Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Advances in lipid-lowering therapy through gene-silencing technologies

Key Points

  • Elevated levels of LDL, remnant lipoproteins, and lipoprotein(a) all cause cardiovascular disease, whereas elevated levels of triglyceride-rich lipoproteins can cause acute pancreatitis in some individuals

  • Small molecule and antibody-based drugs such as statins, ezetimibe, and PCSK9 inhibitors efficiently reduce LDL-cholesterol levels and lower the risk of cardiovascular disease

  • In addition, these drugs can modestly reduce remnant lipoproteins, triglyceride-rich lipoproteins, and/or lipoprotein(a) levels

  • These lipoproteins can be reduced using novel gene-silencing approaches (antisense oligonucleotide inhibition and small interfering RNA technology) by targeting proteins that have an important role in lipoprotein production or removal

  • Using gene-silencing approaches, plasma levels of LDL, remnant lipoproteins, lipoprotein(a), and triglyceride-rich lipoproteins can be reduced by 50–90% when treatment is administered 2–12 times yearly

  • Evidence of long-term safety and cardiovascular disease reduction is needed before the widespread use of gene-silencing therapies; however, these drugs might have a role in patients with rare diseases

Abstract

New treatment opportunities are emerging in the field of lipid-lowering therapy through gene-silencing approaches. Both antisense oligonucleotide inhibition and small interfering RNA technology aim to degrade gene mRNA transcripts to reduce protein production and plasma lipoprotein levels. Elevated levels of LDL, remnant lipoproteins, and lipoprotein(a) all cause cardiovascular disease, whereas elevated levels of triglyceride-rich lipoproteins in some patients can cause acute pancreatitis. The levels of each of these lipoproteins can be reduced using gene-silencing therapies by targeting proteins that have an important role in lipoprotein production or removal (for example, the protein products of ANGPTL3, APOB, APOC3, LPA, and PCSK9). Using this technology, plasma levels of these lipoproteins can be reduced by 50–90% with 2–12 injections per year; such dramatic reductions are likely to reduce the incidence of cardiovascular disease or acute pancreatitis in at-risk patients. The reported adverse effects of these new therapies include injection-site reactions, flu-like symptoms, and low blood platelet counts. However, newer-generation drugs are more efficiently delivered to liver cells, requiring lower drug doses, which leads to fewer adverse effects. Although these findings are promising, robust evidence of cardiovascular disease reduction and long-term safety is needed before these gene-silencing technologies can have widespread implementation. Before the availability of such evidence, these drugs might have roles in patients with unmet medical needs through orphan indications.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Approaches to reduce LDL-cholesterol levels.
Figure 2: Apolipoprotein B-containing lipoproteins that cause cardiovascular disease.
Figure 3: Role of the apolipoprotein B-containing lipoproteins in atherosclerotic plaque initiation, progression, rupture, and regression.
Figure 4: Lipoprotein classes and risk of myocardial infarction.
Figure 5: siRNA-based versus antisense oligonucleotide-based approaches.
Figure 6: Efficacy of different approaches to lipid lowering.

Similar content being viewed by others

References

  1. GBD 2016 Causes of Death Collaborators. Global, regional, and national age-sex specific mortality for 264 causes of death, 1980-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet 390, 1151–1210 (2017).

  2. Baigent, 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 366, 1267–1278 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Ridker, P. M. et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N. Engl. J. Med. 359, 2195–2207 (2008).

    Article  CAS  PubMed  Google Scholar 

  4. Baigent, 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 376, 1670–1681 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Cannon, C. P. et al. Ezetimibe added to statin therapy after acute coronary syndromes. N. Engl. J. Med. 372, 2387–2397 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. Sabatine, M. S. et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N. Engl. J. Med. 376, 1713–1722 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Ridker, P. M. et al. Cardiovascular efficacy and safety of bococizumab in high-risk patients. N. Engl. J. Med. 376, 1527–1539 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Ginsberg, H. N. et al. Effects of combination lipid therapy in type 2 diabetes mellitus. N. Engl. J. Med. 362, 1563–1574 (2010).

    Article  PubMed  Google Scholar 

  9. The HPS3/TIMI55–REVEAL Collaborative Group. Effects of anacetrapib in patients with atherosclerotic vascular disease. N. Engl. J. Med. 377, 1217–1227 (2017).

  10. Ridker, P. M. et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 377, 1119–1131 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Brunzell, J. D. & Deeb, S. S. in The Metabolic & Molecular Bases of Inherited Disease (eds Scriver, C. R., Beaudet, A. L., Sly, W. S. & Valle, D.) 2789–2816 (McGraw-Hill, New York, 2001).

    Google Scholar 

  12. Sullenger, B. A. & Nair, S. From the RNA world to the clinic. Science 352, 1417–1420 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. McClorey, G. & Wood, M. J. An overview of the clinical application of antisense oligonucleotides for RNA-targeting therapies. Curr. Opin. Pharmacol. 24, 52–58 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Geyer, P. E., Holdt, L. M., Teupser, D. & Mann, M. Revisiting biomarker discovery by plasma proteomics. Mol. Syst. Biol. 13, 942 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Nordestgaard, B. G. et al. Fasting is not routinely required for determination of a lipid profile: clinical and laboratory implications including flagging at desirable concentration cutpoints — a joint consensus statement from the European Atherosclerosis Society and European Federation of Clinical Chemistry and Laboratory Medicine. Clin. Chem. 62, 930–946 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Havel, J. H. & Kane, J. P. in The Metabolic & Molecular Bases of Inherited Disease (eds Scriver C. R., Beaudet, A. L., Sly, W. S. & Valle, D.) 2705–2716 (McGraw-Hill, New York, 2001).

    Google Scholar 

  17. Nordestgaard, B. G. A. Test in Context: Lipid Profile, Fasting Versus Nonfasting. J. Am. Coll. Cardiol. 70, 1637–1646 (2017).

    Article  PubMed  Google Scholar 

  18. Nordestgaard, B. G. & Varbo, A. Triglycerides and cardiovascular disease. Lancet 384, 626–635 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Nordestgaard, B. G. Triglyceride-rich lipoproteins and atherosclerotic cardiovascular disease: new insights from epidemiology, genetics, and biology. Circ. Res. 118, 547–563 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Pedersen, S. B., Langsted, A. & Nordestgaard, B. G. Nonfasting mild-to-moderate hypertriglyceridemia and risk of acute pancreatitis. JAMA Intern. Med. 176, 1834–1842 (2016).

    Article  PubMed  Google Scholar 

  21. Nordestgaard, B. G., Benn, M., Schnohr, P. & Tybjaerg-Hansen, A. Nonfasting triglycerides and risk of myocardial infarction, ischemic heart disease, and death in men and women. JAMA 298, 299–308 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Varbo, A. et al. Remnant cholesterol as a causal risk factor for ischemic heart disease. J. Am. Coll. Cardiol. 61, 427–436 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Varbo, A., Benn, M., Tybjaerg-Hansen, A. & Nordestgaard, B. G. Elevated remnant cholesterol causes both low-grade inflammation and ischemic heart disease, whereas elevated low-density lipoprotein cholesterol causes ischemic heart disease without inflammation. Circulation 128, 1298–1309 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Jorgensen, A. B. et al. Genetically elevated non-fasting triglycerides and calculated remnant cholesterol as causal risk factors for myocardial infarction. Eur. Heart J. 34, 1826–1833 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Varbo, A., Freiberg, J. J. & Nordestgaard, B. G. Extreme nonfasting remnant cholesterol versus extreme LDL cholesterol as contributors to cardiovascular disease and all-cause mortality in 90000 individuals from the general population. Clin. Chem. 61, 533–543 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Varbo, A. et al. Remnant cholesterol, low-density lipoprotein cholesterol, and blood pressure as mediators from obesity to ischemic heart disease. Circ. Res. 116, 665–673 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Helgadottir, A. et al. Variants with large effects on blood lipids and the role of cholesterol and triglycerides in coronary disease. Nat. Genet. 48, 634–639 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Wurtz, P. et al. Metabolomic profiling of statin use and genetic inhibition of HMG-CoA reductase. J. Am. Coll. Cardiol. 67, 1200–1210 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jepsen, A. M. et al. Increased remnant cholesterol explains part of residual risk of all-cause mortality in 5414 patients with ischemic heart disease. Clin. Chem. 62, 593–604 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Khera, A. V. et al. Association of rare and common variation in the lipoprotein lipase gene with coronary artery disease. JAMA 317, 937–946 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Varbo, A., Freiberg, J. J. & Nordestgaard, B. G. Remnant cholesterol and myocardial infarction in normal weight, overweight, and obese individuals from the Copenhagen General Population Study. Clin. Chem. 64, 219–230 (2018).

    Article  CAS  PubMed  Google Scholar 

  32. Stary, H. C. et al. A definition of advanced types of atherosclerotic lesions and a histological classification of atherosclerosis. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Arterioscler. Thromb. Vasc. Biol. 15, 1512–1531 (1995).

    Article  CAS  PubMed  Google Scholar 

  33. Stary, H. C. Natural history and histological classification of atherosclerotic lesions: an update. Arterioscler. Thromb. Vasc. Biol. 20, 1177–1178 (2000).

    Article  CAS  PubMed  Google Scholar 

  34. Brown, R. A., Shantsila, E., Varma, C. & Lip, G. Y. Current understanding of atherogenesis. Am. J. Med. 130, 268–282 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Gronholdt, M. L. et al. Macrophages are associated with lipid-rich carotid artery plaques, echolucency on B-mode imaging, and elevated plasma lipid levels. J. Vasc. Surg. 35, 137–145 (2002).

    PubMed  Google Scholar 

  36. Bentzon, J. F., Otsuka, F., Virmani, R. & Falk, E. Mechanisms of plaque formation and rupture. Circ. Res. 114, 1852–1866 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Batt, K. V. et al. Differential effects of low-density lipoprotein and chylomicron remnants on lipid accumulation in human macrophages. Exp. Biol. Med. 229, 528–537 (2004).

    Article  CAS  Google Scholar 

  38. Takahashi, S. et al. The very low-density lipoprotein (VLDL) receptor: characterization and functions as a peripheral lipoprotein receptor. J. Atheroscler. Thromb. 11, 200–208 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Nordestgaard, B. G., Stender, S. & Kjeldsen, K. Reduced atherogenesis in cholesterol-fed diabetic rabbits. Giant lipoproteins do not enter the arterial wall. Arteriosclerosis 8, 421–428 (1988).

    Article  CAS  PubMed  Google Scholar 

  40. Nordestgaard, B. G. & Zilversmit, D. B. Large lipoproteins are excluded from the arterial wall in diabetic cholesterol-fed rabbits. J. Lipid Res. 29, 1491–1500 (1988).

    CAS  PubMed  Google Scholar 

  41. Nordestgaard, B. G., Tybjaerg-Hansen, A. & Lewis, B. Influx in vivo of low density, intermediate density, and very low density lipoproteins into aortic intimas of genetically hyperlipidemic rabbits. Roles of plasma concentrations, extent of aortic lesion, and lipoprotein particle size as determinants. Arterioscler. Thromb. 12, 6–18 (1992).

    Article  CAS  PubMed  Google Scholar 

  42. Shaikh, M. et al. Quantitative studies of transfer in vivo of low density, Sf 12–60, and Sf 60–400 lipoproteins between plasma and arterial intima in humans. Arterioscler. Thromb 11, 569–577 (1991).

    Article  CAS  PubMed  Google Scholar 

  43. Nordestgaard, B. G., Wootton, R. & Lewis, B. Selective retention of VLDL, IDL, and LDL in the arterial intima of genetically hyperlipidemic rabbits in vivo. Molecular size as a determinant of fractional loss from the intima-inner media. Arterioscler. Thromb. Vasc. Biol. 15, 534–542 (1995).

    Article  CAS  PubMed  Google Scholar 

  44. Nielsen, L. B., Nordestgaard, B. G., Stender, S., Niendorf, A. & Kjeldsen, K. Transfer of lipoprotein(a) and LDL into aortic intima in normal and in cholesterol-fed rabbits. Arterioscler. Thromb. Vasc. Biol. 15, 1492–1502 (1995).

    Article  CAS  PubMed  Google Scholar 

  45. Nielsen, L. B., Gronholdt, M. L., Schroeder, T. V., Stender, S. & Nordestgaard, B. G. In vivo transfer of lipoprotein(a) into human atherosclerotic carotid arterial intima. Arterioscler. Thromb. Vasc. Biol. 17, 905–911 (1997).

    Article  CAS  PubMed  Google Scholar 

  46. Nordestgaard, B. G. & Langsted, A. Lipoprotein (a) as a cause of cardiovascular disease: insights from epidemiology, genetics, and biology. J. Lipid Res. 57, 1953–1975 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Boffa, M. B. & Koschinsky, M. L. Lipoprotein (a): truly a direct prothrombotic factor in cardiovascular disease? J. Lipid Res. 57, 745–757 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Nielsen, L. B., Stender, S., Kjeldsen, K. & Nordestgaard, B. G. Specific accumulation of lipoprotein(a) in balloon-injured rabbit aorta in vivo. Circ. Res. 78, 615–626 (1996).

    Article  CAS  PubMed  Google Scholar 

  49. Ference, B. A. et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 38, 2459–2472 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Chapman, M. J. et al. Triglyceride-rich lipoproteins and high-density lipoprotein cholesterol in patients at high risk of cardiovascular disease: evidence and guidance for management. Eur. Heart J. 32, 1345–1361 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hegele, R. A. et al. The polygenic nature of hypertriglyceridaemia: implications for definition, diagnosis, and management. Lancet Diabetes Endocrinol. 2, 655–666 (2014).

    Article  CAS  PubMed  Google Scholar 

  52. Nordestgaard, B. G. et al. Lipoprotein(a) as a cardiovascular risk factor: current status. Eur. Heart J. 31, 2844–2853 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kronenberg, F. & Utermann, G. Lipoprotein(a): resurrected by genetics. J. Intern. Med. 273, 6–30 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation 106, 3143–3421 (2002).

  55. Berglund, L. et al. Evaluation and treatment of hypertriglyceridemia: an Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 97, 2969–2989 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Miller, M. et al. Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation 123, 2292–2333 (2011).

    Article  PubMed  Google Scholar 

  57. Catapano, A. L. et al. 2016 ESC/EAS guidelines for the Management of Dyslipidaemias. Eur. Heart J. 37, 2999–3058 (2016).

    Article  PubMed  Google Scholar 

  58. Piepoli, M. F. et al. 2016 European Guidelines on cardiovascular disease prevention in clinical practice: The Sixth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of 10 societies and by invited experts)Developed with the special contribution of the European Association for Cardiovascular Prevention and Rehabilitation (EACPR). Eur. Heart J. 37, 2315–2381 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Lindkvist, B., Appelros, S., Regner, S. & Manjer, J. A prospective cohort study on risk of acute pancreatitis related to serum triglycerides, cholesterol and fasting glucose. Pancreatology 12, 317–324 (2012).

    Article  CAS  PubMed  Google Scholar 

  60. Murphy, M. J., Sheng, X., MacDonald, T. M. & Wei, L. Hypertriglyceridemia and acute pancreatitis. JAMA Intern. Med. 173, 162–164 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Havel, R. J. Pathogenesis, differentiation and management of hypertriglyceridemia. Adv. Intern. Med. 15, 117–154 (1969).

    CAS  PubMed  Google Scholar 

  62. Saharia, P., Margolis, S., Zuidema, G. D. & Cameron, J. L. Acute pancreatitis with hyperlipemia: studies with an isolated perfused canine pancreas. Surgery 82, 60–67 (1977).

    CAS  PubMed  Google Scholar 

  63. Nordstoga, K., Sorby, R., Olivecrona, G., Smith, A. J. & Christophersen, B. Pancreatitis in hyperlipemic mink (Mustela vison). Vet. Pathol. 49, 557–561 (2012).

    Article  CAS  PubMed  Google Scholar 

  64. Crick, F. Central dogma of molecular biology. Nature 227, 561–563 (1970).

    Article  CAS  PubMed  Google Scholar 

  65. Bennett, C. F. & Swayze, E. E. RNA targeting therapeutics: molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annu. Rev. Pharmacol. Toxicol. 50, 259–293 (2010).

    Article  CAS  PubMed  Google Scholar 

  66. Burnett, J. C. & Rossi, J. J. RNA-based therapeutics: current progress and future prospects. Chem. Biol. 19, 60–71 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Watts, J. K. & Corey, D. R. Silencing disease genes in the laboratory and the clinic. J. Pathol. 226, 365–379 (2012).

    Article  CAS  PubMed  Google Scholar 

  68. Huang, Y. Preclinical and clinical advances of GalNAc-decorated nucleic acid therapeutics. Mol. Ther. Nucleic Acids 6, 116–132 (2017).

    Article  CAS  PubMed  Google Scholar 

  69. Prakash, T. P. et al. Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N-acetyl galactosamine improves potency 10-fold in mice. Nucleic Acids Res. 42, 8796–8807 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Nair, J. K. et al. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J. Am. Chem. Soc. 136, 16958–16961 (2014).

    Article  CAS  PubMed  Google Scholar 

  71. Gaudet, D. & Brisson, D. Gene-based therapies in lipidology: current status and future challenges. Curr. Opin. Lipidol. 26, 553–565 (2015).

    Article  CAS  PubMed  Google Scholar 

  72. Wierzbicki, A. S. & Viljoen, A. Anti-sense oligonucleotide therapies for the treatment of hyperlipidaemia. Expert. Opin. Biol. Ther. 16, 1125–1134 (2016).

    Article  CAS  PubMed  Google Scholar 

  73. Lee, R. G., Crosby, J., Baker, B. F., Graham, M. J. & Crooke, R. M. Antisense technology: an emerging platform for cardiovascular disease therapeutics. J. Cardiovasc. Transl Res. 6, 969–980 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Reyes-Soffer, G. et al. Complex effects of inhibiting hepatic apolipoprotein B100 synthesis in humans. Sci. Transl Med. 8, 323ra12 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Panta, R., Dahal, K. & Kunwar, S. Efficacy and safety of mipomersen in treatment of dyslipidemia: a meta-analysis of randomized controlled trials. J. Clin. Lipidol. 9, 217–225 (2015).

    Article  PubMed  Google Scholar 

  76. Raal, F. J. 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 375, 998–1006 (2010).

    Article  CAS  PubMed  Google Scholar 

  77. Duell, P. B. et al. Long-term mipomersen treatment is associated with a reduction in cardiovascular events in patients with familial hypercholesterolemia. J. Clin. Lipidol. 10, 1011–1021 (2016).

    Article  PubMed  Google Scholar 

  78. Hashemi, N. et al. Liver histology during Mipomersen therapy for severe hypercholesterolemia. J. Clin. Lipidol. 8, 606–611 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Cuchel, M. et al. Homozygous familial hypercholesterolaemia: new insights and guidance for clinicians to improve detection and clinical management. A position paper from the Consensus Panel on Familial Hypercholesterolaemia of the European Atherosclerosis Society. Eur. Heart J. 35, 2146–2157 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Wiegman, A. et al. Familial hypercholesterolaemia in children and adolescents: gaining decades of life by optimizing detection and treatment. Eur. Heart J. 36, 2425–2437 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Jorgensen, A. B., Frikke-Schmidt, R., Nordestgaard, B. G. & Tybjaerg-Hansen, A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N. Engl. J. Med. 371, 32–41 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Crosby, J. et al. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N. Engl. J. Med. 371, 22–31 (2014).

    Article  CAS  PubMed  Google Scholar 

  83. Graham, M. J. et al. Antisense oligonucleotide inhibition of apolipoprotein C-III reduces plasma triglycerides in rodents, nonhuman primates, and humans. Circ. Res. 112, 1479–1490 (2013).

    Article  CAS  PubMed  Google Scholar 

  84. Gaudet, D. et al. Antisense inhibition of apolipoprotein C-III in patients with hypertriglyceridemia. N. Engl. J. Med. 373, 438–447 (2015).

    Article  CAS  PubMed  Google Scholar 

  85. Gaudet, D. et al. Targeting APOC3 in the familial chylomicronemia syndrome. N. Engl. J. Med. 371, 2200–2206 (2014).

    Article  CAS  PubMed  Google Scholar 

  86. Digenio, A. et al. Antisense-mediated lowering of plasma apolipoprotein C-III by volanesorsen improves dyslipidemia and insulin sensitivity in type 2 diabetes. Diabetes Care 39, 1408–1415 (2016).

    Article  CAS  PubMed  Google Scholar 

  87. Crooke, S. T. et al. The effects of 2′-O-methoxyethyl containing antisense oligonucleotides on platelets in human clinical trials. Nucleic Acid Ther. 27, 121–129 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Dewey, F. E. et al. Genetic and pharmacologic inactivation of ANGPTL3 and cardiovascular disease. N. Engl. J. Med. 377, 211–221 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Graham, M. J. et al. Cardiovascular and metabolic effects of ANGPTL3 antisense oligonucleotides. N. Engl. J. Med. 377, 222–232 (2017).

    Article  CAS  PubMed  Google Scholar 

  90. Gaudet, D. et al. ANGPTL3 inhibition in homozygous familial hypercholesterolemia. N. Engl. J. Med. 377, 296–297 (2017).

    Article  PubMed  Google Scholar 

  91. Kamstrup, P. R., Tybjaerg-Hansen, A., Steffensen, R. & Nordestgaard, B. G. Genetically elevated lipoprotein(a) and increased risk of myocardial infarction. JAMA 301, 2331–2339 (2009).

    Article  CAS  PubMed  Google Scholar 

  92. Clarke, R. et al. Genetic variants associated with Lp(a) lipoprotein level and coronary disease. N. Engl. J. Med. 361, 2518–2528 (2009).

    Article  CAS  PubMed  Google Scholar 

  93. Thanassoulis, G. et al. Genetic associations with valvular calcification and aortic stenosis. N. Engl. J. Med. 368, 503–512 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Kamstrup, P. R., Tybjaerg-Hansen, A. & Nordestgaard, B. G. Elevated lipoprotein(a) and risk of aortic valve stenosis in the general population. J. Am. Coll. Cardiol. 63, 470–477 (2014).

    Article  CAS  PubMed  Google Scholar 

  95. Graham, M. J., Viney, N., Crooke, R. M. & Tsimikas, S. Antisense inhibition of apolipoprotein (a) to lower plasma lipoprotein (a) levels in humans. J. Lipid Res. 57, 340–351 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Tsimikas, S. et al. Antisense therapy targeting apolipoprotein(a): a randomised, double-blind, placebo-controlled phase 1 study. Lancet 386, 1472–1483 (2015).

    Article  CAS  PubMed  Google Scholar 

  97. Viney, N. J. et al. Antisense oligonucleotides targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind, placebo-controlled, dose-ranging trials. Lancet 388, 2239–2253 (2016).

    Article  CAS  PubMed  Google Scholar 

  98. Mora, S. et al. Lipoprotein(a) and risk of type 2 diabetes. Clin. Chem. 56, 1252–1260 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Kamstrup, P. R. & Nordestgaard, B. G. Lipoprotein(a) concentrations, isoform size, and risk of type 2 diabetes: a Mendelian randomisation study. Lancet Diabetes Endocrinol. 1, 220–227 (2013).

    Article  CAS  PubMed  Google Scholar 

  100. Ding, L. et al. Serum lipoprotein (a) concentrations are inversely associated with T2D, prediabetes, and insulin resistance in a middle-aged and elderly Chinese population. J. Lipid Res. 56, 920–926 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Ye, Z. et al. The association between circulating lipoprotein(a) and type 2 diabetes: is it causal? Diabetes 63, 332–342 (2014).

    Article  CAS  PubMed  Google Scholar 

  102. Tolbus, A. et al. Kringle IV type 2, not low lipoprotein(a), as a cause of diabetes: a novel genetic approach using SNPs associated selectively with lipoprotein(a) concentrations or with Kringle IV type 2 repeats. Clin. Chem. 63, 1866–1876 (2017).

    Article  CAS  PubMed  Google Scholar 

  103. Langsted, A., Kamstrup, P. R. & Nordestgaard, B. G. High lipoprotein(a) and low risk of major bleeding in brain and airways in the general population: a Mendelian randomization study. Clin. Chem. 63, 1714–1723 (2017).

    Article  CAS  PubMed  Google Scholar 

  104. Ishikawa, S. et al. Inverse association between serum lipoprotein(a) and cerebral hemorrhage in the Japanese population. Thromb. Res. 131, e54–e58 (2013).

    Article  CAS  PubMed  Google Scholar 

  105. Fire, A. et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391, 806–811 (1998).

    Article  CAS  PubMed  Google Scholar 

  106. Elbashir, S. M. et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 411, 494–498 (2001).

    Article  CAS  PubMed  Google Scholar 

  107. Abifadel, M. et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat. Genet. 34, 154–156 (2003).

    Article  CAS  PubMed  Google Scholar 

  108. Cohen, J. C., Boerwinkle, E., Mosley, T. H. Jr & Hobbs, H. H. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N. Engl. J. Med. 354, 1264–1272 (2006).

    Article  CAS  PubMed  Google Scholar 

  109. Benn, M. & Nordestgaard, B. G., Grande, P., Schnohr, P. & Tybjaerg-Hansen, A. PCSK9 R46L, low-density lipoprotein cholesterol levels, and risk of ischemic heart disease: 3 independent studies and meta-analyses. J. Am. Coll. Cardiol. 55, 2833–2842 (2010).

    Article  CAS  PubMed  Google Scholar 

  110. Nicholls, S. J. et al. Effect of evolocumab on progression of coronary disease in statin-treated patients: the GLAGOV randomized clinical trial. JAMA 316, 2373–2384 (2016).

    Article  CAS  PubMed  Google Scholar 

  111. Stein, E. A. et al. Effect of a monoclonal antibody to PCSK9 on LDL cholesterol. N. Engl. J. Med. 366, 1108–1118 (2012).

    Article  CAS  PubMed  Google Scholar 

  112. Fitzgerald, K. et al. A highly durable RNAi therapeutic inhibitor of PCSK9. N. Engl. J. Med. 376, 41–51 (2017).

    Article  CAS  PubMed  Google Scholar 

  113. Ray, K. K. et al. Inclisiran in patients at high cardiovascular risk with elevated LDL cholesterol. N. Engl. J. Med. 376, 1430–1440 (2017).

    Article  CAS  PubMed  Google Scholar 

  114. Morrissey, D. V. et al. Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nat. Biotechnol. 23, 1002–1007 (2005).

    Article  CAS  PubMed  Google Scholar 

  115. Jackson, A. L. et al. Position-specific chemical modification of siRNAs reduces “off-target” transcript silencing. RNA 12, 1197–1205 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Garber, K. Alnylam terminates revusiran program, stock plunges. Nat. Biotechnol. 34, 1213–1214 (2016).

    Article  CAS  PubMed  Google Scholar 

  117. Mullard, A. RNAi hits another rut. Nat. Rev. Drug Discov. 15, 738 (2016).

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

All the authors contributed to researching data for the article, discussion of its content, writing the article, and reviewing and editing the manuscript before submission.

Corresponding author

Correspondence to Børge G. Nordestgaard.

Ethics declarations

Competing interests

B.G.N. is a consultant for Amgen, AstraZeneca, Denka Seiken, Dezima Pharma, Ionis Pharmaceuticals, Kowa, Merck, Regeneron Pharmaceuticals, and Sanofi. S.J.N. has received research funding from Amgen, Anthera Pharmaceuticals, AstraZeneca, Cerenis Therapeutics, Eli Lilly, InfraReDx, LipoScience, Novartis, Regeneron Pharmaceuticals, Resverlogix, Roche, Sanofi, and The Medicines Company, and is a consultant for Anthera Pharmaceuticals, AstraZeneca, Boehringer Ingelheim, CSL Behring, Eli Lilly, Esperion Therapeutics, Merck, Omthera Pharmaceuticals, Regeneron Pharmaceuticals, Resverlogix, Sanofi, and Takeda. K.K.R. has received research funding from Amgen, Merck, Pfizer, Regeneron, and Sanofi, and is a consultant for AbbVie, Algorithm, Amgen, AstraZeneca, Boehringer Ingelheim, Cerenis Therapeutics, Cipla, Eli Lilly, Esperion, Ionis Pharmaceuticals, Kowa, Merck, Novo Nordisk, Pfizer, Regeneron, Resverlogix, Sanofi, Takeda, and The Medicines Company. The other authors declare no competing interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nordestgaard, B., Nicholls, S., Langsted, A. et al. Advances in lipid-lowering therapy through gene-silencing technologies. Nat Rev Cardiol 15, 261–272 (2018). https://doi.org/10.1038/nrcardio.2018.3

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrcardio.2018.3

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing