Editorial Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Jul 15, 2017; 8(7): 311-316
Published online Jul 15, 2017. doi: 10.4239/wjd.v8.i7.311
PCSK9 and carbohydrate metabolism: A double-edged sword
Theodosios D Filippatos, Sebastian Filippas-Ntekouan, Eleni Pappa, Thalia Panagiotopoulou, Vasilios Tsimihodimos, Moses S Elisaf, Department of Internal Medicine, School of Medicine, University of Ioannina, 45110 Ioannina, Greece
Author contributions: Filippatos TD wrote the manuscript; Filippas-Ntekouan S and Panagiotopoulou T searched the bibliography; Pappa E, Tsimihodimos V and Elisaf MS made appropriate comments and checked the final version of the manuscript.
Conflict-of-interest statement: This review was written independently. Professor Elisaf MS reports personal fees from ASTRA ZENECA, grants and personal fees from MSD, personal fees from PFIZER, ABBOTT, SANOFI, BOEHRINGER INGELHEIM, ELI LILLY, GSK. The authors have given talks and attended conferences sponsored by various pharmaceutical companies, including Bristol-Myers Squibb, Pfizer, Lilly, Abbott, Amgen, AstraZeneca, Novartis, Vianex, Teva and MSD.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Moses S Elisaf, Professor, Department of Internal Medicine, School of Medicine, University of Ioannina, Staurou Niarchou Avenue, 45110 Ioannina, Greece. melisaf54@gmail.com
Telephone: +30-26-51007509 Fax: +30-26-51007016
Received: December 24, 2016
Peer-review started: December 28, 2016
First decision: February 17, 2017
Revised: February 27, 2017
Accepted: March 23, 2017
Article in press: March 24, 2017
Published online: July 15, 2017

Abstract

Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a paramount role in the degradation of low-density lipoprotein (LDL) receptors (LDLR) on the hepatic cells surface and subsequently affects LDL particles catabolism and LDL cholesterol (LDL-c) levels. The anti-PCSK9 monoclonal antibodies lead to substantial decrease of LDL-c concentration. PCSK9 (which is also expressed in pancreatic delta-cells) can decrease LDLR and subsequently decrease cholesterol accumulation in pancreatic beta-cells, which impairs glucose metabolism and reduces insulin secretion. Thus, a possible adverse effect of PCSK9 inhibitors on carbohydrate metabolism may be expected by this mechanism, which has been supported by the mendelian studies results. On the other hand, clinical data have suggested a detrimental association of PCSK9 with glucose metabolism. So, the inhibition of PCSK9 may be seen as a double-edged sword regarding carbohydrate metabolism. Completed clinical trials have not shown a detrimental effect of PCSK9 inhibitors on diabetes risk, but their short-term duration does not allow definite conclusions.

Key Words: Proprotein convertase subtilisin/kexin type 9, Diabetes, Carbohydrate metabolism, Low-density lipoprotein, Proprotein convertase subtilisin/kexin type 9 inhibitors

Core tip: Proprotein convertase subtilisin/kexin type 9 (PCSK9) may play a beneficial role in carbohydrate metabolism because it can decrease low-density lipoprotein receptor and subsequently decrease cholesterol accumulation in pancreatic beta-cells, which impairs glucose metabolism and reduces insulin secretion. In contrast, clinical data have suggested a detrimental association of PCSK9 with glucose metabolism. These conflicting mechanisms may lead to a neutral effect on carbohydrate variables and explain the results of short-term clinical trials with PCSK9 inhibitors, which have not shown an increased diabetes risk.



INTRODUCTION

Statins can dose dependently increase the incidence of new-onset diabetes mainly in patients with underlying abnormalities of carbohydrate metabolism. This effect is at least partially an “on target’’ effect related to the statin-induced inhibition of 5-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase[1-4]. These observations have led ongoing research to focus on the possible association of newer hypolipidemic drugs with incident diabetes. Proprotein convertase subtilisin/kexin type 9 (PCSK9) has been identified as a key protein in lipid and lipoprotein metabolism, which plays a paramount role in the degradation of low-density lipoprotein (LDL) receptors (LDLR) on the hepatic cells surface and subsequently affects LDL particles catabolism and LDL cholesterol (LDL-c) levels (Figure 1)[5]. The anti-PCSK9 monoclonal antibodies bind circulating PCSK9, thus preventing PCSK9-induced degradation of LDLR. The administration of these drugs on top of conventional lipid lowering treatment substantially decreases LDL-c concentration by approximately 50% in various groups of high-risk patients, while the treatment is well tolerated[6]. Even though significant differences in the incidence of most adverse events were not observed between PCSK9 inhibitors-treated and placebo-treated patients, an increased incidence of neurocognitive events was observed, which needs further evaluation[7].

Figure 1
Figure 1 The effect of proprotein convertase subtilisin/kexin type 9 (A) and proprotein convertase subtilisin/kexin type 9 inhibition (B) on liver cells low-density lipoprotein receptors expression and serum low-density lipoprotein-cholesterol levels. PCSK9: Proprotein convertase subtilisin/kexin type 9; LDL: Low-density lipoprotein; LDLR: Low-density lipoprotein receptors.

It has been shown that PCSK9 can decrease LDLR and subsequently decrease cholesterol concentrations in pancreatic beta-cells; thus, it may beneficially affect beta cell function, since the accumulation of cholesterol in beta-cells impairs glucose metabolism, reduces insulin secretion and can be associated with a diabetic phenotype[8]. Based on this concept, a crucial question emerges whether PCSK9 inhibitors can increase diabetes risk by inhibiting this beneficial effect (Table 1). This question is particularly relevant, because the results of genetic studies have shown contradictory results. Thus, even though no increased risk of diabetes or other changes in glucose homeostasis were found in individuals with PCSK9 loss-of function variants[9,10], carriage of the loss-of-function PCSK9 p.R46L mutation was associated with insulin resistance [increased homeostasis model assessment-insulin resistance (HOMA-IR) index] in those with apolipoprotein E3/E2 genotype[11]. However, another study did not confirm these results and showed that the p.R46L mutation was not associated with markers of glucose homeostasis, while p.R46L carriers did not experience an increased risk of new-onset diabetes mellitus[12]. Additionally, experimental data from animal models have also provided conflicting results. One study showed that PCSK9 deficiency does not alter insulin secretion and glucose tolerance in mice[13], while another study showed that PCSK9 deficient mice (PCSK9-/-) exhibit hyperglycemia, impaired glucose tolerance associated with hypoinsulinemia and pancreatic islet abnormalities (malformation, apoptosis and inflammation)[14]. Interestingly, PCSK9, whereas it is not expressed in α- and β-cells, is co-localized specifically with somatostatin in human pancreatic delta-cells, a finding which may be implicated in the previously mentioned results[13]. These findings support the previously mentioned statement concerning the detrimental role of LDLR-associated cholesterol accumulation in pancreatic beta-cells on insulin secretion and carbohydrate homeostasis. Accordingly, three recently published genetic studies showed that PCSK9 variants-associated genetically predicted reduction of LDL-c was related with an increased risk for type 2 diabetes (Table 2)[15-17]. Overall, these observations point to a possible adverse effect of PCSK9 inhibitors on carbohydrate metabolism.

Table 1 Studies that examined the association of proprotein convertase subtilisin/kexin type 9 with carbohydrate metabolism.
Ref.TypeMain findings
Studies pointing to a positive effect of PCSK9 on carbohydrate metabolism
Mbikay et al[14]Experimental (mice)PCSK9-null male mice over 4 mo of age carried more LDLR and less insulin in their pancreas; islets exhibited signs of malformation, apoptosis and inflammation
Awan et al[11]Genetic studyCarriage of the loss-of-function PCSK9 p.R46L mutation was associated with insulin resistance in subjects with apolipoprotein E3/E2 genotype
Studies pointing to a negative effect of PCSK9 on carbohydrate metabolism
Langhi et al[13]Experimental (mice)PCSK9 deficiency does not alter insulin secretion and glucose tolerance
Baass et al[18]Clinical study (children)Significant correlation of PCSK9 levels with glucose, insulin and HOMA-IR levels; an increase in PCSK9 levels by 1%-2% was associated with 10% higher fasting insulin levels in both sexes
Arsenault et al[21]Clinical study (abdominally obese men)PCSK9 levels are associated with dyslipidemia and with increased HOMA-IR
Studies pointing to a neutral effect of PCSK9 on carbohydrate metabolism
Bonnefond et al[12]Genetic studyThe p.R46L mutation is not associated with markers of glucose homeostasis; p.R46L carriers did not experience an increased risk of new-onset diabetes mellitus
Colhoun et al[22]Analysis of 10 phase 3 clinical trials with alirocumab (3448 non-diabetic individuals)Hazard ratio for diabetes-related treatment adverse effects 0.64 (95%CI: 0.36-1.14) in alirocumab-treated patients vs placebo-treated and 0.55 (95%CI: 0.22-1.41) vs ezetimibe-treated patients
Blom et al[23]Post hoc analysis of the DESCARTES trial (evolocumab)No changes in parameters of carbohydrate metabolism in patients with pre-existing dysglycemia or metabolic syndrome
Ongoing trials that may better delineate the role of PCSK9 inhibition on carbohydrate metabolism
Fourier trial (ClinicalTrials.gov Identifier: NCT01764633)Ongoing trialPrimary hypothesis is that additional LDL-c lowering with evolocumab decreases the risk of cardiovascular events in subjects with clinically evident cardiovascular disease
Odyssey trial (ClinicalTrials.gov Identifier: NCT01663402)Ongoing trialPrimary hypothesis is that additional LDL-c lowering with alirocumab decreases the risk of cardiovascular events in patients who have experienced an acute coronary syndrome event 4 to 52 wk prior to randomization
Table 2 Proprotein convertase subtilisin/kexin type 9 inhibitors and diabetes mellitus: Results of the mendelian randomization studies.
PCSK9 variantsDecrease in serum LDL cholesterolOdds ratio for type 2 diabetes mellitus
rs 11591147[15]1 mmol/L (38.4 mg/dL)1.19 (95%CI: 1.02-1.38)
14 independent variants[16] (rs 11583680, rs 11591147, rs 2479109, rs 11206510)1 mmol/L (38.4 mg/dL)1.29 (95%CI: 1.11-1.50)
2Genetic score[17]10 mg/dL1.11 (95%CI: 1.04-1.19)

On the other hand, available clinical data have suggested a detrimental association of PCSK9 with glucose metabolism (Table 1). Thus, in children a significant correlation of PCSK9 levels with glucose, insulin, and HOMA-IR levels was observed, while an increase in PCSK9 levels by 1%-2% was associated with 10% higher fasting insulin levels in both sexes[18]. It has been reported that hepatic PCSK9 expression is regulated by insulin via the sterol regulatory element-binding protein I-C (SREBP-1C); thus PCSK9 is secreted in an insulin-dependent fashion[19], underlying an association between PCSK9 and carbohydrate metabolism[20]. Additionally, in abdominally obese men PCSK9 levels were associated with dyslipidemia (with small dense LDL particles and increased apolipoprotein CIII levels) but also with insulin resistance (increased HOMA-IR)[21].

The results of the clinical trials, however, do not support any significant effect of these drugs on carbohydrate metabolism (Table 1). In fact, a recently published analysis of 10 phase 3 clinical trials with alirocumab showed that the hazard ratio for diabetes-related treatment adverse effects among 3448 non-diabetic individuals was 0.64 (95%CI: 0.36-1.14) in alirocumab-treated patients vs placebo-treated and 0.55 (95%CI: 0.22-1.41) vs ezetimibe-treated patients[22]. In prediabetic individuals, the hazard ratio associated with transition of prediabetes to new-onset diabetes for alirocumab was 0.90 (95%CI: 0.63-1.29) vs placebo and 1.10 (95%CI: 0.57-2.12) vs ezetimibe. Furthermore, no change in plasma glucose and glycated hemoglobin (HbA1c) levels was observed between treated groups in non-diabetic individuals of these results[22]. Additionally, a post hoc analysis of the DESCARTES showed that the administration of evolocumab (420 mg monthly) was not associated with any changes in parameters of carbohydrate metabolism in patients with pre-existing dysglycemia or metabolic syndrome[23]. Finally, the available data suggest similar effects of these drugs on the levels of serum lipid parameters in diabetic vs non-diabetic individuals[24]. However, the relatively small number of patients, the short-follow up, the design of the studies (administration on top of statin therapy) may reduce the significance of these observations.

Thus, the effects of PCSK9 and accordingly of PCSK9 inhibitors on carbohydrate metabolism may be seen under different points of view (Figure 2). The potential detrimental consequences of PCSK9 inhibitors on pancreatic cells leading to reduced insulin secretion due to a direct effect on pancreatic cells or to increased intracellular cholesterol levels may be counterbalanced by their direct beneficial effects on carbohydrate homeostasis. Alternatively, the relatively short duration of the above mentioned clinical trials is not adequate for any detrimental effect of PCSK9 inhibition to be evident. It should be also mentioned that in the clinical trials the addition of PCSK9 inhibitors to statins may have partially masked their effects on glucose metabolism if there are shared mechanisms of action between these two drug classes. Finally, a generally non-significant effect of PCSK9 inhibition on glucose metabolism cannot be excluded. Thus, the results of both Fourier (Clinical Trials.gov Identifier: NCT01764633) and Odyssey (Clinical Trials.gov Identifier: NCT01663402) outcome trials may better delineate the role of PCSK9 inhibitors on the parameters of glucose homeostasis and their long-term effect on the incidence of new-onset diabetes mellitus.

Figure 2
Figure 2 The role of proprotein convertase subtilisin/kexin type 9 on carbohydrate homeostasis. Accordingly, PCSK9 inhibitors may be associated with a neutral effect on carbohydrate homeostasis at least in the short term. PCSK9: Proprotein convertase subtilisin/kexin type 9; LDL: Low-density lipoprotein; LDLR: LDL receptors; HbA1c: Glycated hemoglobin; SREBP-1C: Sterol regulatory element-binding protein I-C; HOMA-IR: Homeostasis model assessment-insulin resistance.
Footnotes

Manuscript source: Invited manuscript

Specialty type: Endocrinology and metabolism

Country of origin: Greece

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B, B, B, B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Dinc M, Goemann IM, Hussain SAR, Kushiyama A, Zhao JB S- Editor: Ji FF L- Editor: A E- Editor: Lu YJ

References
1.  Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008;359:2195-2207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4679]  [Cited by in F6Publishing: 4528]  [Article Influence: 283.0]  [Reference Citation Analysis (0)]
2.  Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM, de Craen AJ, Seshasai SR, McMurray JJ, Freeman DJ, Jukema JW. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet. 2010;375:735-742.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1657]  [Cited by in F6Publishing: 1626]  [Article Influence: 116.1]  [Reference Citation Analysis (0)]
3.  Preiss D, Seshasai SR, Welsh P, Murphy SA, Ho JE, Waters DD, DeMicco DA, Barter P, Cannon CP, Sabatine MS. Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis. JAMA. 2011;305:2556-2564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1001]  [Cited by in F6Publishing: 972]  [Article Influence: 74.8]  [Reference Citation Analysis (0)]
4.  Swerdlow DI, Preiss D, Kuchenbaecker KB, Holmes MV, Engmann JE, Shah T, Sofat R, Stender S, Johnson PC, Scott RA. HMG-coenzyme A reductase inhibition, type 2 diabetes, and bodyweight: evidence from genetic analysis and randomised trials. Lancet. 2015;385:351-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 508]  [Cited by in F6Publishing: 462]  [Article Influence: 51.3]  [Reference Citation Analysis (0)]
5.  Horton JD, Cohen JC, Hobbs HH. Molecular biology of PCSK9: its role in LDL metabolism. Trends Biochem Sci. 2007;32:71-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 412]  [Cited by in F6Publishing: 429]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
6.  Do RQ, Vogel RA, Schwartz GG. PCSK9 Inhibitors: potential in cardiovascular therapeutics. Curr Cardiol Rep. 2013;15:345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
7.  Lipinski MJ, Benedetto U, Escarcega RO, Biondi-Zoccai G, Lhermusier T, Baker NC, Torguson R, Brewer HB, Waksman R. The impact of proprotein convertase subtilisin-kexin type 9 serine protease inhibitors on lipid levels and outcomes in patients with primary hypercholesterolaemia: a network meta-analysis. Eur Heart J. 2016;37:536-545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 171]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
8.  Fryirs M, Barter PJ, Rye KA. Cholesterol metabolism and pancreatic beta-cell function. Curr Opin Lipidol. 2009;20:159-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 68]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
9.  Cohen JC, Boerwinkle E, Mosley TH, Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med. 2006;354:1264-1272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2340]  [Cited by in F6Publishing: 2222]  [Article Influence: 123.4]  [Reference Citation Analysis (0)]
10.  Guella I, Asselta R, Ardissino D, Merlini PA, Peyvandi F, Kathiresan S, Mannucci PM, Tubaro M, Duga S. Effects of PCSK9 genetic variants on plasma LDL cholesterol levels and risk of premature myocardial infarction in the Italian population. J Lipid Res. 2010;51:3342-3349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
11.  Awan Z, Delvin EE, Levy E, Genest J, Davignon J, Seidah NG, Baass A. Regional distribution and metabolic effect of PCSK9 insLEU and R46L gene mutations and apoE genotype. Can J Cardiol. 2013;29:927-933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
12.  Bonnefond A, Yengo L, Le May C, Fumeron F, Marre M, Balkau B, Charpentier G, Franc S, Froguel P, Cariou B. The loss-of-function PCSK9 p.R46L genetic variant does not alter glucose homeostasis. Diabetologia. 2015;58:2051-2055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 38]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
13.  Langhi C, Le May C, Gmyr V, Vandewalle B, Kerr-Conte J, Krempf M, Pattou F, Costet P, Cariou B. PCSK9 is expressed in pancreatic delta-cells and does not alter insulin secretion. Biochem Biophys Res Commun. 2009;390:1288-1293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 83]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
14.  Mbikay M, Sirois F, Mayne J, Wang GS, Chen A, Dewpura T, Prat A, Seidah NG, Chretien M, Scott FW. PCSK9-deficient mice exhibit impaired glucose tolerance and pancreatic islet abnormalities. FEBS Lett. 2010;584:701-706.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 148]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
15.  Lotta LA, Sharp SJ, Burgess S, Perry JR, Stewart ID, Willems SM, Luan J, Ardanaz E, Arriola L, Balkau B. Association Between Low-Density Lipoprotein Cholesterol-Lowering Genetic Variants and Risk of Type 2 Diabetes: A Meta-analysis. JAMA. 2016;316:1383-1391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 266]  [Article Influence: 33.3]  [Reference Citation Analysis (0)]
16.  Schmidt AF, Swerdlow DI, Holmes MV, Patel RS, Fairhurst-Hunter Z, Lyall DM, Hartwig FP, Horta BL, Hyppönen E, Power C. PCSK9 genetic variants and risk of type 2 diabetes: a mendelian randomisation study. Lancet Diabetes Endocrinol. 2017;5:97-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 263]  [Article Influence: 37.6]  [Reference Citation Analysis (0)]
17.  Ference BA, Robinson JG, Brook RD, Catapano AL, Chapman MJ, Neff DR, Voros S, Giugliano RP, Davey Smith G, Fazio S. Variation in PCSK9 and HMGCR and Risk of Cardiovascular Disease and Diabetes. N Engl J Med. 2016;375:2144-2153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 507]  [Cited by in F6Publishing: 501]  [Article Influence: 62.6]  [Reference Citation Analysis (0)]
18.  Baass A, Dubuc G, Tremblay M, Delvin EE, O’Loughlin J, Levy E, Davignon J, Lambert M. Plasma PCSK9 is associated with age, sex, and multiple metabolic markers in a population-based sample of children and adolescents. Clin Chem. 2009;55:1637-1645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 170]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
19.  Costet P, Cariou B, Lambert G, Lalanne F, Lardeux B, Jarnoux AL, Grefhorst A, Staels B, Krempf M. Hepatic PCSK9 expression is regulated by nutritional status via insulin and sterol regulatory element-binding protein 1c. J Biol Chem. 2006;281:6211-6218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 241]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
20.  Cariou B, Le Bras M, Langhi C, Le May C, Guyomarc’h-Delasalle B, Krempf M, Costet P. Association between plasma PCSK9 and gamma-glutamyl transferase levels in diabetic patients. Atherosclerosis. 2010;211:700-702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
21.  Arsenault BJ, Pelletier-Beaumont E, Alméras N, Tremblay A, Poirier P, Bergeron J, Després JP. PCSK9 levels in abdominally obese men: association with cardiometabolic risk profile and effects of a one-year lifestyle modification program. Atherosclerosis. 2014;236:321-326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
22.  Colhoun HM, Ginsberg HN, Robinson JG, Leiter LA, Müller-Wieland D, Henry RR, Cariou B, Baccara-Dinet MT, Pordy R, Merlet L. No effect of PCSK9 inhibitor alirocumab on the incidence of diabetes in a pooled analysis from 10 ODYSSEY Phase 3 studies. Eur Heart J. 2016;37:2981-2989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 123]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
23.  Blom DJ, Koren MJ, Roth E, Monsalvo ML, Djedjos CS, Nelson P, Elliott M, Wasserman SM, Ballantyne CM, Holman RR. Evaluation of the efficacy, safety and glycaemic effects of evolocumab (AMG 145) in hypercholesterolaemic patients stratified by glycaemic status and metabolic syndrome. Diabetes Obes Metab. 2017;19:98-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
24.  Sattar N, Preiss D, Robinson JG, Djedjos CS, Elliott M, Somaratne R, Wasserman SM, Raal FJ. Lipid-lowering efficacy of the PCSK9 inhibitor evolocumab (AMG 145) in patients with type 2 diabetes: a meta-analysis of individual patient data. Lancet Diabetes Endocrinol. 2016;4:403-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 112]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]