISIS 301012, targeting human apolipoprotein B-100. Drug Metab Dispos. 2007;35:460–8.

    Article  CAS  PubMed  Google Scholar 

  83. Agarwala A, Jones P, Nambi V. The role of antisense oligonucleotide therapy in patients with familial hypercholesterolemia: risks, benefits, and management recommendations. Curr Atheroscler Rep. 2015;17:467.

    Article  PubMed  Google Scholar 

  84. Graham MJ, Lee RG, Bell III TA, 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. This study provided comprehensive preclinical evaluation and phase I study of the anti-APOC3 antisense oligonucleotide, the newest apoC-III focused therapy in development. It showed in mice, rats, nonhuman primates and humans that ASO treatment was safe and efficacious in reducing circulating apoC-III levels.

  85. Duivenvoorden I, Teusink B, Rensen PC, Romijn JA, Havekes LM, Voshol PJ. Apolipoprotein C3 deficiency results in diet-induced obesity and aggravated insulin resistance in mice. Diabetes. 2005;54:664–71.

    Article  CAS  PubMed  Google Scholar 

  86. Sundaram M, Zhong S, Bou Khalil M, Zhou H, Jiang ZG, Zhao Y, et al. Functional analysis of the missense APOC3 mutation Ala23Thr associated with human hypotriglyceridemia. J Lipid Res. 2010;51:1524–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Sundaram M, Zhong S, Bou Khalil M, Links PH, Zhao Y, Iqbal J, et al. Expression of apolipoprotein C-III in McA-RH7777 cells enhances VLDL assembly and secretion under lipid-rich conditions. J Lipid Res. 2010;51:150–61.

    Article  PubMed  PubMed Central  Google Scholar 

  88. 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. This small clinical study was the first to assess the APOC3 ASO in hypertriglyceridemic patients. It showed that apoC-III inhibition in patients with familial chylomicronemia syndrome genetically lacking LPL potently lowered TG levels, suggesting that apoC-III modulates TGs at least in part by LPL-independent mechanisms.

  89. Gaudet D, Alexander VJ, Baker BF, Brisson D, Tremblay K, Singleton W, et al. Antisense inhibition of apolipoprotein C-III in patients with hypertriglyceridemia. N Engl J Med. 2015;373:438–47. This phase II study of the APOC3 ASO in 57 hypertriglyceridemic patients showed that the ASO was efficacious in reducing apoC-III levels and TG while raising HDL-C.

    Article  CAS  PubMed  Google Scholar 

  90. Yang, X., Lee, S.R., Choi, Y.S., Alexander, V.J., Digenio, A., Yang, Q., Miller, Y.I., Witztum, J.L., and Tsimikas, S. 2016. Reduction in lipoprotein-associated apoC-III levels following volanesorsen therapy: phase 2 randomized trial results. J Lipid Res

  91. Digenio, A., Dunbar, R.L., Alexander, V.J., Hompesch, M., Morrow, L., Lee, R.G., Graham, M.J., Hughes, S.G., Yu, R., Singleton, W., et al. 2016. Antisense-mediated lowering of plasma apolipoprotein C-III by volanesorsen improves dyslipidemia and insulin sensitivity in type 2 diabetes. Diabetes Care.

  92. Kevin Fitzgerald, A.B., William Querbes, Jessica Sutherland, Renta Hutabarat, Stuart Milstein, Satya Kuchimanchi, Rajeev Kallanthottathil, Klaus Charisse, Kristina Yucias, Abigail Liebow, Andrew Sprague, Martin Maier, David Kallend2 Jay Horton, Amy Simon. 2014. A subcutaneous, potent and durable RNAi platform targeting metabolic diseases, genes PCSK9, APOC3 and ANGPLT3. In Arterioscler Thromb Vasc Biol.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daniel J. Rader.

Ethics declarations

Conflict of Interest

Sumeet A. Khetarpal, Arman Qamar, John S. Millar, and Daniel J. Rader declare that they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

This article is part of the Topical Collection on Nonstatin Drugs

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Khetarpal, S.A., Qamar, A., Millar, J.S. et al. Targeting ApoC-III to Reduce Coronary Disease Risk. Curr Atheroscler Rep 18, 54 (2016). https://doi.org/10.1007/s11883-016-0609-y

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11883-016-0609-y

Keywords

Navigation