5,557

The Role of PCSK9 in Lipid Metabolism and its Relationship to New Therapies for Lowering Cholesterol and Reducing Cardiac Disease

Yipin Han, Monte S. Willis

Yipin Han, North Carolina State University, Department of Engineering, Raleigh, NC, the United States
Monte S. Willis, University of North Carolina at Chapel Hill, McAllister Heart Institute, Department of Pathology & Laboratory Medicine and Pharmacology, Chapel Hill, NC 27599-7525, the United States

Correspondence to: Monte S. Willis, MD, PhD, MBA, Vice Chair of Academic Affairs, Department of Pathology, Associate Professor, McAllister Heart Institute, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, 27599, the United States.
Email: monte_willis@med.unc.edu
Telephone: +1-919-843-1938
Received: May 22, 2015
Revised: August 19, 2015
Accepted: August 23, 2015
Published online: October 10, 2015

ABSTRACT

The treatment of atherosclerosis includes changing lifestyle (e.g. changing diet and exercise regimens, losing weight, stopping smoking, managing stress), performing medical procedures (e.g. bypass surgery) and medical interventions involving primarily statins. Despite excellent safety profiles, the major limitation of statins is their muscle and liver toxicity that occurs in some patients. Since non-statin therapies only modestly reduce LDL cholesterol, alternatives are needed for patients not tolerating statins or in clinical cases of familial hypercholesterolemia where even statins are ineffective. Over the past 12 years, an amazing series of studies have identified proprotein convertase subtilisin/kexin type-9 (PCSK9) as a new target to treat high cholesterol that is the underlying cause of atherosclerosis and cardiovascular disease. The liver-derived PCSK9, a gene found on the short arm of chromosome 1 that encodes a 692 amino acid protein, was initially discovered as neural apoptosis-regulated convertase 1 (NARC-1) in cancer cell lines and implicated in the differentiation of cortical neurons in 2003. Subsequently, neutralizing PCSK9 antibodies were developed and proven clinically effective in pre-clinical studies. Since then, Phase I, II, and most recently Phase III human trials have tested applied anti-PCSK9 antibodies to patients intolerant of statins and familial hypercholesterolemia. In this review, we discuss the role of PCSK9 in regulating cholesterol and summarize the promising findings of blocking PCSK9 with antibodies in human clinical trials. While the efficacy of blocking PCSK9 using antibodies to treat high cholesterol is still forthcoming in at least 12 ongoing Phase III clinical studies, anti-PCSK9 antibodies appear effective with minimal safety or tolerance issues identified to date. Blocking PCSK9 may offer a viable complement to statin therapies, which are not universally tolerated or effective with all patient populations.

Key words: Atherosclerosis; PCSK9; Statin; Cholesterol; Lipid metabolism

© 2015 The Authors. Published by ACT Group Ltd.

Han Y, Willis MS. The Role of PCSK9 in Lipid Metabolism and its Relationship to New Therapies for Lowering Cholesterol and Reducing Cardiac Disease. Journal of Cardiology and Therapy 2015; 2(5): 393-399 Available from: URL: http://www.ghrnet.org/index.php/jct/article/view/1223

Nonstandard Abbreviation and Acronyms : vLDL: very-low-density lipoprotein; IDL: intermediate density lipoprotein; LDL: low-density lipoprotein; HDL: high density lipoprotein; LDLR: low-density lipoprotein receptor; PCSK9: proprotein convertase subtilisin-like/kexin type 9; IHD: ischemic heart disease; TC: total cholesterol; PCR: polymerase chain reaction; CAD: coronary artery disease; IS: ischemic stroke.

Treating Atherosclerosis: Today

The treatment of atherosclerosis, the underlying cause of cardiovascular disease, involves lifestyle changes (e.g. changing diet and exercise regimens, loosing weight, stopping smoking, managing stress) and medical interventions involving primarily statins in those that can tolerate them. The goal in treating atherosclerosis is to slow the buildup of plaques, lower the risk of blood clots, relieve symptoms (if present), and prevent heart attacks (stroke, intestinal ischemia, peripheral artery disease). Lowering LDL, triglycerides, or overall cholesterol are the three primary ways in which medicines target atherosclerosis. Despite the excellent safety profile of statins, muscle toxicity is the major limitation for its use. Non-statin therapies are available, but their effectiveness in reducing LDL cholesterol concentrations is modest (15%-20%) compared with those obtained with a full-dose statin regimen. Thus, patients at high cardiovascular risk who are unable to achieve LDL cholesterol goals or to tolerate effective doses of statins because of muscle-related side effects represent a difficult population to treat. This population is steadily increasing and needs alternative strategies to reduce cardiovascular risk.

The class of medicine most widely used to lower cholesterol today are statins, which are HMG-CoA reductase inhibitors, which lower LDL cholesterol by reducing cholesterol synthesis in the liver (Figure 1, see **). This class of drugs lowers LDL cholesterol, while often raising HDL and lowering triglycerides, all beneficial to treating atherosclerosis[1]. While statins are the most effective cholesterol lowering drugs in use today, most individuals do not reach the desired outcome[2]. In the United Kingdom, only 28% of individuals achieve targeted level of cholesterol with statin therapy[3]. But they have many side effects and risks of liver damage and muscle fatigue[4]. Fibrates, such as Gemfibrozil and Fenofibrate, are used to more specifically reduce triglycerides, which contribute to atherosclerosis[5]. Similarly, niacin can improve cholesterol by reducing triglycerides and LDL by reducing mobilization of free fatty acids, decreasing substrate for hepatic synthesis of triglycerides and VLDL[6,7], along with other recently described mechanisms that increase HDL[8]. Zetia (ezetimibe) reduces cholesterol uptake in the intestines by blocking its absorption­. While it can lower LDL, it does not do as good of a job as statins[10]. Bile acid sequestrants (cholestryramine) bind bile acids in the intestines to lower cholesterol, and lovaza and vascepa (containing omega-3 fatty acids) are prescription drugs used to lower triglycerides[11]. Reducing blood pressure, in addition to lowering cholesterol, is a last important treatment in reducing atherosclerotic disease. Since none of these therapies are direct regulators of LDL cholesterol metabolism and have many side effects preventing their use, additional therapies are necessary to treat atherosclerosis.

Treating Atherosclerosis and High Cholesterol: Targeting PCSK9 in the Future?

A number of studies were published recently investigating proprotein convertase subtilisin/kexin type-9 (PCSK9) as a new target to treat atherosclerosis. How it became a new target of interest has involved some new discoveries teaching us that our understanding of how lipid metabolism is regulated is more complex than previously thought. The liver-derived PCSK9, a gene found on the short arm of chromosome 1 that encodes a 692 amino acid protein, was initially discovered as neural apoptosis-regulated convertase 1 (NARC-1) in cancer cell lines and implicated in the differentiation of cortical neurons in 2003[12]. Since then, therapies targeting PCSK9 to lower LDL-C have been successfully developed in pre-clinical trials and more recently multiple ongoing and completed Phase III human trials, which we discuss here.

The role of regulation of PCSK9 in regulating lipid metabolism

PCSK9 influence on cholesterol was evident in observational Dallas Heart Studies of multi-ethnic probability-based sample of Dallas County[13]. One striking finding in these studies was the variability of circulating plasma PCSK9 concentrations found in patients. PCSK9 levels varied 100 fold, ranging from 33 to 2988 ng/mL (median 487 ng/ml), and correlating closely with LDL-C[13]. PCSK9 levels were significantly higher in women (517 ng/mL) than men (450 ng/mL) and correlated with LDL-C, plasma triglycerides, insulin, and glucose[13]. Follow-up studies identified patients with loss of function PCSK9 mutations and reduced plasma levels of LDL-C had significantly lower levels of PCSK9, adjusting for age, gender, and LDL-C levels (p<0.0001)[14]. While these results suggested that inhibiting PCSK9 activity may be therapeutically beneficial, the underlying mechanism at the time was unclear[14]. One of the unique characteristics of PCSK9 is that plasma PCSK9 concentrations vary minimally in response to short term high fat challenges and is not accompanied by changes in hypercholesterolemia induced by high-fructose diets[14]. Short term high-fructose diet, however, increases plasma PCSK9 suggesting that PCSK9 is regulated by SREBP-2, associated with insulin resistance, hepatic steatosis, and plasma triglycerides[14]. Together, these studies suggest that PCSK9 regulates triglyceride-rich lipoproteins, a common contributor to cardiovascular disease in diabetes mellitus and a difficult to control factor in its pathogenesis and disease progression.

PCSK9 and LDL-apoB Catabolism

The association between PCSK9 and the degradation of the LDL receptor in patients was investigated next in patients with a wide range of body mass index values[15]. Like the relationship identified in PCSK9 mutations, plasma PCSK9 concentration correlated with cholesterol, LDL-C, and LDL apoB-100 concentrations and inversely correlated with the LDL Apo B-100 fractional catabolic rate[15]. This association remained statistically significant after adjusting for age, obesity, plasma insulin, insulin resistance measures (HOMA), and dietary energy intake[15]. These studies were vital in determining that PCSK9 levels were independent of obesity, insulin resistance, energy intake and age and directly linked the Apo B-100 catabolism with PCSK9, accounting for nearly 50% of the correlation (r=-0.456, p=0.038)[15]. Subsequently, mice expressing human PCSK9 were created to identify how PCSK9 and LDLR interact. These studies supported a role for LDLR in eliminating PCSK9, which reciprocally reduces LDLR[16]. In this way, serum PCSK9 levels and hepatic LDLR expression and serum LDL levels reciprocally balance each others activity and offer a mechanism in which reducing PCSK9 results in the reduction of serum lipoproteins and cholesterol[16].

Critical Role of cleaving PCSK9 to inactivate in vivo

The formation and inactivation of PCSK9 is a multi-step process, with the 62 kDa proprotein secreted in liver, intestine, and kidney tissues[17]. As an escort protein to the lysosomal degradatory pathway, it is cleaved after the R218 residue, resulting in the detachment and formation of a 55 kDa inactive form of PCSK9[18]. The proteolysis of PCSK9 is mediated by two proprotein convertases in hepatocytes, named furin and PC5/6A[19,20]. Evidence supporting this cleavage site includes missense mutations in the R218 residue that prevent inactivation in PCSK9 (e.g. R218S), result in an autosomal dominant hypercholesterolemia[21]. Mechanistically, these studies indicate that PCSK9 inactivation by furin and PC5/6A lead to an increased in proteins degraded by PCSK9 (e.g. LDLR, VLDL, ApoER2) responsible for clearing circulating lipoproteins. In turn, hypercholesterolemia results.

In summary, PCSK9 was found to be responsible for inhibiting the clearance involved in lipid metabolism, at the level of the liver. Specifically, PCSK9 was found to clear the LDL receptor (LDLR), VLDL receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2), all of which are critical to clearing and ultimately reducing circulating cholesterol levels (see Figure 1)[22]. Since PCSK9 activity reduces the clearance mechanisms needed to prevent lipid accumulation, inhibiting its activity would be predicted to reduce cholesterol levels, as patients with PCSK9 loss of function mutations demonstrated. This hypothesis was tested by the development of therapies to reduce PCSK9 activity and expression.

[12,23-39]Targeting PCSK9 Therapeutically to Reduce Cholesterol and Risk of CV Disease

Cholesterol lowering drugs, such as statins, and fibrates lower PCSK9, despite being developed against other targets[40]. Statin therapies up-regulate PCSK9 in hepatocytes to a greater extent than they increase the expression of the LDLR itself[41]. This finding reveals the paradox of statins, which increase LDLR activity, resulting in lowering LDL-C on the one hand, while simultaneously increasing the expression of PCSK9 on the other hand, an effect resulting in LDLR degradation (and opposing LDL-C lowering effects)[42]. Treating hypercholestermic patients with statins results in increased PSCK9 vs. controls, while increasing statin dose and addicting ezetimibe further enhanced PCSK9 levels, opposing the observed decreases in cholesterol seen[43]. Later studies reported different results. In these randomized clinical trials, it was confirmed that simvastatin, but not the fibrate ezetimib, resulted in upregulating circulating PCSK9 levels[44]. Since fibrates significantly modify PCSK9 concentrations, it may be by indirectly modifying cholesterol levels[45]. Bile acid binding resins have also been found to increase PCSK9 gene expression in human liver biopsies[46], while reduction of lipoproteins by apheresis results in a 50% reduction of PCSK9 levels by trapping LDL-bound and Apo B-free PCSK9 in the apheresis.

PCSK9 inhibitors in Clinical Trials

Phase I Clinical Trials. An anti-sense nucleotide targeting PCSK9 was begun in a Phase I clinical trial. However, it was terminated prematurely and no ongoing trials pursuing anti-sense oligonucleotides of PCSK9 are currently underway[53]. The use of small interfering RNA (siRNA) against PCSK9, which binds messenger RNA of PCSK9 and reduces protein expression, has been used to reduce LDL-C. In Phase I clinical trials, healthy volunteers given siRNA had a 70% reduction in circulating PCSK9 concentrations, resulting in a 40% reduction in LDL-C[54]. Monoclonal antibodies against PCSK9 have been developed to prevent PCSK9 binding to the LDL receptor. In a Phase I clinical trial of the AMG145 monoclonal antibody, a single injection reduced LDL level by up to 64% compared to placebo[55]. Patients with baseline LDL levels of 70-200 mg/dL were given AMG145 repeatedly, after 6-8 weeks[55], AMG145 injections resulted in 75% reduction of LDL level.

Phase II Clinical Trials. Determining the safety and frequency of dosing

PCSK9 effects with statin therapy

To assess the efficacy and tolerability of AMG145 in patients with statin intolerance due to muscle-related side effects, a 12 week randomized, double-blind, placebo and ezetimibe-controlled, dose-ranging study was performed at 33 international locations. With a primary end point of change in LDL-C measured by ultracentrifugation, 160 randomized patients were found to have a lowering of LDL-C of 67 mg/dL (-41%, 95% CI -49% to -33%) for the 280 mg AMG145 group[56]. Lowering of LDL-C by -70 mg/dL and -92 mg/dL were seen in the 350 and 420 mg groups; a lowering of -110 mg/dL for the 420 mg/ezetimibe group was seen compared to the -14 mg/dL seen in the placebo/ezetimibe group (p<0.001)[56]. The short-term tolerability was good in these studies, with only myalgia occurring in 15.6% of the 280 mg group, 3.2% in the 350 mg group, and 3.1% in the 420 mg group[56].

Frequency of administration

Phase II trials to assess the LDL-C lowering by PCSK9 inhibitors using different administration frequencies have been completed[57]. SAR236553, a fully human monoclonal antibody to PCSK9, has been tested in a double blind, parallel group, placebo controlled randomized trial of 183 patients with LDL-C≥ 100 mg/dL on stable-dose atorvastatin 10, 20, or 40 for ≥ 6 weeks in Phase 2 clinical trials[57]. SAR236553 had a clear dose-response with respect to LCL-C lowering when given every 2 weeks (Q2W) or every 4 weeks (Q4W). Reductions in 40%, 64%, and 72% with 50, 100, and 150 mg Q2W were seen, with reductions in 43% and 48% with 200 and 300 mg Q4W[57]. Notably, patients enrolled found the drug generally well tolerated, with only one patient experiencing an adverse event (leukocytoclastic vasculitis)[57].

Phase III Clinical Trials of monoclonal anti-PCSK9 antibodies

Six (6) Phase III clinical trials investigating monoclonal antibodies against PCSK9 have been completed, with nine (9) ongoing, including Evolocumab, Bococizumab, and Alirocumab. In the completed studies, Alirocumab is the only drug that has been tested to date. The focus of these studies has been on patients with statin intolerance and familial hypercholesterolemia. Statin intolerance occurs in ~5-10% of patients taking statins resulting in myalgia, rhabdomyolysis, and elevated liver enzymes, while familial hypercholesterolemia, occurring in 1/200 – 1/500 people, is characterized by extremely high LDL-C concentrations and the development of atherosclerosis and cardiovascular disease at a young age[58]. Patients unable to reach LDL cholesterol goals with high dose statins may also benefit from PCSK9 inhibitors, which have also been investigated in these Phase III studies. Since PCSK9 inhibitors effectively reduce LDL-C when added to statins, they may be effective against cardiovascular disease complications.

In the ODYSSEY ALTERNATIVE (NCT01709513), the efficacy and safety of Alirocumab was tested against two comparator therapies: ezetimibe (a drug inhibiting cholesterol absorption from the gut) and a statin (atorvastatin), along with two placebos for 24 weeks in this Phase III trial[59]. In this ongoing randomized study, patients with primary hypercholesterolemia intolerant to statins are followed for the primary outcomes of change in LDL-C at baseline compared to 24 weeks of therapy.

In the ODYSSEY OPTIONS I trial, patients with inadequately controlled hypercholesterolemia and high cardiovascular disease risk were studied for Alirocumab LDL-C reduction as an add-on therapy to atorvastatin compared to ezetimibe as an add-on to atorvastatin, compared to double atorvastatin dose, compared to switching from atorvastatin to rosuvastatin after 24 weeks of treatment[60]. The ODYSSEY OPTIONS II trials tested Alirocumab in hypercholesterolemia not adequately controlled in patients with high cardiovascular disease risk to evaluate Alirocumab LDL-C reduction as an add-on therapy to rosuvastatin, ezetimibe as an add-on to rosuvastatin, compared to double atorvastatin dose, compared to switching from atorvastatin to rosuvastatin after 24 weeks of treatment[60]. Together, these studies demonstrated the long-term efficacy and safety in high-risk patients when given with a maximally tolerated statin, allowing for therapy based on the degree of LDL-C lowering needed on an individual basis to achieve the goal treatment response.

In the ODYSSEY FH I (NCT01623115) trials, patients with heterozygous familial hypercholesterolemia and inadequately controlled LDL-C with current lipid-modifying therapy were evaluated for the effect of Alirocumab compared to placebo on LDL-C after 24 weeks of treatment[61]. In this trial completed in Dec. 2014, heterozygous familial hypercholesterolemia patients inadequately controlled with maximally tolerated statin with or without other lipid-modifying therapy were treated with Alirocumab to demonstrate reduction in LDL-C compared to placebo as an add-on therapy to stable, maximally tolerated daily statin therapy with or without other lipid-modifying therapy for 24 hours[61]. The ODYSSEY FH II (NCT01709500) phase 3 trial comparing atorvastatin, simvastatin, or rosuvastatin vs Alirocumab, vs. placebo, was completed in January 2015. The ODDYSSEY HIGH FH (NCT01617655) investigated Alirocumab in heterozygous familial hypercholesterolemia and LDL-C levels ≥160 mg/dL) for its efficacy, safety, and tolerability.

What Phase III Studies have learned: Safety and tolerance of anti-PCSK9 antibodies. While the final clinical results have yet to be published, we do have information on the safety and tolerance of anti-PCSK9 preliminarily. Phase III trials have found that 36-75% of patients given PSCK9 inhibitors had adverse events, with 2-6% of them serious. Discontinuation of the drug occurred in 2-10% of patients, a rate comparable to the group being compared[62]. Liver function tests >3 times upper normal limit (aminotransferases) were found in <2% of the patients; abnormal elevations in creatine kinase were similarly rare[63]. Anti-drug antibodies were not detected, which could potentially alter PCSK9 inhibitor pharmacokinetics in either LDL-C concentrations or safety in any Phase Ⅲ trials.

Injection site reactions were observed in 1/52 patients in the Odyssey MONO trials vs. 2/51 in the placebo group at 24 weeks[63]. At 52 weeks, the OSLER trials reported only 1/38 patients with an injection-site reaction[64]. The DESCARTES study found 34/599 patients (5.7%) in the Evolocumab group and 14/302 (5.0%) in the placebo group experiences injection-site reactions, with only 1 discontinuing Evolocumab[65]. In the ODYSSEY COMBO II Study, approximately 85% of patients were taking medication (or placebo) at 1 year, indicating a good tolerance of parenteral LDL-C lowering therapy[66]. While more detail of these profiles will be forthcoming, the studies have been suggestive of a generally safe and well tolerated therapy relative to currently available treatments.

Discussion

The major limitation of statin therapies has been their muscle and liver toxicity that occurs in some patients. Alternatives to statins only modestly reduce LDL-C, while patients with extreme hypercholesterolemia due to genetic mutations often need more than statins alone to to reduce LDL-C. Here we presented the emerging evidence for PCSK9 as a new target to treat high cholesterol, in addition to specific therapies inhibiting PCSK9 activity to reduce cholesterol in Phase I-III clinical trials. While the efficacy of blocking PCSK9 using antibodies to treat high cholesterol is still forthcoming in at least 12 ongoing Phase III clinical studies (Table 1), anti-PCSK9 antibodies appear effective with minimal safety or tolerance issues identified to date. Blocking PCSK9 may offer a viable complement to statin therapies, which are not universally tolerated or effective with all patient populations. While there is much to learn about PCSK9 before being accepted in practice, they offer a new hope and alternative to treating cardiovascular disease and atherosclerosis and will likely become a milestone on the road to curing hypercholesterolemia.

CONFLICT OF INTERESTS

There are no conflicts of interest with regard to the present study.

REFERENCES

1. Artom N, Montecucco F, Dallegri F, Pende A. Carotid atherosclerotic plaque stenosis: the stabilizing role of statins. Eur J Clin Invest 2014;44:1122-34.

2. Dadu RT, Ballantyne CM. Lipid lowering with PCSK9 inhibitors. Nat Rev Cardiol 2014;11:563-75.

3. Jameson K, Amber V, D'Oca K, Mills D, Giles A, Ambegaonkar B. Impact of lipid-lowering therapy on the prevalence of dyslipidaemia in patients at high-risk of cardiovascular events in UK primary care - a retrospective database study. Int J Clin Pract 2013;67:1228-37.

4. Desai CS, Martin SS, Blumenthal RS. Non-cardiovascular effects associated with statins. BMJ 2014;349:g3743.

5. Agouridis AP, Rizos CV, Elisaf MS, Filippatos TD. Does combination therapy with statins and fibrates prevent cardiovascular disease in diabetic patients with atherogenic mixed dyslipidemia? Rev Diabet Stud 2013;10:171-90.

6. Wu ZH, Zhao SP. Niacin promotes cholesterol efflux through stimulation of the PPARgamma-LXRalpha-ABCA1 pathway in 3T3-L1 adipocytes. Pharmacology 2009;84:282-7.

7. Grundy SM, Mok HY, Zech L, Berman M. Influence of nicotinic acid on metabolism of cholesterol and triglycerides in man. J Lipid Res 1981;22:24-36.

8. Hochholzer W, Berg DD, Giugliano RP. The Facts Behind Niacin. Ther Adv Cardiovasc Dis 2011;5:227-40.

9. Catapano AL, Farnier M, Foody JM, et al. Combination therapy in dyslipidemia: Where are we now? Atherosclerosis 2014;237:319-35.

10. Meyers CD, Moon YS, Ghanem H, Wong ND. Type of preexisting lipid therapy predicts LDL-C response to ezetimibe. Ann Pharmacother 2006;40:818-23.

11. Weintraub HS. Overview of prescription omega-3 fatty acid products for hypertriglyceridemia. Postgrad Med 2014;126:7-18.

12. Seidah NG, Benjannet S, Wickham L, et al. The secretory proprotein convertase neural apoptosis-regulated convertase 1 (NARC-1): liver regeneration and neuronal differentiation. Proc Natl Acad Sci U S A 2003;100:928-33.

13. Lakoski SG, Lagace TA, Cohen JC, Horton JD, Hobbs HH. Genetic and metabolic determinants of plasma PCSK9 levels. J Clin Endocrinol Metab 2009;94:2537-43.

14. Cariou B, Langhi C, Le Bras M, et al. Plasma PCSK9 concentrations during an oral fat load and after short term high-fat, high-fat high-protein and high-fructose diets. Nutr Metab (Lond) 2013;10:4.

15. Chan DC, Lambert G, Barrett PH, Rye KA, Ooi EM, Watts GF. Plasma proprotein convertase subtilisin/kexin type 9: a marker of LDL apolipoprotein B-100 catabolism? Clin Chem 2009;55:2049-52.

16. Tavori H, Fan D, Blakemore JL, et al. Serum proprotein convertase subtilisin/kexin type 9 and cell surface low-density lipoprotein receptor: evidence for a reciprocal regulation. Circulation 2013;127:2403-13.

17. Urban D, Poss J, Bohm M, Laufs U. Targeting the proprotein convertase subtilisin/kexin type 9 for the treatment of dyslipidemia and atherosclerosis. J Am Coll Cardiol 2013;62:1401-8.

18. Lambert G, Sjouke B, Choque B, Kastelein JJ, Hovingh GK. The PCSK9 decade. J Lipid Res 2012;53:2515-24.

19. Benjannet S, Rhainds D, Hamelin J, Nassoury N, Seidah NG. The proprotein convertase (PC) PCSK9 is inactivated by furin and/or PC5/6A: functional consequences of natural mutations and post-translational modifications. J Biol Chem 2006;281:30561-72.

20. Essalmani R, Susan-Resiga D, Chamberland A, et al. In vivo evidence that furin from hepatocytes inactivates PCSK9. J Biol Chem 2011;286:4257-63.

21. Allard D, Amsellem S, Abifadel M, et al. Novel mutations of the PCSK9 gene cause variable phenotype of autosomal dominant hypercholesterolemia. Hum Mutat 2005;26:497.

22. Poirier S, Mayer G, Benjannet S, et al. The proprotein convertase PCSK9 induces the degradation of low density lipoprotein receptor (LDLR) and its closest family members VLDLR and ApoER2. J Biol Chem 2008;283:2363-72.

23. Abifadel M, Varret M, Rabes JP, et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet 2003;34:154-6.

24. 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.

25. Lagace TA, Curtis DE, Garuti R, et al. Secreted PCSK9 decreases the number of LDL receptors in hepatocytes and in livers of parabiotic mice. J Clin Invest 2006;116:2995-3005.

26. Benjannet S, Rhainds D, Essalmani R, et al. NARC-1/PCSK9 and its natural mutants: zymogen cleavage and effects on the low density lipoprotein (LDL) receptor and LDL cholesterol. J Biol Chem 2004;279:48865-75.

27. Poirier S, Mayer G. The biology of PCSK9 from the endoplasmic reticulum to lysosomes: new and emerging therapeutics to control low-density lipoprotein cholesterol. Drug Des Devel Ther 2013;7:1135-48.

28. Stein EA, Swergold GD. Potential of proprotein convertase subtilisin/kexin type 9 based therapeutics. Curr Atheroscler Rep 2013;15:310.

29. Lagace TA. PCSK9 and LDLR degradation: regulatory mechanisms in circulation and in cells. Curr Opin Lipidol 2014;25:387-93.

30. Davignon J, Dubuc G, Seidah NG. The influence of PCSK9 polymorphisms on serum low-density lipoprotein cholesterol and risk of atherosclerosis. Curr Atheroscler Rep 2010;12:308-15.

31. Abifadel M, Rabes JP, Devillers M, et al. Mutations and polymorphisms in the proprotein convertase subtilisin kexin 9 (PCSK9) gene in cholesterol metabolism and disease. Hum Mutat 2009;30:520-9.

32. Timms KM, Wagner S, Samuels ME, et al. A mutation in PCSK9 causing autosomal-dominant hypercholesterolemia in a Utah pedigree. Hum Genet 2004;114:349-53.

33. Shioji K, Mannami T, Kokubo Y, et al. Genetic variants in PCSK9 affect the cholesterol level in Japanese. J Hum Genet 2004;49:109-14.

34. Awan Z, Delvin EE, Levy E, et al. Regional distribution and metabolic effect of PCSK9 insLEU and R46L gene mutations and apoE genotype. Can J Cardiol 2013;29:927-33.

35. Aung LH, Yin RX, Miao L, et al. The proprotein convertase subtilisin/kexin type 9 gene E670G polymorphism and serum lipid levels in the Guangxi Bai Ku Yao and Han populations. Lipids Health Dis 2011;10:5.

36. Chen SN, Ballantyne CM, Gotto AM, Jr., Tan Y, Willerson JT, Marian AJ. A common PCSK9 haplotype, encompassing the E670G coding single nucleotide polymorphism, is a novel genetic marker for plasma low-density lipoprotein cholesterol levels and severity of coronary atherosclerosis. J Am Coll Cardiol 2005;45:1611-9.

37. Slimani A, Harira Y, Trabelsi I, et al. Effect of E670G Polymorphism in PCSK9 Gene on the Risk and Severity of Coronary Heart Disease and Ischemic Stroke in a Tunisian Cohort. J Mol Neurosci 2014;53:150-7.

38. Hunninghake DB, Miller VT, LaRosa JC, et al. Long-term treatment of hypercholesterolemia with dietary fiber. Am J Med 1994;97:504-8.

39. Mayne J, Dewpura T, Raymond A, et al. Novel loss-of-function PCSK9 variant is associated with low plasma LDL cholesterol in a French-Canadian family and with impaired processing and secretion in cell culture. Clin Chem 2011;57:1415-23.

40. Guo YL, Zhang W, Li JJ. PCSK9 and lipid lowering drugs. Clin Chim Acta 2014;437:66-71.

41. Dubuc G, Chamberland A, Wassef H, et al. Statins upregulate PCSK9, the gene encoding the proprotein convertase neural apoptosis-regulated convertase-1 implicated in familial hypercholesterolemia. Arterioscler Thromb Vasc Biol 2004;24:1454-9.

42. Attie AD, Seidah NG. Dual regulation of the LDL receptor--some clarity and new questions. Cell Metab 2005;1:290-2.

43. Davignon J, Dubuc G. Statins and ezetimibe modulate plasma proprotein convertase subtilisin kexin-9 (PCSK9) levels. Trans Am Clin Climatol Assoc 2009;120:163-73.

44. Berthold HK, Seidah NG, Benjannet S, Gouni-Berthold I. Evidence from a randomized trial that simvastatin, but not ezetimibe, upregulates circulating PCSK9 levels. PLoS One 2013;8:e60095.

45. Mayne J, Dewpura T, Raymond A, et al. Plasma PCSK9 levels are significantly modified by statins and fibrates in humans. Lipids Health Dis 2008;7:22.

46. Nilsson LM, Abrahamsson A, Sahlin S, et al. Bile acids and lipoprotein metabolism: effects of cholestyramine and chenodeoxycholic acid on human hepatic mRNA expression. Biochem Biophys Res Commun 2007;357:707-11.

47. Chan JC, Piper DE, Cao Q, et al. A proprotein convertase subtilisin/kexin type 9 neutralizing antibody reduces serum cholesterol in mice and nonhuman primates. Proc Natl Acad Sci U S A 2009;106:9820-5.

48. Zhang L, McCabe T, Condra JH, et al. An anti-PCSK9 antibody reduces LDL-cholesterol on top of a statin and suppresses hepatocyte SREBP-regulated genes. Int J Biol Sci 2012;8:310-27.

49. Ni YG, Di Marco S, Condra JH, et al. A PCSK9-binding antibody that structurally mimics the EGF(A) domain of LDL-receptor reduces LDL cholesterol in vivo. J Lipid Res 2011;52:78-86.

50. Frank-Kamenetsky M, Grefhorst A, Anderson NN, et al. Therapeutic RNAi targeting PCSK9 acutely lowers plasma cholesterol in rodents and LDL cholesterol in nonhuman primates. Proc Natl Acad Sci U S A 2008;105:11915-20.

51. Lindholm MW, Elmen J, Fisker N, et al. PCSK9 LNA antisense oligonucleotides induce sustained reduction of LDL cholesterol in nonhuman primates. Mol Ther 2012;20:376-81.

52. Ding Q, Strong A, Patel KM, et al. Permanent alteration of PCSK9 with in vivo CRISPR-Cas9 genome editing. Circ Res 2014;115:488-92.

53. Do RQ, Vogel RA, Schwartz GG. PCSK9 Inhibitors: potential in cardiovascular therapeutics. Curr Cardiol Rep 2013;15:345.

54. Kuczkowski J, Lewandowski D. [2 cases of facial neurofibroma in children]. Otolaryngol Pol 1985;39:345-9.

55. Dias CS, Shaywitz AJ, Wasserman SM, et al. Effects of AMG 145 on low-density lipoprotein cholesterol levels: results from 2 randomized, double-blind, placebo-controlled, ascending-dose phase 1 studies in healthy volunteers and hypercholesterolemic subjects on statins. J Am Coll Cardiol 2012;60:1888-98.

56. Sullivan D, Olsson AG, Scott R, et al. Effect of a monoclonal antibody to PCSK9 on low-density lipoprotein cholesterol levels in statin-intolerant patients: the GAUSS randomized trial. JAMA 2012;308:2497-506.

57. McKenney JM, Koren MJ, Kereiakes DJ, Hanotin C, Ferrand AC, Stein EA. Safety and efficacy of a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 serine protease, SAR236553/REGN727, in patients with primary hypercholesterolemia receiving ongoing stable atorvastatin therapy. J Am Coll Cardiol 2012;59:2344-53.

58. Nordestgaard BG, Chapman MJ, Humphries SE, et al. Familial hypercholesterolaemia is underdiagnosed and undertreated in the general population: guidance for clinicians to prevent coronary heart disease: consensus statement of the European Atherosclerosis Society. Eur Heart J 2013;34:3478-90a.

59. Moriarty PM, Jacobson TA, Bruckert E, et al. Efficacy and safety of alirocumab, a monoclonal antibody to PCSK9, in statin-intolerant patients: design and rationale of ODYSSEY ALTERNATIVE, a randomized phase 3 trial. J Clin Lipidol 2014;8:554-61.

60. Colhoun HM, Robinson JG, Farnier M, et al. Efficacy and safety of alirocumab, a fully human PCSK9 monoclonal antibody, in high cardiovascular risk patients with poorly controlled hypercholesterolemia on maximally tolerated doses of statins: rationale and design of the ODYSSEY COMBO I and II trials. BMC Cardiovasc Disord 2014;14:121.

61. Kastelein JJ, Robinson JG, Farnier M, et al. Efficacy and safety of alirocumab in patients with heterozygous familial hypercholesterolemia not adequately controlled with current lipid-lowering therapy: design and rationale of the ODYSSEY FH studies. Cardiovasc Drugs Ther 2014;28:281-9.

62. Shimada YJ, Cannon CP. PCSK9 (Proprotein convertase subtilisin/kexin type 9) inhibitors: past, present, and the future. Eur Heart J 2015.

63. Roth EM, Taskinen MR, Ginsberg HN, et al. Monotherapy with the PCSK9 inhibitor alirocumab versus ezetimibe in patients with hypercholesterolemia: results of a 24 week, double-blind, randomized Phase 3 trial. Int J Cardiol 2014;176:55-61.

64. Koren MJ, Giugliano RP, Raal FJ, et al. Efficacy and safety of longer-term administration of evolocumab (AMG 145) in patients with hypercholesterolemia: 52-week results from the Open-Label Study of Long-Term Evaluation Against LDL-C (OSLER) randomized trial. Circulation 2014;129:234-43.

65. Blom DJ, Hala T, Bolognese M, et al. A 52-week placebo-controlled trial of evolocumab in hyperlipidemia. N Engl J Med 2014;370:1809-19.

66. Cannon CP, Cariou B, Blom D, et al. Efficacy and safety of alirocumab in high cardiovascular risk patients with inadequately controlled hypercholesterolaemia on maximally tolerated doses of statins: the ODYSSEY COMBO II randomized controlled trial. Eur Heart J 2015;36:1186-94.

Peer reviewers: Guixue Wang, Professor, College of Bioengineering, Chongqing University, Chongqing, 400044 China; Manfredi Rizzo, BioMedical Department of Internal Medicine and Medical Specialties, University of Palermo, Via del Vespro, Palermo, Italy; Juan Pedro-Botet, Department of Medicine, Hospital del Mar, Psseig Marítim, Barcelona, Spain.

Refbacks

  • There are currently no refbacks.


Creative Commons License
This work is licensed under a Creative Commons Attribution 3.0 License.