1
|
Gianazza E, Macchi C, Banfi C, Ruscica M. Proteomics and Lipidomics to unveil the contribution of PCSK9 beyond cholesterol lowering: a narrative review. Front Cardiovasc Med 2023; 10:1191303. [PMID: 37378405 PMCID: PMC10291627 DOI: 10.3389/fcvm.2023.1191303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9), one of the key regulators of the low-density lipoprotein receptor (LDLR), can play a direct role in atheroma development. Although advances in the understandings of genetic PCSK9 polymorphisms have enabled to reveal the role of PCSK9 in the complex pathophysiology of cardiovascular diseases (CVDs), increasing lines of evidence support non-cholesterol-related processes mediated by PCSK9. Owing to major improvements in mass spectrometry-based technologies, multimarker proteomic and lipidomic panels hold the promise to identify novel lipids and proteins potentially related to PCSK9. Within this context, this narrative review aims to provide an overview of the most significant proteomics and lipidomics studies related to PCSK9 effects beyond cholesterol lowering. These approaches have enabled to unveil non-common targets of PCSK9, potentially leading to the development of novel statistical models for CVD risk prediction. Finally, in the era of precision medicine, we have reported the impact of PCSK9 on extracellular vesicles (EVs) composition, an effect that could contribute to an increased prothrombotic status in CVD patients. The possibility to modulate EVs release and cargo could help counteract the development and progression of the atherosclerotic process.
Collapse
Affiliation(s)
- Erica Gianazza
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
2
|
Zhou X, Li Z, Liu H, Li Y, Zhao D, Yang Q. Antithrombotic therapy and bleeding risk in the era of aggressive lipid-lowering: current evidence, clinical implications, and future perspectives. Chin Med J (Engl) 2023; 136:645-652. [PMID: 36806078 PMCID: PMC10129148 DOI: 10.1097/cm9.0000000000002057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Indexed: 02/23/2023] Open
Abstract
ABSTRACT The clinical efficacy of proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) in reducing major cardiovascular adverse events related to atherosclerotic cardiovascular disease (ASCVD) has been well established in recent large randomized outcome trials. Although the cardiovascular and all-cause mortality benefit of PCSK9i remains inconclusive, current cholesterol management guidelines have been modified toward more aggressive goals for lowering low-density lipoprotein cholesterol (LDL-C). Consequently, the emerging concept of "the lower the better" has become the paradigm of ASCVD prevention. However, there is evidence from observational studies of a U-shaped association between baseline LDL-C levels and all-cause mortality in population-based cohorts. Among East Asian populations, low LDL-C was associated with an increased risk for hemorrhagic stroke in patients not on antithrombotic therapy. Accumulating evidence showed that low LDL-C was associated with an enhanced bleeding risk in patients on dual antiplatelet therapy following percutaneous coronary intervention. Additionally, low LDL-C was associated with a higher risk for incident atrial fibrillation and thereby, a possible increase in the risk for intracranial hemorrhage after initiation of anticoagulation therapy. The mechanism of low-LDL-C-related bleeding risk has not been fully elucidated. This review summarizes recent evidence of low-LDL-C-related bleeding risk in patients on antithrombotic therapy and discusses potential measures for reducing this risk, underscoring the importance of carefully weighing the pros and cons of aggressive LDL-C lowering in patients on antithrombotic therapy.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ziping Li
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hangkuan Liu
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yongle Li
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Dong Zhao
- Department of Epidemiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Qing Yang
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
3
|
Sarkar SK, Matyas A, Asikhia I, Hu Z, Golder M, Beehler K, Kosenko T, Lagace TA. Pathogenic gain-of-function mutations in the prodomain and C-terminal domain of PCSK9 inhibit LDL binding. Front Physiol 2022; 13:960272. [PMID: 36187800 PMCID: PMC9515655 DOI: 10.3389/fphys.2022.960272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/23/2022] [Indexed: 11/30/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type-9 (PCSK9) is a secreted protein that binds and mediates endo-lysosomal degradation of low-density lipoprotein receptor (LDLR), limiting plasma clearance of cholesterol-rich LDL particles in liver. Gain-of-function (GOF) point mutations in PCSK9 are associated with familial hypercholesterolemia (FH). Approximately 30%–40% of PCSK9 in normolipidemic human plasma is bound to LDL particles. We previously reported that an R496W GOF mutation in a region of PCSK9 known as cysteine-histidine–rich domain module 1 (CM1) prevents LDL binding in vitro [Sarkar et al., J. Biol. Chem. 295 (8), 2285–2298 (2020)]. Herein, we identify additional GOF mutations that inhibit LDL association, localized either within CM1 or a surface-exposed region in the PCSK9 prodomain. Notably, LDL binding was nearly abolished by a prodomain S127R GOF mutation, one of the first PCSK9 mutations identified in FH patients. PCSK9 containing alanine or proline substitutions at amino acid position 127 were also defective for LDL binding. LDL inhibited cell surface LDLR binding and degradation induced by exogenous PCSK9-D374Y but had no effect on an S127R-D374Y double mutant form of PCSK9. These studies reveal that multiple FH-associated GOF mutations in two distinct regions of PCSK9 inhibit LDL binding, and that the Ser-127 residue in PCSK9 plays a critical role.
Collapse
Affiliation(s)
- Samantha K. Sarkar
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Angela Matyas
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Ikhuosho Asikhia
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Zhenkun Hu
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Mia Golder
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | | | - Tanja Kosenko
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Thomas A. Lagace
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- *Correspondence: Thomas A. Lagace,
| |
Collapse
|
4
|
Ferri N, Ruscica M, Lupo MG, Vicenzi M, Sirtori CR, Corsini A. Pharmacological rationale for the very early treatment of acute coronary syndrome with monoclonal antibodies anti-PCSK9. Pharmacol Res 2022; 184:106439. [PMID: 36100012 DOI: 10.1016/j.phrs.2022.106439] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/07/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022]
Abstract
Immediate and aggressive lipid lowering therapies after acute coronary syndromes (ACS) and percutaneous coronary interventions (PCI) are supported by the ESC/EAS dyslipidemia guidelines, recommending the initiation of high-intensity statin therapy within the first 1-4 days of hospitalization. However, whether non statin lipid-lowering agents, added to statin treatment, could produce a further reduction in the risk of major adverse cardiovascular events (MACE) is still unknown. Thus, the efficacy of early treatment post-ACS with monoclonal antibodies (mAbs) anti PCSK9, evolocumab and alirocumab, is under investigation. The rationale to explore the rapid and aggressive pharmacological intervention with PCSK9 mAbs is supported by at least five confirmatory data in ACS: 1) circulating PCSK9 levels are raised during ACS 2) PCSK9 may stimulate platelet reactivity, this last being pivotal in the recurrence of ischemic events; 3) PCSK9 is associated with intraplaque inflammation, macrophage activation and endothelial dysfunction; 4) PCSK9 concentrations are associated with inflammation in the acute phase of ACS; and 5) statins raise PCSK9 levels promptly and, at times, dramatically. In this scenario, appropriate pharmacodynamic characteristics of anti PCSK9 therapies are a prerequisite for an effective response. Monoclonal antibodies act on circulating PCSK9 with a direct and rapid binding by blocking the interaction with the low-density lipoprotein receptor (LDLR). Evolocumab and alirocumab show a very rapid (within 4 h) and effective suppression of circulating unbound PCSK9 (- 95 % ÷ - 97 %). This inhibition results in a significant reduction of LDL-cholesterol (LDL-C) after 48 h (- 35 %) post injection with a full effect after 7-10 days (55-75 %). The complete and swift inhibitory action by evolocumab and alirocumab could have a potential clinical impact in ACS patients, also considering their potential inhibition of PCSK9 within the atherosclerotic plaque. Thus, administration of evolocumab or alirocumab is effective in lowering LDL-C levels in ACS, although the efficacy to prevent further cardiovascular (CV) events is still undetermined. The answer to this question will be provided by the ongoing clinical trials with evolocumab and alirocumab in ACS. In the present review we will discuss the pharmacological and biological rationale supporting the potential use of PCSK9 mAbs in ACS patients and the emerging evidence of evolocumab and alirocumab treatment in this clinical setting.
Collapse
Affiliation(s)
- Nicola Ferri
- Dipartimento di Medicina, Università degli Studi di Padova, Padua, Italy.
| | - Massimiliano Ruscica
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | | | - Marco Vicenzi
- Cardiovascular Disease Unit, Internal Medicine Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cesare R Sirtori
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Alberto Corsini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
5
|
Dafnis I, Tsouka AN, Gkolfinopoulou C, Tellis CC, Chroni A, Tselepis AD. PCSK9 is minimally associated with HDL but impairs the anti-atherosclerotic HDL effects on endothelial cell activation. J Lipid Res 2022; 63:100272. [PMID: 36067830 PMCID: PMC9526147 DOI: 10.1016/j.jlr.2022.100272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 07/30/2022] [Accepted: 07/30/2022] [Indexed: 11/30/2022] Open
Abstract
Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9) regulates the cell-surface localization of LDL receptors in hepatocytes and is associated with LDL and lipoprotein(a) [Lp(a)] uptake, reducing blood concentrations. However, the connection between PCSK9 and HDL is unclear. Here, we investigated the association of plasma PCSK9 with HDL subpopulations and examined the effects of PCSK9 on the atheroprotective function of HDL. We examined the association of PCSK9 with HDL in apoB-depleted plasma by ELISA, native PAGE, and immunoblotting. Our analyses showed that upon apoB-depletion, total circulating PCSK9 levels were 32% of those observed in normolipidemic plasma, and only 6% of PCSK9 in the apoB-depleted plasma, including both the mature and furin-cleaved forms, was associated with HDL. We also show human recombinant PCSK9 abolished the capacity of reconstituted HDL to reduce the formation of ROS in endothelial cells, while a PCSK9-blocking antibody enhanced the capacity of human HDL (in apoB-depleted plasma) to reduce ROS formation in endothelial cells and promote endothelial cell migration. Overall, our findings suggest that PCSK9 is only minimally associated with HDL particles, but PCSK9 in apoB-depleted plasma can affect the atheroprotective properties of HDL related to preservation of endothelial function. This study contributes to the elucidation of the pathophysiological role of plasma PCSK9 and highlights further the anti-atherosclerotic effect of PCSK9 inhibition.
Collapse
Affiliation(s)
- Ioannis Dafnis
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Aikaterini N Tsouka
- Atherothrombosis Research Centre, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Christina Gkolfinopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Constantinos C Tellis
- Atherothrombosis Research Centre, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Alexandros D Tselepis
- Atherothrombosis Research Centre, Department of Chemistry, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
6
|
Pirillo A, Catapano AL. Evinacumab: a new option in the treatment of homozygous familial hypercholesterolemia. Expert Opin Biol Ther 2022; 22:813-820. [PMID: 35698895 DOI: 10.1080/14712598.2022.2090242] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Familial hypercholesterolemia is a genetic disorder characterized by elevated levels of low-density lipoprotein cholesterol (LDL-C) since birth and an exceedingly high risk of premature cardiovascular disease, especially in the homozygous form (HoFH). Despite the availability of effective cholesterol-lowering drugs, substantial LDL-C and cardiovascular risk reductions in these patients are still problematic, especially in those carrying mutations in the low-density lipoprotein receptor (LDLR) gene. AREAS COVERED Loss-of-function mutations in angiopoietin-like 3 (ANGPTL3) encoding gene are associated with lower levels of LDL-C and reduced cardiovascular risk; the pharmacological inhibition of ANGPTL3 reduces LDL-C levels independently of LDLR. This approach can thus improve the treatment of HoFH using a monoclonal antibody targeting ANGPTL3 (evinacumab). EXPERT OPINION Most lipid-lowering agents available so far are insufficient to achieve an appropriate response in HoFH patients. The inhibition of ANGPTL3 with evinacumab halves LDL-C levels in HoFH patients by an LDLR-independent mechanism. The results obtained so far have clearly indicated a promising improvement in the management of these patients. As the reduction of CV risk is proportional to the absolute reduction in LDL-C levels, we can expect that treatment with evinacumab, added to the maximally tolerated lipid-lowering therapy, will turn into a significant clinical benefit.
Collapse
Affiliation(s)
- Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, Milan, Italy.,Center for the Study of Dyslipidaemias, IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Alberico L Catapano
- Center for the Study of Dyslipidaemias, IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
7
|
Altabas V, Biloš LSK. The Role of Endothelial Progenitor Cells in Atherosclerosis and Impact of Anti-Lipemic Treatments on Endothelial Repair. Int J Mol Sci 2022; 23:ijms23052663. [PMID: 35269807 PMCID: PMC8910333 DOI: 10.3390/ijms23052663] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/16/2022] [Accepted: 02/26/2022] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular complications are associated with advanced atherosclerosis. Although atherosclerosis is still regarded as an incurable disease, at least in its more advanced stages, the discovery of endothelial progenitor cells (EPCs), with their ability to replace old and injured cells and differentiate into healthy and functional mature endothelial cells, has shifted our view of atherosclerosis as an incurable disease, and merged traditional theories of atherosclerosis pathogenesis with evolving concepts of vascular biology. EPC alterations are involved in the pathogenesis of vascular abnormalities in atherosclerosis, but many questions remain unanswered. Many currently available drugs that impact cardiovascular morbidity and mortality have shown a positive effect on EPC biology. This review examines the role of endothelial progenitor cells in atherosclerosis development, and the impact standard antilipemic drugs, including statins, fibrates, and ezetimibe, as well as more novel treatments such as proprotein convertase subtilisin/kexin type 9 (PCSK9) modulating agents and angiopoietin-like proteins (Angtpl3) inhibitors have on EPC biology.
Collapse
Affiliation(s)
- Velimir Altabas
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia
- Correspondence: ; Tel.: +385-1-3787-692
| | | |
Collapse
|
8
|
Canclini L, Malvandi AM, Uboldi P, Jabnati N, Grigore L, Zambon A, Baragetti A, Catapano AL. The Association of Proprotein Convertase Subtilisin/Kexin Type 9 to Plasma Low-Density Lipoproteins: An Evaluation of Different Methods. Metabolites 2021; 11:metabo11120861. [PMID: 34940619 PMCID: PMC8706035 DOI: 10.3390/metabo11120861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 11/18/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type-9 (PCSK9) is key regulator of low-density lipoprotein (LDL) metabolism. A significant proportion of PCSK9 is believed to be associated with LDL in plasma as it circulates, although this finding is still a matter of debate. The purpose of this study was to establish an experimental method to investigate the presence of such an interaction in the bloodstream. We compared a number of well-established methods for lipoprotein (LP) isolation to clarify whether PCSK9 associates differently to circulating lipoproteins, such as KBr gradient ultracentrifugation, physical precipitation of ApoB-LPs, fast protein liquid chromatography (FPLC) and iodixanol gradient ultracentrifugation. Our data show heterogeneity in PCSK9 association to lipoproteins according to the method used. Two methods, iodixanol ultracentrifugation and column chromatography, which did not involve precipitation or high salt concentration, consistently showed an interaction of PCSK9 with a subfraction of LDL that appeared to be more buoyant and have a lower size than average LDL. The percent of PCSK9 association ranged from 2 to 30% and did not appear to correlate to plasma or LDL cholesterol levels. The association of PCSK9 to LDL appeared to be sensitive to high salt concentrations. FPLC and iodixanol gradient ultracentrifugation appeared to be the most suitable methods for the study of this association.
Collapse
Affiliation(s)
- Laura Canclini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.C.); (P.U.); (N.J.); (A.B.)
- IRCCS Multimedica, 20138 Milan, Italy;
| | | | - Patrizia Uboldi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.C.); (P.U.); (N.J.); (A.B.)
| | - Najoua Jabnati
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.C.); (P.U.); (N.J.); (A.B.)
| | - Liliana Grigore
- IRCCS Multimedica, 20099 Sesto San Giovanni, Italy; (L.G.); (A.Z.)
- Center for the Study of Atherosclerosis, Bassini Hospital, 20092 Cinisello Balsamo, Italy
| | - Alberto Zambon
- IRCCS Multimedica, 20099 Sesto San Giovanni, Italy; (L.G.); (A.Z.)
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.C.); (P.U.); (N.J.); (A.B.)
- IRCCS Multimedica, 20138 Milan, Italy;
| | - Alberico Luigi Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.C.); (P.U.); (N.J.); (A.B.)
- IRCCS Multimedica, 20138 Milan, Italy;
- Correspondence: ; Tel.:+39-02-50318302-401; Fax: +39-02-50318386
| |
Collapse
|
9
|
Dal Pino B, Sbrana F, Bigazzi F, Sampietro T. Is treating severe He FH so easy? A combined treatment between lipoprotein apheresis and PCSK9 inhibitors. Transfus Apher Sci 2021; 60:103258. [PMID: 34674949 DOI: 10.1016/j.transci.2021.103258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/09/2021] [Accepted: 08/17/2021] [Indexed: 11/30/2022]
Abstract
Despite advance in pharmacotherapy of lipid disorders, many heterozygous Familial Hypercholesterolemia patients do not achieve a desirable lipid target to significantly reduce the risk of atherosclerotic cardiovascular disease. The aim of the present work is to evaluate the interaction between Lipoprotein apheresis (LA) and PCSK9i in a small FH cohort in which the guidelines therapeutic target is not achieved. During one year, together with a complete adherence to PCSK9i therapy, we recorded a 3 to 5 LA sessions less per year in each patient. This therapeutic approach suggests: i) the possibility of increasing the number of patients treated with LA, ii) the improvement of their quality of life, and iii) the costs reduction for the single patient-treatment.
Collapse
Affiliation(s)
- Beatrice Dal Pino
- Lipoapheresis Unit - Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana "Gabriele Monasterio", Via Moruzzi 1, Pisa, 56124, Italy
| | - Francesco Sbrana
- Lipoapheresis Unit - Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana "Gabriele Monasterio", Via Moruzzi 1, Pisa, 56124, Italy.
| | - Federico Bigazzi
- Lipoapheresis Unit - Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana "Gabriele Monasterio", Via Moruzzi 1, Pisa, 56124, Italy
| | - Tiziana Sampietro
- Lipoapheresis Unit - Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana "Gabriele Monasterio", Via Moruzzi 1, Pisa, 56124, Italy
| |
Collapse
|
10
|
Yang Q, Sun D, Pei C, Zeng Y, Wang Z, Li Z, Hao Y, Song X, Li Y, Liu G, Tang Y, Smith SC, Han Y, Huo Y, Ge J, Ma C, Fonarow GC, Morgan L, Liu J, Liu J, Zhou M, Zhao D, Zhou Y, Zhou X. LDL cholesterol levels and in-hospital bleeding in patients on high-intensity antithrombotic therapy: findings from the CCC-ACS project. Eur Heart J 2021; 42:3175-3186. [PMID: 34347859 DOI: 10.1093/eurheartj/ehab418] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/29/2020] [Accepted: 06/17/2021] [Indexed: 11/12/2022] Open
Abstract
AIMS Emerging evidence has linked cholesterol metabolism with platelet responsiveness. We sought to examine the dose-response relationship between low-density lipoprotein cholesterol (LDL-C) and major in-hospital bleeds in acute coronary syndrome (ACS) patients. METHODS AND RESULTS Among 42 378 ACS patients treated with percutaneous coronary intervention (PCI) enrolled in 240 hospitals in the Improving Care for Cardiovascular Disease in China-ACS project from 2014 to 2019, a total of 615 major bleeds, 218 ischaemic events, and 337 deaths were recorded. After controlling for baseline variables, a non-linear relationship was observed for major bleeds, with the higher risk at lower LDL-C levels. No dose-response relationship was identified for ischaemic events and mortality. A threshold value of LDL-C <70 mg/dL was associated with an increased risk for major bleeds (adjusted odds ratio: 1.49; 95% confidence interval: 1.21-1.84) in multivariable-adjusted logistic regression models and in propensity score-matched cohorts. The results were consistent in multiple sensitivity analyses. Among ticagrelor-treated patients, the LDL-C threshold for increased bleeding risk was observed at <88 mg/dL, whereas for clopidogrel-treated patients, the threshold was <54 mg/dL. Across a full spectrum of LDL-C levels, the treatment effect size associated with ticagrelor vs. clopidogrel on major bleeds favoured clopidogrel at lower LDL-C levels, but no difference at higher LDL-C levels. CONCLUSIONS In a nationwide ACS registry, a non-linear association was identified between LDL-C levels and major in-hospital bleeds following PCI, with the higher risk at lower levels. As the potential for confounding may exist, further studies are warranted. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02306616.
Collapse
Affiliation(s)
- Qing Yang
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Dongdong Sun
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Chongzhe Pei
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Yuhong Zeng
- Departments of Epidemiology and Cardiology, Beijing Anzhen Hospital, Capital Medical University, the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Zhuoqun Wang
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Ziping Li
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Yongchen Hao
- Departments of Epidemiology and Cardiology, Beijing Anzhen Hospital, Capital Medical University, the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Xiwen Song
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Yongle Li
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Gang Liu
- Heart Center, the First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang 050000, Hebei, China
| | - Yida Tang
- Department of Cardiology, Peking University Third Hospital, 49 Huayuanbei Road, Haidian District, Beijing 100191, China
| | - Sidney C Smith
- Division of Cardiology, University of North Carolina at Chapel Hill, 6031 Burnett-Womack Building, Chapel Hill, NC 27599-7075, USA
| | - Yaling Han
- Department of Cardiology, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenhe District, Shenyang 110801, Liaoning, China
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, 100034 Beijing, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, 200032 Shanghai, China
| | - Changsheng Ma
- Departments of Epidemiology and Cardiology, Beijing Anzhen Hospital, Capital Medical University, the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Gregg C Fonarow
- Division of Cardiology, David Geffen School of Medicine at University of California, Los Angeles, 100 UCLA Medical Plaza, Los Angeles, CA 90095, USA
| | - Louise Morgan
- International Quality Improvement Department, American Heart Association, 7272 Greenville Ave, Dallas, TX 75231, USA
| | - Jing Liu
- Departments of Epidemiology and Cardiology, Beijing Anzhen Hospital, Capital Medical University, the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Jun Liu
- Departments of Epidemiology and Cardiology, Beijing Anzhen Hospital, Capital Medical University, the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Mengge Zhou
- Departments of Epidemiology and Cardiology, Beijing Anzhen Hospital, Capital Medical University, the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Dong Zhao
- Departments of Epidemiology and Cardiology, Beijing Anzhen Hospital, Capital Medical University, the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Yujie Zhou
- Departments of Epidemiology and Cardiology, Beijing Anzhen Hospital, Capital Medical University, the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Xin Zhou
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| |
Collapse
|
11
|
Farmaki P, Damaskos C, Garmpis N, Garmpi A, Savvanis S, Diamantis E. PCSK9 Inhibitors and Cardiovascular Disease: Impact on Cardiovascular Outcomes. Curr Drug Discov Technol 2021; 17:138-146. [PMID: 30526464 DOI: 10.2174/1570163816666181211112358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/21/2018] [Accepted: 11/09/2018] [Indexed: 12/20/2022]
Abstract
Cardiovascular Disease (CVD) remains the leading cause of morbidity and mortality in the western world. Hypolipidemic drugs have long been used for the primary and secondary prevention of heart disease. However, the high frequency of recurrent events in patients despite hypolipidemic therapy has increased the need for new more targeted therapeutic approaches. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors are monoclonal antibodies to the PCSK9 gene and represent a new class of drugs that have been shown to further decrease LDL-C when administered as a monotherapy or in combination with statins. In addition to LDL reduction, PCSK9 inhibitors are shown to decrease apolipoprotein B and lipoprotein (a) levels without major adverse effects. Whether or not PCSK9 inhibitors can actually reduce the incidence of cardiovascular events and ameliorate CVD prognosis is yet to be clarified. This review summarizes recent literature on the safety and efficacy of PCSK9 inhibitors on CVD outcome and its potential role in the management of patients with high-risk cardiovascular disease.
Collapse
Affiliation(s)
- Paraskevi Farmaki
- First Department of Pediatrics, Aghia Sophia Children's Hospital, Athens, Greece
| | - Christos Damaskos
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Garmpis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Garmpi
- Internal Medicine Department, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Spyridon Savvanis
- Department of Internal Medicine, General Hospital of Athens "Elpis", Athens, Greece
| | - Evangelos Diamantis
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos Hospital, Athens, Greece
| |
Collapse
|
12
|
Ramin-Mangata S, Thedrez A, Nativel B, Diotel N, Blanchard V, Wargny M, Aguesse A, Billon-Crossouard S, Vindis C, Le May C, Hulin P, Armanet M, Gmyr V, Pattou F, Croyal M, Meilhac O, Nobécourt E, Cariou B, Lambert G. Effects of proprotein convertase subtilisin kexin type 9 modulation in human pancreatic beta cells function. Atherosclerosis 2021; 326:47-55. [PMID: 33933263 DOI: 10.1016/j.atherosclerosis.2021.03.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/09/2021] [Accepted: 03/30/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND AIMS Proprotein Convertase Subtilisin Kexin Type 9 (PCSK9) is an endogenous inhibitor of the LDL receptor (LDLR). Mendelian randomization studies suggest that PCSK9 deficiency increases diabetes risk, but the underlying mechanisms remain unknown. The aim of our study was to investigate whether PCSK9 or its inhibition may modulate beta cell function. METHODS We assessed PCSK9 and insulin colocalization in human pancreatic sections by epifluorescent and confocal microscopy. We also investigated the expression and the function of PCSK9 in the human EndoC-βH1 beta cell line, by ELISA and flow cytometry, respectively. PCSK9 was inhibited with Alirocumab or siRNA. LDLR expression and LDL uptake were assessed by flow cytometry. RESULTS PCSK9 was expressed and secreted from beta cells isolated from human pancreas as well as from EndoC-βH1 cells. PCSK9 secretion was enhanced by statin treatment. Recombinant PCSK9 decreased LDLR abundance at the surface of these cells, an effect abrogated by Alirocumab. Alirocumab as well as PCSK9 silencing increased LDLR expression at the surface of EndoC-βH1 cells. Neither exogenous PCSK9, nor Alirocumab, nor PCSK9 silencing significantly altered glucose-stimulated insulin secretion (GSIS) from these cells. High-low density lipoproteins (LDL) concentrations decreased GSIS, but the addition of PCSK9 or its inhibition did not modulate this phenomenon. CONCLUSIONS While PCSK9 regulates LDLR abundance in beta cells, inhibition of exogenous or endogenous PCSK9 does not appear to significantly impact insulin secretion. This is reassuring for the safety of PCSK9 inhibitors in terms of beta cell function.
Collapse
Affiliation(s)
| | - Aurélie Thedrez
- Université de Nantes, CRNH Ouest, Inra UMR 1280 PhAN, Nantes, France; L'institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Brice Nativel
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France
| | - Nicolas Diotel
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France
| | - Valentin Blanchard
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France
| | - Matthieu Wargny
- L'institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France; CHU Nantes, INSERM, CIC 1413, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des Données, Nantes, F-44093, France
| | - Audrey Aguesse
- Université de Nantes, CRNH Ouest, Inra UMR 1280 PhAN, Nantes, France
| | | | | | - Cédric Le May
- L'institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Philippe Hulin
- Université de Nantes, CHU de Nantes, Inserm UMS 016, Cnrs UMS 3556, Structure Fédérative de Recherche François Bonamy, Micropicell Facility, Nantes, France
| | - Mathieu Armanet
- Cell Therapy Unit, Hôpital Saint Louis, AP-HP, Université Paris Diderot, Paris, France
| | - Valery Gmyr
- European Genomic Institute for Diabetes, Inserm UMR 1190 Translational Research for Diabetes, University of Lille 2, Lille, France
| | - François Pattou
- European Genomic Institute for Diabetes, Inserm UMR 1190 Translational Research for Diabetes, University of Lille 2, Lille, France; Lille University Hospital, Lille, France
| | - Mikaël Croyal
- Université de Nantes, CRNH Ouest, Inra UMR 1280 PhAN, Nantes, France
| | - Olivier Meilhac
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France
| | - Estelle Nobécourt
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France; CHU de La Réunion, Service d'Endocrinologie Nutrition, Saint-Pierre, France
| | - Bertrand Cariou
- L'institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Gilles Lambert
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France.
| |
Collapse
|
13
|
Burnap SA, Mayr M. Lipoprotein compartmentalisation as a regulator of PCSK9 activity. J Mol Cell Cardiol 2021; 155:21-24. [PMID: 33617869 DOI: 10.1016/j.yjmcc.2021.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/29/2021] [Accepted: 02/09/2021] [Indexed: 10/22/2022]
Affiliation(s)
- Sean A Burnap
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - Manuel Mayr
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom.
| |
Collapse
|
14
|
Oleaga C, Hay J, Gurcan E, David LL, Mueller PA, Tavori H, Shapiro MD, Pamir N, Fazio S. Insights into the kinetics and dynamics of the furin-cleaved form of PCSK9. J Lipid Res 2020; 62:100003. [PMID: 33429337 PMCID: PMC7890205 DOI: 10.1194/jlr.ra120000964] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/27/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates cholesterol metabolism by inducing the degradation of hepatic low density lipoprotein receptors (LDLRs). Plasma PCSK9 has 2 main molecular forms: a 62 kDa mature form (PCSK9_62) and a 55 kDa, furin-cleaved form (PCSK9_55). PCSK9_55 is considered less active than PCSK9_62 in degrading LDLRs. We aimed to identify the site of PCSK9_55 formation (intracellular vs. extracellular) and to further characterize the LDLR-degradative function of PCSK9_55 relative to PCSK9_62. Coexpressing PCSK9_62 with furin in cell culture induced formation of PCSK9_55, most of which was found in the extracellular space. Under the same conditions, we found that i) adding a cell-permeable furin inhibitor preferentially decreased the formation of PCSK9_55 extracellularly; ii) using pulse-chase analysis, we observed the formation of PCSK9_55 exclusively extracellularly in a time-dependent manner. A recombinant form of PCSK9_55 was efficiently produced but displayed impaired secretion that resulted in its intracellular trapping. However, the nonsecreted PCSK9_55 was able to induce degradation of LDLR, though with 50% lower efficiency than PCSK9_62. Collectively, our data show that 1) PCSK9_55 is formed extracellularly; 2) PCSK9_55 has a shorter half-life; 3) there is a small intracellular pool of PCSK9_55 that is not secreted; and 4) PCSK9_55 retained within the cell maintains a reduced efficiency to cause LDLR degradation.
Collapse
Affiliation(s)
- Carlota Oleaga
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Joshua Hay
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Emma Gurcan
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Larry L David
- Proteomics Shared Resource, Oregon Health & Science University, Portland, OR, USA
| | - Paul A Mueller
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Hagai Tavori
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Michael D Shapiro
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Nathalie Pamir
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA.
| | - Sergio Fazio
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
15
|
Banerjee S, Luo P, Reda DJ, Latif F, Hastings JL, Armstrong EJ, Bagai J, Abu-Fadel M, Baskar A, Kamath P, Lippe D, Wei Y, Scrymgeour A, Gleason TC, Brilakis ES. Plaque Regression and Endothelial Progenitor Cell Mobilization With Intensive Lipid Elimination Regimen (PREMIER). Circ Cardiovasc Interv 2020; 13:e008933. [PMID: 32791950 DOI: 10.1161/circinterventions.119.008933] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Low-density lipoproteins (LDLs) are removed by extracorporeal filtration during LDL apheresis. It is mainly used in familial hyperlipidemia. The PREMIER trial (Plaque Regression and Progenitor Cell Mobilization With Intensive Lipid Elimination Regimen) evaluated LDL apheresis in nonfamilial hyperlipidemia acute coronary syndrome patients treated with percutaneous coronary intervention. METHODS We randomized 160 acute coronary syndrome patients at 4 Veterans Affairs centers within 72 hours of percutaneous coronary intervention to intensive lipid-lowering therapy (ILLT) comprising single LDL apheresis and statins versus standard medical therapy (SMT) with no LDL apheresis and statin therapy alone. Trial objectives constituted primary safety and primary efficacy end points and endothelial progenitor cell colony-forming unit mobilization in peripheral blood. RESULTS Mean LDL reduction at discharge was 53% in ILLT and 17% in SMT groups (P<0.0001) from baseline levels of 116.3±34.3 and 110.7±32 mg/dL (P=0.2979), respectively. The incidence of the primary safety end point of major peri-percutaneous coronary intervention adverse events was similar in both groups (ILLT, 3; SMT, 0). The primary efficacy end point, percentage change in total plaque volume at 90 days by intravascular ultrasound, on average decreased by 4.81% in the ILLT group and increased by 2.31% in the SMT group (difference of means, -7.13 [95% CI, -14.59 to 0.34]; P=0.0611). The raw change in total plaque volume on average decreased more in the ILLT group than in the SMT group (-6.01 versus -0.95 mm3; difference of means, -5.06 [95% CI, -11.61 to 1.48]; P=0.1286). Similar results were obtained after adjusting for participating sites, age, preexisting coronary artery disease, diabetes mellitus, baseline LDL levels, and baseline plaque burden. There was robust endothelial progenitor cell colony-forming unit mobilization from baseline to 90 days in the ILLT group (P=0.0015) but not in SMT (P=0.0844). CONCLUSIONS PREMIER is the first randomized clinical trial to demonstrate safety and a trend for early coronary plaque regression with LDL apheresis in nonfamilial hyperlipidemia acute coronary syndrome patients treated with percutaneous coronary intervention. Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT01004406 and NCT02347098.
Collapse
Affiliation(s)
- Subhash Banerjee
- Veterans Affairs North Texas Health Care System, Dallas (S.B., J.L.H.).,University of Texas Southwestern Medical Center, Dallas (S.B., J.L.H., A.B., P.K.)
| | - Ping Luo
- Cooperative Studies Program Coordinating Center, Edward Hines, Jr Veterans Affairs Hospital, Hines, IL (P.L., D.J.R., D.L., Y.W.)
| | - Domenic J Reda
- Cooperative Studies Program Coordinating Center, Edward Hines, Jr Veterans Affairs Hospital, Hines, IL (P.L., D.J.R., D.L., Y.W.)
| | - Faisal Latif
- Oklahoma City Veterans Affairs Medical Center (F.L.).,University of Oklahoma Health Sciences Center (F.L., M.A.-F.)
| | - Jeffrey L Hastings
- Veterans Affairs North Texas Health Care System, Dallas (S.B., J.L.H.).,University of Texas Southwestern Medical Center, Dallas (S.B., J.L.H., A.B., P.K.)
| | - Ehrin J Armstrong
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO (E.J.A.)
| | - Jayant Bagai
- Veterans Affairs Tennessee Valley Health Care System, Nashville (J.B.)
| | - Mazen Abu-Fadel
- University of Oklahoma Health Sciences Center (F.L., M.A.-F.)
| | - Amutharani Baskar
- University of Texas Southwestern Medical Center, Dallas (S.B., J.L.H., A.B., P.K.)
| | - Preeti Kamath
- University of Texas Southwestern Medical Center, Dallas (S.B., J.L.H., A.B., P.K.)
| | - Daniel Lippe
- Cooperative Studies Program Coordinating Center, Edward Hines, Jr Veterans Affairs Hospital, Hines, IL (P.L., D.J.R., D.L., Y.W.)
| | - Yongliang Wei
- Cooperative Studies Program Coordinating Center, Edward Hines, Jr Veterans Affairs Hospital, Hines, IL (P.L., D.J.R., D.L., Y.W.)
| | - Alexandra Scrymgeour
- Cooperative Studies Program Clinical Research Pharmacy Coordinating Center, Albuquerque, NM (A.S.)
| | - Theresa C Gleason
- Department of Veterans Affairs, Office of Research and Development, Washington, DC (T.C.G.)
| | - Emmanouil S Brilakis
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, MN (E.S.B.)
| |
Collapse
|
16
|
Apolipoprotein(a) phenotype determines the correlations of lipoprotein(a) and proprotein convertase subtilisin/kexin type 9 levels in patients with potential familial hypercholesterolemia. Atherosclerosis 2019; 277:477-482. [PMID: 30270088 DOI: 10.1016/j.atherosclerosis.2018.08.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 06/01/2018] [Accepted: 08/17/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND AIMS The aim of this study is to investigate the relation between lipoprotein(a) [Lp(a)] and proprotein convertase subtilisin/kexin type 9 (PCSK9) concentrations, and their complex, in patients with potential familial hypercholesterolemia (FH), depending on apo(a) phenotype. METHODS The study included 205 patients with total cholesterol (TC) > 7.5 mmol/L and/or low density lipoprotein cholesterol (LDL-C)>4.9 mmol/L, 32 (15%) patients suffered from ischemic heart disease (IHD), 64 were taking statins. The diagnosis of FH was estimated according to the Dutch Lipid Clinics Network criteria. Lipid parameters, apoB-containing lipoprotein subfractions, Lp(a), PCSK9, Lp(a)-PCSK9 complex levels and apo(a) phenotype were determined. Depending on the apo(a) phenotype, all patients were divided into 2 groups: with high molecular weight (HMW) (n = 145) and low molecular weight (LMW) (n = 60) apo(a) phenotype. RESULTS The groups were comparable by all major clinical characteristics and biochemical parameters. In the whole group, PCSK9 concentration correlated with age, statins intake, Lp(a), TC and TG levels. Correlation between Lp(a) and PCSK9 levels was found only in the LMW apo(a) phenotype group independently of statins intake (r = 0.46, p < 0.001). Associations between Lp(a)-PCSK9 complex and large subfractions of intermediate (r = 0.30) and low-density lipoproteins (r = 0.30, p < 0.05 for both) were observed, with more significance in group 2 (r = 0.59, p < 0.005 and r = 0.40, p < 0.05, respectively). CONCLUSIONS In patients with potential familial hypercholesterolemia, positive correlations between concentrations of Lp(a) and PCSK9, as well as of Lp(a)-PCSK9 plasma complex with large subfractions of intermediate and low-density lipoproteins (IDL-1 and LDL-C), were determined by the LMW apo(a) phenotype.
Collapse
|
17
|
Tsimikas S, Fazio S, Ferdinand KC, Ginsberg HN, Koschinsky ML, Marcovina SM, Moriarty PM, Rader DJ, Remaley AT, Reyes-Soffer G, Santos RD, Thanassoulis G, Witztum JL, Danthi S, Olive M, Liu L. NHLBI Working Group Recommendations to Reduce Lipoprotein(a)-Mediated Risk of Cardiovascular Disease and Aortic Stenosis. J Am Coll Cardiol 2019; 71:177-192. [PMID: 29325642 DOI: 10.1016/j.jacc.2017.11.014] [Citation(s) in RCA: 326] [Impact Index Per Article: 65.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
Pathophysiological, epidemiological, and genetic studies provide strong evidence that lipoprotein(a) [Lp(a)] is a causal mediator of cardiovascular disease (CVD) and calcific aortic valve disease (CAVD). Specific therapies to address Lp(a)-mediated CVD and CAVD are in clinical development. Due to knowledge gaps, the National Heart, Lung, and Blood Institute organized a working group that identified challenges in fully understanding the role of Lp(a) in CVD/CAVD. These included the lack of research funding, inadequate experimental models, lack of globally standardized Lp(a) assays, and inadequate understanding of the mechanisms underlying current drug therapies on Lp(a) levels. Specific recommendations were provided to facilitate basic, mechanistic, preclinical, and clinical research on Lp(a); foster collaborative research and resource sharing; leverage expertise of different groups and centers with complementary skills; and use existing National Heart, Lung, and Blood Institute resources. Concerted efforts to understand Lp(a) pathophysiology, together with diagnostic and therapeutic advances, are required to reduce Lp(a)-mediated risk of CVD and CAVD.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Vascular Medicine Program, Sulpizio Cardiovascular Center, Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, California.
| | - Sergio Fazio
- Oregon Health & Science University, Portland, Oregon
| | | | - Henry N Ginsberg
- College of Physicians and Surgeons, Columbia University, New York, New York
| | - Marlys L Koschinsky
- Robarts Research Institute and Department of Physiology & Pharmacology Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | | | | | - Daniel J Rader
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alan T Remaley
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Raul D Santos
- Heart Institute (InCor) University of Sao Paulo Medical School Hospital and Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | | | - Joseph L Witztum
- Division of Endocrinology, Department of Medicine, University of California San Diego, La Jolla, California
| | - Simhan Danthi
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Michelle Olive
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lijuan Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
18
|
Makino H, Koezuka R, Tamanaha T, Ogura M, Matsuki K, Hosoda K, Harada-Shiba M. Familial Hypercholesterolemia and Lipoprotein Apheresis. J Atheroscler Thromb 2019; 26:679-687. [PMID: 31231083 PMCID: PMC6711846 DOI: 10.5551/jat.rv17033] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Lipoprotein apheresis has been developed as the treatment for refractory familial hypercholesterolemia (FH) to remove low-density lipoprotein (LDL), which is the main pathogenic factor. Currently, three procedures are available in Japan, including the plasma exchange, double-membrane filtration, and selective LDL adsorption. Selective LDL adsorption, which was developed in Japan, has been one of the most common treatment methods in the world. Lipoprotein apheresis enabled the prevention of atherosclerosis progression even in homozygous FH (HoFH) patients. However, in our observational study, HoFH patients who started lipoprotein apheresis in adulthood had a poorer prognosis than those who started in childhood. Therefore, HoFH patients need to start lipoprotein apheresis as early as possible. Although the indication for lipoprotein apheresis in heterozygous FH (HeFH) patients has been decreasing with the advent of strong statins, our observational study showed that HeFH patients who discontinued lipoprotein apheresis had a poorer prognosis than patients who continued apheresis therapy. These results suggest that it is beneficial for very-high-risk HeFH patients to be treated by lipoprotein apheresis even if their LDL cholesterol is controlled well by lipid-lowering agents. Since launching a new class of lipid-lowering agents, proprotein convertase subtilisin/kexin type 9 (PCSK9) antibody and microsome triglyceride transfer protein inhibitors, the indication for lipoprotein apheresis in FH has been changing. However, despite the development of these drugs, lipoprotein apheresis is still an option with a high therapeutic effect for FH patients with severe atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Hisashi Makino
- Department of Diabetes and Lipid Metabolism, National Cerebral and Cardiovascular Center
| | - Ryo Koezuka
- Department of Diabetes and Lipid Metabolism, National Cerebral and Cardiovascular Center
| | - Tamiko Tamanaha
- Department of Diabetes and Lipid Metabolism, National Cerebral and Cardiovascular Center
| | - Masatsune Ogura
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute
| | - Kota Matsuki
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute
| | - Kiminori Hosoda
- Department of Diabetes and Lipid Metabolism, National Cerebral and Cardiovascular Center
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute
| |
Collapse
|
19
|
Adorni MP, Ruscica M, Ferri N, Bernini F, Zimetti F. Proprotein Convertase Subtilisin/Kexin Type 9, Brain Cholesterol Homeostasis and Potential Implication for Alzheimer's Disease. Front Aging Neurosci 2019; 11:120. [PMID: 31178716 PMCID: PMC6538876 DOI: 10.3389/fnagi.2019.00120] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/07/2019] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) has been associated with dysregulation of brain cholesterol homeostasis. Proprotein convertase subtilisin/kexin type 9 (PCSK9), beyond the known role in the regulation of plasma low-density lipoprotein cholesterol, was first identified in the brain with a potential involvement in brain development and apoptosis. However, its role in the central nervous system (CNS) and in AD pathogenesis is still far from being understood. While in vitro and in vivo evidence led to controversial results, genetic studies apparently did not find an association between PCSK9 loss of function mutations and AD risk or prevalence. In addition, a potential impairment of cognitive performances by the treatment with the PCSK9 inhibitors, alirocumab and evolocumab, have been excluded, although ongoing studies with longer follow-up will provide further insights. PCSK9 is able to affect the expression of neuronal receptors involved in cholesterol homeostasis and neuroinflammation, and higher PCSK9 concentrations have been found in the cerebrospinal fluid (CSF) of AD patients. In this review article, we critically examined the science of PCSK9 with respect to its modulatory role of the mechanisms underlying the pathogenesis of AD. In addition, based on literature data, we made the hypothesis to consider brain PCSK9 as a negative modulator of brain cholesterol homeostasis and neuroinflammation and a potential pharmacological target for treatment.
Collapse
Affiliation(s)
- Maria Pia Adorni
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Massimiliano Ruscica
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Nicola Ferri
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padova, Italy
| | - Franco Bernini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Francesca Zimetti
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| |
Collapse
|
20
|
Macchi C, Banach M, Corsini A, Sirtori CR, Ferri N, Ruscica M. Changes in circulating pro-protein convertase subtilisin/kexin type 9 levels - experimental and clinical approaches with lipid-lowering agents. Eur J Prev Cardiol 2019; 26:930-949. [PMID: 30776916 DOI: 10.1177/2047487319831500] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Regulation of pro-protein convertase subtilisin/kexin type 9 (PCSK9) by drugs has led to the development of a still small number of agents with powerful activity on low-density lipoprotein cholesterol levels, associated with a significant reduction of cardiovascular events in patients in secondary prevention. The Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk (FOURIER) and Evaluation of Cardiovascular Outcomes After an Acute Coronary Syndrome During Treatment With Alirocumab (ODYSSEY OUTCOMES) studies, with the two available PCSK9 antagonists, i.e. evolocumab and alirocumab, both reported a 15% reduction in major adverse cardiovascular events. Regulation of PCSK9 expression is dependent upon a number of factors, partly genetic and partly associated to a complex transcriptional system, mainly controlled by sterol regulatory element binding proteins. PCSK9 is further regulated by concomitant drug treatments, particularly by statins, enhancing PCSK9 secretion but decreasing its stimulatory phosphorylated form (S688). These complex transcriptional mechanisms lead to variable circulating levels making clinical measurements of plasma PCSK9 for cardiovascular risk assessment a debated matter. Determination of total PCSK9 levels may provide a diagnostic tool for explaining an apparent resistance to PCSK9 inhibitors, thus indicating the need for other approaches. Newer agents targeting PCSK9 are in clinical development with a major interest in those with a longer duration of action, e.g. RNA silencing, allowing optimal patient compliance. Interest has been expanded to areas not only limited to low-density lipoprotein cholesterol reduction but also investigating other non-lipid pathways raising cardiovascular risk, in particular inflammation associated to raised high-sensitivity C-reactive protein levels, not significantly affected by the present PCSK9 antagonists.
Collapse
Affiliation(s)
- C Macchi
- 1 Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Italy
| | - M Banach
- 2 Department of Hypertension, Medical University of Lodz, Poland.,3 Polish Mother's Memorial Hospital Research Institute (PMMHRI), Poland.,4 Cardiovascular Research Centre, University of Zielona Gora, Poland
| | - A Corsini
- 1 Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Italy.,5 Multimedica IRCCS, Italy
| | - C R Sirtori
- 6 Dyslipidemia Center, A.S.S.T. Grande Ospedale Metropolitano Niguarda, Italy
| | - N Ferri
- 7 Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Italy
| | - M Ruscica
- 1 Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Italy
| |
Collapse
|
21
|
Galvan AM, Chorba JS. Cell-associated heparin-like molecules modulate the ability of LDL to regulate PCSK9 uptake. J Lipid Res 2018; 60:71-84. [PMID: 30463987 DOI: 10.1194/jlr.m087189] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 10/30/2018] [Indexed: 01/12/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) targets the LDL receptor (LDLR) for degradation, increasing plasma LDL and, consequently, cardiovascular risk. Uptake of secreted PCSK9 is required for its effect on the LDLR, and LDL itself inhibits this uptake, though how it does so remains unclear. In this study, we investigated the relationship between LDL, the PCSK9:LDLR interaction, and PCSK9 uptake. We show that LDL inhibits binding of PCSK9 to the LDLR in vitro more impressively than it inhibits PCSK9 uptake in cells. Furthermore, cell-surface heparin-like molecules (HLMs) can partly explain this difference, consistent with heparan sulfate proteoglycans (HSPGs) acting as coreceptors for PCSK9. We also show that HLMs can interact with either PCSK9 or LDL to modulate the inhibitory activity of LDL on PCSK9 uptake, with such inhibition rescued by competition with the entire PCSK9 prodomain, but not its truncated variants. Additionally, we show that the gain-of-function PCSK9 variant, S127R, located in the prodomain near the HSPG binding site, exhibits increased affinity for HLMs, potentially explaining its phenotype. Overall, our findings suggest a model where LDL acts as a negative regulator of PCSK9 function by decreasing its uptake via direct interactions with either the LDLR or HLMs.
Collapse
Affiliation(s)
- Adri M Galvan
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA 94143
| | - John S Chorba
- Division of Cardiology, Zuckerberg San Francisco General, Department of Medicine, University of California San Francisco, San Francisco, CA 94110
| |
Collapse
|
22
|
Ruscica M, Simonelli S, Botta M, Ossoli A, Lupo MG, Magni P, Corsini A, Arca M, Pisciotta L, Veglia F, Franceschini G, Ferri N, Calabresi L. Plasma PCSK9 levels and lipoprotein distribution are preserved in carriers of genetic HDL disorders. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:991-997. [DOI: 10.1016/j.bbalip.2018.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 04/18/2018] [Accepted: 05/27/2018] [Indexed: 01/27/2023]
|
23
|
Zenti MG, Altomari A, Lupo MG, Botta M, Bonora E, Corsini A, Ruscica M, Ferri N. From lipoprotein apheresis to proprotein convertase subtilisin/kexin type 9 inhibitors: Impact on low-density lipoprotein cholesterol and C-reactive protein levels in cardiovascular disease patients. Eur J Prev Cardiol 2018; 25:1843-1851. [DOI: 10.1177/2047487318792626] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this observational study, we compared the effect of lipoprotein apheresis and evolocumab or alirocumab on levels of lipoprotein cholesterol, triglycerides and inflammatory markers (C reactive protein and interleukin 6) in cardiovascular patients ( n = 9). Patients were monitored during the last year of lipoprotein apheresis followed by six months of treatment with proprotein convertase subtilisin/kexin type 9 inhibitors. The biochemical parameters were determined pre- and post- every apheresis procedure for 12 months and then after one, three and six months of treatment with evolocumab (140 mg every two weeks [Q2W]) or alirocumab (75 mg or 150 mg every two weeks [Q2W]). Lipoprotein apheresis significantly reduced low-density lipoprotein cholesterol levels from 138 ± 32 mg/dl to 46 ± 16 mg/dl ( p < 0.001), with an inter-apheresis level of 114 ± 26 mg/dl. Lipoprotein(a) was also reduced from a median of 42 mg/dl to 17 mg/dl ( p < 0.01). Upon anti-proprotein convertase subtilisin/kexin type 9 therapy, low-density lipoprotein cholesterol levels were similar to post-apheresis (59 ± 25, 41 ± 22 and 42 ± 21mg/dl at one, three and six months, respectively) as well as those of lipoprotein(a) (18 mg/dl). However, an opposite effect was observed on high-density lipoprotein cholesterol levels: –16.0% from pre- to post-apheresis and +34.0% between pre-apheresis and proprotein convertase subtilisin/kexin type 9 inhibitors. Apheresis significantly reduced high-sensitivity C-reactive protein levels (1.5 ± 1.2 mg/l pre-apheresis to 0.6 ± 0.6 mg/l post-apheresis), while no changes were found upon proprotein convertase subtilisin/kexin type 9 mAbs administration. In conclusion, our study demonstrated that, by switching from lipoprotein apheresis to anti-proprotein convertase subtilisin/kexin type 9 therapies, patients reached similar low-density lipoprotein cholesterol and lipoprotein(a) levels, increased those of high-density lipoprotein cholesterol, and showed no changes on high-sensitivity C-reactive protein.
Collapse
Affiliation(s)
- Maria G Zenti
- Divisione di Endocrinologia, Diabetologia e Metabolismo, Università degli Studi di Verona, Italy
| | - Anna Altomari
- Divisione di Endocrinologia, Diabetologia e Metabolismo, Università degli Studi di Verona, Italy
| | - Maria G Lupo
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Italy
| | - Margherita Botta
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Italy
| | - Enzo Bonora
- Divisione di Endocrinologia, Diabetologia e Metabolismo, Università degli Studi di Verona, Italy
| | - Alberto Corsini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Italy
- IRCCS, Multimedica, Italy
| | - Massimiliano Ruscica
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Italy
| | - Nicola Ferri
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Italy
| |
Collapse
|
24
|
Rosenson RS, Hegele RA, Fazio S, Cannon CP. The Evolving Future of PCSK9 Inhibitors. J Am Coll Cardiol 2018; 72:314-329. [DOI: 10.1016/j.jacc.2018.04.054] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 01/09/2023]
|
25
|
PCSK9 inhibition with alirocumab reduces lipoprotein(a) levels in nonhuman primates by lowering apolipoprotein(a) production rate. Clin Sci (Lond) 2018; 132:1075-1083. [DOI: 10.1042/cs20180040] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/12/2018] [Accepted: 05/03/2018] [Indexed: 12/24/2022]
Abstract
Therapeutic antibodies targeting proprotein convertase subtilisin kexin type 9 (PCSK9) (e.g. alirocumab) lower low-density lipoprotein cholesterol (LDL-C) and lipoprotein (a) [Lp(a)] levels in clinical trials. We recently showed that PCSK9 enhances apolipoprotein(a) [apo(a)] secretion from primary human hepatocytes but does not affect Lp(a) cellular uptake. Here, we aimed to determine how PCSK9 neutralization modulates Lp(a) levels in vivo.
Six nonhuman primates (NHP) were treated with alirocumab or a control antibody (IgG1) in a crossover protocol. After the lowering of lipids reached steady state, NHP received an intravenous injection of [2H3]-leucine, and blood samples were collected sequentially over 48 h. Enrichment of apolipoproteins in [2H3]-leucine was assessed by liquid chromatography–tandem mass spectrometry (LC–MS/MS). Kinetic parameters were calculated using numerical models with the SAAMII software. Compared with IgG1, alirocumab significantly reduced total cholesterol (TC) (−28%), LDL-C (−67%), Lp(a) (−56%), apolipoprotein B100 (apoB100) (−53%), and apo(a) (−53%). Alirocumab significantly increased the fractional catabolic rate of apoB100 (+29%) but not that of apo(a). Conversely, alirocumab sharply and significantly reduced the production rate (PR) of apo(a) (−42%), but not significantly that of apoB100, compared with IgG1, respectively.
In line with the observations made in human hepatocytes, the present kinetic study establishes that PCSK9 neutralization with alirocumab efficiently reduces circulating apoB100 and apo(a) levels by distinct mechanisms: apoB primarily by enhancing its catabolism and apo(a) primarily by lowering its production.
Collapse
|
26
|
Abstract
Unknown 15 years ago, PCSK9 (proprotein convertase subtilisin/kexin type 9) is now common parlance among scientists and clinicians interested in prevention and treatment of atherosclerotic cardiovascular disease. What makes this story so special is not its recent discovery nor the fact that it uncovered previously unknown biology but rather that these important scientific insights have been translated into an effective medical therapy in record time. Indeed, the translation of this discovery to novel therapeutic serves as one of the best examples of how genetic insights can be leveraged into intelligent target drug discovery. The PCSK9 saga is unfolding quickly but is far from complete. Here, we review major scientific understandings as they relate to the role of PCSK9 in lipoprotein metabolism and atherosclerotic cardiovascular disease and the impact that therapies designed to inhibit its action are having in the clinical setting.
Collapse
Affiliation(s)
- Michael D Shapiro
- From the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland
| | - Hagai Tavori
- From the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland
| | - Sergio Fazio
- From the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland.
| |
Collapse
|
27
|
El Khoury P, Roussel R, Fumeron F, Abou-Khalil Y, Velho G, Mohammedi K, Jacob MP, Steg PG, Potier L, Ghaleb Y, Elbitar S, Ragot S, Andreata F, Caligiuri G, Hadjadj S, Boileau C, Marre M, Abifadel M, Varret M, Hansel B. Plasma proprotein-convertase-subtilisin/kexin type 9 (PCSK9) and cardiovascular events in type 2 diabetes. Diabetes Obes Metab 2018; 20:943-953. [PMID: 29205760 DOI: 10.1111/dom.13181] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/13/2017] [Accepted: 11/30/2017] [Indexed: 12/12/2022]
Abstract
AIM To investigate whether plasma concentrations of proprotein-convertase-subtilisin/kexin type 9 (PCSK9) were associated with cardiovascular (CV) events in two cohorts of patients with type 2 diabetes mellitus. METHODS We considered patients from the DIABHYCAR (n = 3137) and the SURDIAGENE (n = 1468) studies. Baseline plasma PCSK9 concentration was measured using an immunofluorescence assay. In post hoc, but preplanned, analyses we assessed the relationship between PCSK9 and the following endpoints: (1) a combined endpoint of major CV events: CV death, non-fatal myocardial infarction (MI), stroke and heart failure-related hospital admission; (2) a composite of all CV events: MI, stroke, heart failure-related hospital admission, coronary/peripheral angioplasty or bypass, CV death; (3) MI; (4) stroke/transient ischaemic attack (TIA); and (5) CV death. RESULTS In the DIABHYCAR study, plasma PCSK9 tertiles were associated with the incidence of MI, all CV events and stroke/TIA (P for trend <.05). In adjusted Cox analysis, plasma PCSK9 was associated, independently of classic risk factors, with the incidence of major CV events (hazard ratio [HR] for 1-unit increase of log[PCSK9] 1.28 [95% confidence interval {CI} 1.06-1.55]), the incidence of MI (HR 1.66 [95% CI 1.05-2.63]), and the incidence of all CV events (HR 1.22 [95% CI 1.04-1.44]), but not with CV death. Plasma PCSK9 was not associated with the incidence of CV disease in the participants of the SURDIAGENE study with high CV risk treated with statins and insulin. CONCLUSIONS We found that PCSK9 was inconsistently associated with CV events in populations with type 2 diabetes. The association may depend on the level of CV risk and the background treatment.
Collapse
Affiliation(s)
- Petra El Khoury
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
| | - Ronan Roussel
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Frederic Fumeron
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Yara Abou-Khalil
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
| | - Gilberto Velho
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Kamel Mohammedi
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Marie-Paule Jacob
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Philippe Gabriel Steg
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- FACT, HUPNVS, Département de Cardiologie, AP-HP, CHU Xavier Bichat, Paris, France
- NHLI, Imperial College, Royal Brompton Hospital, London, UK
| | - Louis Potier
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Youmna Ghaleb
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Sandy Elbitar
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Stephanie Ragot
- Centre Investigation Clinique 1402, University of Poitiers, Poitiers, France
- Centre Investigation Clinique, CHU Poitiers, Poitiers, France
- Centre Investigation Clinique CIC1402, INSERM, Poitiers, France
| | - Francesco Andreata
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Giusepinna Caligiuri
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Samy Hadjadj
- Centre Investigation Clinique 1402, University of Poitiers, Poitiers, France
- Centre Investigation Clinique, CHU Poitiers, Poitiers, France
- Centre Investigation Clinique CIC1402, INSERM, Poitiers, France
- Pole DUNE, CHU Poitiers, Poitiers, France
- U1082, INSERM, Poitiers, France
| | - Catherine Boileau
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Département de génétique, AP-HP, CHU Xavier Bichat, Paris, France
| | - Michel Marre
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| | - Marianne Abifadel
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratory of Biochemistry and Molecular Therapeutics, Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Pôle Technologie- Santé, Saint-Joseph University, Beirut, Lebanon
| | - Mathilde Varret
- INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Boris Hansel
- Département d'Endocrinologie, Diabétologie et Nutrition, DHU-FIRE, HUPNVS, AP-HP, CHU Xavier Bichat, Paris, France
- Université Paris Diderot-Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, U-1138, Paris, France
| |
Collapse
|
28
|
Viney NJ, Yeang C, Yang X, Xia S, Witztum JL, Tsimikas S. Relationship between "LDL-C", estimated true LDL-C, apolipoprotein B-100, and PCSK9 levels following lipoprotein(a) lowering with an antisense oligonucleotide. J Clin Lipidol 2018; 12:702-710. [PMID: 29574075 DOI: 10.1016/j.jacl.2018.02.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/21/2018] [Accepted: 02/24/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND The laboratory measurement of "low-density lipoprotein cholesterol (LDL-C)" includes the cholesterol content of lipoprotein(a) (Lp(a)-C). OBJECTIVE To estimate the "true" LDL-C in relation to changes in apolipoprotein B-100 (apoB-100) and assess changes in proprotein convertase subtilisin/kexin 9 (PCSK9) levels in patients with elevated Lp(a) treated with IONIS-APO(a)Rx. METHODS: A pooled placebo group (n = 29), and cohort A (n = 24, baseline Lp(a) 50-175 mg/dL) and cohort B (n = 8, baseline Lp(a) > 175 mg/dL) treated with IONIS-APO(a)Rx were studied. Lp(a) particle number, ultracentrifugation-measured "LDL-C", apoB-100, total PCSK9, and lipoprotein-associated PCSK9 (PCSK9-Lp(a), PCSK9-apoB, PCSK9-apoAI) were measured. Lp(a)-cholesterol (Lp(a)-C) and LDL-C corrected for Lp(a)-C (LDL-Ccorr) were calculated. RESULTS Baseline mean (standard deviation) "LDL-C" was 120 (42), 128 (45), and 112 (39) mg/dL in placebo, cohorts A and B, respectively, whereas LDL-Ccorr was 86 (48), 96 (43), and 57 (37) mg/dL (P < .001 compared with placebo), representing 28%, 25%, and 50% lower levels than "LDL-C". Following IONIS-APO(a)Rx treatment at day 85/99, Lp(a) particle number and Lp(a)-C decreased -66.8% and -71.6%, apoB-100 -10.3% and -17.5%, "LDL-C" -11.8% and -22.7%, (P < .001 for all vs placebo), whereas LDL-Ccorr increased +10.4% (P = .66) and +49.9% (P < .001) in cohorts A and B, respectively. Total PCSK9 did not change but PCSK9-Lp(a) decreased with IONIS-APO(a)Rx vs placebo (-39.0% vs +8.4%, P < .001). CONCLUSION LDL-Ccorr is lower than laboratory "LDL-C" in patients with elevated Lp(a). Following apolipoprotein(a) inhibition and decline in Lp(a) and Lp(a)-C, the decline in apoB-100 is consistent with the notion that LDL devoid of apo(a) is cleared faster than Lp(a). These types of analyses may provide insights into the mechanisms of drugs affecting Lp(a) levels in clinical trials.
Collapse
Affiliation(s)
| | - Calvin Yeang
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Xiaohong Yang
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Joseph L Witztum
- Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| | - Sotirios Tsimikas
- Ionis Pharmaceuticals, Carlsbad, CA, USA; Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
29
|
|
30
|
PCSK9: BIOLOGICAL ACTIVITY REGULATION AND CONNECTION WITH LIPID AND CARBOHYDRATE METABOLISM. КЛИНИЧЕСКАЯ ПРАКТИКА 2017. [DOI: 10.17816/clinpract8370-75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a serine protease which plays an important role in the regulation of LDL receptor (LDLR) expression and apolipoprotein B (apoB) lipoprotein cholesterol metabolism. It is well known that hepatic PCSK9 expression, its activity and secretion influence cholesterol homeostasis. An upregulation of PCSK9 causes an increase of LDLR degradation, which results in decrease of apoB lipoprotein uptake, and a consequent increase in plasma lipoprotein concentration, including LDL. Therefore, PCSK9 has become a new target for lipid lowering therapy. The aim of this review is to consider current data on metabolic and dietary regulation of PCSK9 and its effect on cholesterol and apoB lipoproteins metabolism and risk of cardiovascular disease.
Collapse
|
31
|
Fazio S, Minnier J, Shapiro MD, Tsimikas S, Tarugi P, Averna MR, Arca M, Tavori H. Threshold Effects of Circulating Angiopoietin-Like 3 Levels on Plasma Lipoproteins. J Clin Endocrinol Metab 2017; 102. [PMID: 28633452 PMCID: PMC5587068 DOI: 10.1210/jc.2016-4043] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
CONTEXT Angiopoietin-like 3 (ANGPTL3) deficiency in plasma due to loss-of-function gene mutations results in familial combined hypobetalipoproteinemia type 2 (FHBL2) in homozygotes. However, the lipid phenotype in heterozygotes is much milder and does not appear to relate directly to ANGPTL3 levels. Furthermore, the low-density lipoprotein (LDL) phenotype in carriers of ANGPTL3 mutations is unexplained. OBJECTIVE To determine whether reduction below a critical threshold in plasma ANGPTL3 levels is a determinant of lipoprotein metabolism in FHBL2, and to determine whether proprotein convertase subtilisin kexin type 9 (PCSK9) is involved in determining low LDL levels in this condition. DESIGN We studied subjects from 19 families with ANGPTL3 mutations and subjects with familial combined hypobetalipoproteinemia type 1 (FHBL1) due to truncated apolipoprotein B (apoB) species. RESULTS First, total cholesterol, high-density lipoprotein (HDL) cholesterol, triglycerides, and HDL and LDL particle concentration correlated with plasma ANGPTL3 levels but only when the latter was <25% of normal (<60 ng/dL). Second, the very low-density lipoprotein particle concentration correlated strongly with plasma ANGPTL3 when the latter was <58% of normal. Third, both FHBL1 and FHBL2 subjects showed low levels of mature and LDL-bound PCSK9 and higher levels of its furin-cleaved form. Finally, LDL-bound PCSK9 is protected from cleavage by furin and binds to the LDL receptor more strongly than apoB-free PCSK9. CONCLUSIONS Our results suggest that the hypolipidemic effects of ANGPTL3 mutations in FHBL2 are dependent on a threshold of plasma ANGPTL3 levels, with differential effects on various lipoprotein particles. The increased inactivation of PCSK9 by furin in FHBL1 and FHBL2 is likely to cause increased LDL clearance and suggests novel therapeutic avenues.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hagai Tavori
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon 97239
| |
Collapse
|
32
|
Dixon DL, Buckley LF, Trankle CR, Kadariya D, Abbate A. Clinical utility of evolocumab in the management of hyperlipidemia: patient selection and follow-up. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:2121-2129. [PMID: 28744103 PMCID: PMC5513834 DOI: 10.2147/dddt.s114091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Inhibition of PCSK9 is a novel therapeutic strategy aimed at reducing low-density-lipoprotein cholesterol (LDL-C) and cardiovascular risk. Evolocumab is a fully humanized monoclonal antibody that inhibits PCSK9, an enzyme that binds to LDL receptors and prevents them from recycling to the hepatocyte surface. Clinical trials have demonstrated 50%-70% reductions in LDL-C with evolocumab when used in combination with statin therapy. The recent FOURIER trial demonstrated that evolocumab further reduces cardiovascular events, but not mortality, in high-risk patients already receiving statin therapy. Furthermore, evolocumab did not affect neurocognitive function and was not associated with antidrug-antibody production in over 60,000 patient-years of drug exposure. Appropriate candidates for evolocumab primarily are individuals at high cardiovascular risk, including those with familial hypercholesterolemia and/or established cardiovascular disease, who are already on statin therapy. At this time, the use of evolocumab monotherapy seems appropriate only for individuals deemed statin-intolerant despite attempting several statins. Consideration must be given toward patient willingness to self-inject evolocumab and issues concerning third-party coverage, given the current costs of evolocumab.
Collapse
Affiliation(s)
- Dave L Dixon
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy
| | - Leo F Buckley
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy
| | | | - Dinesh Kadariya
- Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | | |
Collapse
|
33
|
Toward an international consensus—Integrating lipoprotein apheresis and new lipid-lowering drugs. J Clin Lipidol 2017; 11:858-871.e3. [DOI: 10.1016/j.jacl.2017.04.114] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/30/2017] [Accepted: 04/15/2017] [Indexed: 12/11/2022]
|
34
|
Krysa JA, Ooi TC, Proctor SD, Vine DF. Nutritional and Lipid Modulation of PCSK9: Effects on Cardiometabolic Risk Factors. J Nutr 2017; 147:473-481. [PMID: 28179493 DOI: 10.3945/jn.116.235069] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/13/2016] [Accepted: 01/10/2017] [Indexed: 11/14/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a serine protease involved in the regulation of LDL receptor (LDLR) expression and apolipoprotein B lipoprotein cholesterol metabolism. Hepatic PCSK9 protein expression, activity, and secretion have been shown to affect cholesterol homeostasis. An upregulation of hepatic PSCK9 protein leads to increased LDLR degradation, resulting in decreased uptake of apoB lipoproteins and a consequent increase in the plasma concentration of these lipoproteins, including LDL and chylomicron remnants. Hence, PCSK9 has become a novel target for lipid-lowering therapies. The aim of this review is to outline current findings on the metabolic and dietary regulation of PCSK9 and effects on cholesterol, apoB lipoprotein metabolism, and cardiovascular disease (CVD) risk. PCSK9 gene and protein expression have been shown to be regulated by metabolic status and the diurnal pattern. In the fasting state, plasma PCSK9 is reduced via modulation of the nuclear transcriptional factors, including sterol regulatory element-binding protein (SREBP) 1c, SREBP2, and hepatocyte nuclear factor 1α. Plasma PCSK9 concentrations are also known to be positively associated with plasma insulin and homeostasis model assessment of insulin resistance, and appear to be regulated by SREBP1c independently of glucose status. Plasma PCSK9 concentrations are stable in response to high-fat or high-protein diets in healthy individuals; however, this response may differ in altered metabolic conditions. Dietary n-3 polyunsaturated fatty acids have been shown to reduce plasma PCSK9 concentration and hepatic PCSK9 mRNA expression, consistent with their lipid-lowering effects, whereas dietary fructose appears to upregulate PCSK9 mRNA expression and plasma PCSK9 concentrations. Further studies are needed to elucidate the mechanisms of how dietary components regulate PCSK9 and effects on cholesterol and apoB lipoprotein metabolism, as well as to delineate the clinical impact of diet on PCSK9 in terms of CVD risk.
Collapse
Affiliation(s)
- Jacqueline A Krysa
- Metabolic and Cardiovascular Diseases Laboratory, University of Alberta, Edmonton, Canada
| | - Teik Chye Ooi
- Department of Medicine, University of Ottawa, Ottawa, Canada; and.,Chronic Disease Program, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, Canada
| | - Spencer D Proctor
- Metabolic and Cardiovascular Diseases Laboratory, University of Alberta, Edmonton, Canada
| | - Donna F Vine
- Metabolic and Cardiovascular Diseases Laboratory, University of Alberta, Edmonton, Canada;
| |
Collapse
|
35
|
Abstract
Even though it is only a little over a decade from the discovery of proprotein convertase subtilisin/kexin type 9 (PCSK9) as a plasma protein that associates with both high and low cholesterol syndromes, a rich body of knowledge has developed, and drugs inhibiting this target have been approved in many markets. While the majority of research in recent years has focused on the impact of therapeutic antagonism of this molecule, important lines of investigation have emerged characterizing its unique physiology as it relates to cholesterol metabolism and atherosclerosis. The PCSK9 story is unfolding rapidly but is far from complete. One chapter that is of particular interest is the possible direct link between PCSK9 and atherosclerosis. This review specifically examines this relationship drawing from data produced from experimental models of plaque biology and inflammation, atherosclerosis imaging studies, and observational epidemiology.
Collapse
Affiliation(s)
- Michael D Shapiro
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology
| | - Sergio Fazio
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology
| |
Collapse
|
36
|
The complexity of lipoprotein (a) lowering by PCSK9 monoclonal antibodies. Clin Sci (Lond) 2017; 131:261-268. [DOI: 10.1042/cs20160403] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 01/04/2023]
Abstract
Since 2012, clinical trials dedicated to proprotein convertase subtilisin kexin type 9 (PCSK9) inhibition with monoclonal antibodies (mAbs) have unambiguously demonstrated robust reductions not only in low-density lipoprotein (LDL) cholesterol (LDL-C) but also in lipoprotein (a) [Lp(a)] levels. The scientific literature published prior to those studies did not provide any evidence for a link between PCSK9 and Lp(a) metabolism. More recent investigations, either in vitro or in vivo, have attempted to unravel the mechanism(s) by which PCSK9 mAbs reduce circulating Lp(a) levels, with some showing a specific implication of the LDL receptor (LDLR) in Lp(a) clearance whereas others found no significant role for the LDLR in that process. This elusive pathway appears clearly distinct from that of the widely prescribed statins that also enhance LDLR function but do not lower circulating Lp (a) levels in humans. So how does PCSK9 inhibition with mAbs reduce Lp(a)? This still remains to be established.
Collapse
|
37
|
Arca M. Old challenges and new opportunities in the clinical management of heterozygous familial hypercholesterolemia (HeFH): The promises of PCSK9 inhibitors. Atherosclerosis 2017; 256:134-145. [DOI: 10.1016/j.atherosclerosis.2016.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 12/17/2022]
|
38
|
Watts GF, Stefanutti C. ODYSSEY ESCAPE: is PCSK9 inhibition the Trojan Horse for the use of lipoprotein apheresis in familial hypercholesterolaemia? Eur Heart J 2016; 37:3596-3599. [PMID: 28087751 DOI: 10.1093/eurheartj/ehw497] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Gerald F Watts
- School of Medicine and Pharmacology, University of Western Australia, Western Australia, Australia .,Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Western Australia, Australia
| | - Claudia Stefanutti
- Extracorporeal Therapeutic Techniques Unit, Immunohematology and Transfusion Medicine, Lipid Clinic and Atherosclerosis Prevention Centre, 'Umberto I' Hospital, Rome, Italy.,Department of Molecular Medicine, 'Sapienza', University of Rome, Rome, Italy
| |
Collapse
|
39
|
Yadav K, Sharma M, Ferdinand KC. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors: Present perspectives and future horizons. Nutr Metab Cardiovasc Dis 2016; 26:853-862. [PMID: 27352986 DOI: 10.1016/j.numecd.2016.05.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 05/04/2016] [Accepted: 05/17/2016] [Indexed: 12/31/2022]
Abstract
AIMS Our comprehensive review highlights the drug development and pharmacogenomics leading to the recent approval of PCSK9 inhibitors. We also review the anticipated future advances into the uses of PCSK9 inhibition. BACKGROUND Despite the present advances in pharmacotherapy, atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of mortality worldwide. Low density lipoprotein-cholesterol (LDL-C) lowering is the primary target for ASCVD risk reduction, showing demonstrable benefits in mortality. However, 70% of events occur even in the presence of statins. This residual risk may be approached with additional LDL-C reduction. Statin intolerance is a common clinical concern affecting adherence and the benefit with statins. There is also significant variation of individual lipid-lowering. Following rapid development, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors have progressed from genetic observations, to mechanistic studies, to closer realization of the goal of CVD risk reduction. This review discusses the science behind PCSK9 inhibition, evidence of trials involving efficacy and safety, and reflections of its present and future role in clinical care, especially in high-risk patients with ASCVD, persons with suboptimal responses to statins and familial hyperlipidemia. Monoclonal antibodies have demonstrated LDL-C lowering of up to 57% as monotherapy and up to 73% when added to statins. Statins have limited efficacy in reduction of LDL-C due to an increased number of LDL-receptors. Elevated lipoprotein (a) levels may also be significantly lowered by PCSK9i. The journey from discovery to PSCK9 target validation took less than five years, and development and approval of therapeutic modalities for PCSK9 inhibitors happened over the next seven. This review highlights the drug development and pharmacogenomics leading to the recent approval of two agents, alirocumab and evolocumab, with a third bococizumab, and other novel approaches to the pathway pending. DATA SYNTHESIS We searched MEDLINE database via Pubmed for reviews, research publications and relevant trials available on PCSK9 inhibition. CONCLUSION Despite decades of medical advances, ASCVD remains one of the major causes of morbidity and mortality worldwide. Statin use has multiplied since the validation of LDL hypothesis, however, it is undeniable a more effective and well-tolerated agent is needed in significant number or patients. With the arrival of the era of unprecedented CV protection with PCSK9 inhibition, this exciting new therapy holds a pivotal promise as the future of lipid management. The data available already indicate safety, tolerability and superb efficacy of these agents, which are already changing contemporary cholesterol management. The rapid translation of innovative basic science research into drug development may lead to CV outcomes reduction and confirm that this pathway will become prominently utilized.
Collapse
Affiliation(s)
- K Yadav
- Tulane Heart and Vascular Institute, Tulane University School of Medicine, 1430 Tulane Avenue, #8548, New Orleans, LA, 70112, USA
| | - M Sharma
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - K C Ferdinand
- Tulane Heart and Vascular Institute, Tulane University School of Medicine, 1430 Tulane Avenue, #8548, New Orleans, LA, 70112, USA.
| |
Collapse
|
40
|
Villard EF, Thedrez A, Blankenstein J, Croyal M, Tran TTT, Poirier B, Le Bail JC, Illiano S, Nobécourt E, Krempf M, Blom DJ, Marais AD, Janiak P, Muslin AJ, Guillot E, Lambert G. PCSK9 Modulates the Secretion But Not the Cellular Uptake of Lipoprotein(a) Ex Vivo: An Effect Blunted by Alirocumab. ACTA ACUST UNITED AC 2016; 1:419-427. [PMID: 29308438 PMCID: PMC5753417 DOI: 10.1016/j.jacbts.2016.06.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Unlike LDL uptake, Lp(a) uptake is not altered by PCSK9 or PCSK9 inhibition in primary human hepatocytes and in primary dermal fibroblasts isolated from familial hypercholesterolemic and non–familial hypercholesterolemic patients. Lp(a) uptake is occurring in the absence of a functional LDL receptor and is not affected by LDL receptor blockade with monoclonal antibodies. Lp(a) cellular binding and whole particle uptake are not altered by PCSK9. The secretion of Lp(a) from primary human hepatocytes is enhanced by PCSK9, an effect that is blunted by PCSK9 inhibition with alirocumab.
To elucidate how the proprotein convertase subtilisin kexin type 9 (PCSK9) inhibitor alirocumab modulates lipoprotein(a) [Lp(a)] plasma levels, the authors performed a series of Lp(a) uptake studies in primary human hepatocytes and dermal fibroblasts and measured Lp(a) secretion from human hepatocytes. They found that Lp(a) cellular uptake occurred in a low-density lipoprotein receptor–independent manner. Neither PCSK9 nor alirocumab altered Lp(a) internalization. By contrast, the secretion of apolipoprotein (a) from human hepatocytes was sharply increased by PCSK9, an effect that was reversed by alirocumab. They propose that PCSK9 does not significantly modulate Lp(a) catabolism, but rather enhances the secretion of Lp(a) from liver cells.
Collapse
Affiliation(s)
| | - Aurélie Thedrez
- Inra UMR 1280, Nantes, France.,Université de Nantes UMR1280, Faculté de Médecine, Nantes, France
| | | | | | | | - Bruno Poirier
- Sanofi Recherche Développement, Chilly-Mazarin, France
| | | | | | - Estelle Nobécourt
- Inra UMR 1280, Nantes, France.,Université de Nantes UMR1280, Faculté de Médecine, Nantes, France
| | - Michel Krempf
- Inra UMR 1280, Nantes, France.,Université de Nantes UMR1280, Faculté de Médecine, Nantes, France
| | - Dirk J Blom
- Division of Lipidology, Department of Internal Medicine, University of Cape Town, Cape Town, South Africa
| | - A David Marais
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Philip Janiak
- Sanofi Recherche Développement, Chilly-Mazarin, France
| | | | | | - Gilles Lambert
- Inra UMR 1280, Nantes, France.,Inserm UMR 1188, Sainte-Clotilde, France.,Université de la Réunion UMR1188, Faculté de Médecine, Saint-Denis, France.,CHU de la Réunion, Saint Denis, France
| |
Collapse
|
41
|
Abstract
Reducing plasma levels of low-density lipoprotein cholesterol (LDL-C) remains the cornerstone in the primary and secondary prevention of cardiovascular disease. However, lack of efficacy and adverse effects mean that a substantial proportion of patients fail to achieve acceptable LDL-C levels with currently available lipid-lowering drugs. Over the last decade, inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as a promising therapeutic strategy to reduce residual cardiovascular disease risk. Binding of PCSK9 to the LDL receptor targets the receptor for lysosomal degradation. The recognition that inhibition of PCSK9 increases LDL receptor activity has led to the development of a number of approaches to directly target PCSK9. Numerous monoclonal antibodies against PCSK9 are currently being evaluated in phase 3 trials, involving various patient categories on different background lipid-lowering therapies. Current evidence shows reductions in LDL-C levels of up to 70 % may be achieved with PCSK9 inhibition, independent of background statin therapy. This review examines the most recent evidence and future prospects for the use of PCSK9 inhibitors in the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- James Latimer
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, M3.131, 3rd Floor William Leech Building, Newcastle upon Tyne, NE2 4HH, UK
| | - Jonathan A Batty
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, M3.131, 3rd Floor William Leech Building, Newcastle upon Tyne, NE2 4HH, UK
- Freeman Hospital, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - R Dermot G Neely
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, M3.131, 3rd Floor William Leech Building, Newcastle upon Tyne, NE2 4HH, UK
- Royal Victoria Infirmary, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Vijay Kunadian
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, M3.131, 3rd Floor William Leech Building, Newcastle upon Tyne, NE2 4HH, UK.
- Freeman Hospital, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
42
|
Norata GD, Tavori H, Pirillo A, Fazio S, Catapano AL. Biology of proprotein convertase subtilisin kexin 9: beyond low-density lipoprotein cholesterol lowering. Cardiovasc Res 2016; 112:429-42. [PMID: 27496869 DOI: 10.1093/cvr/cvw194] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/06/2016] [Indexed: 12/17/2022] Open
Abstract
Proprotein convertase subtilisin kexin 9 (PCSK9) is a key regulator of low-density lipoprotein receptor levels and LDL-cholesterol levels. Loss-of-function mutations in PCSK9 gene are associated with hypocholesterolaemia and protection against cardiovascular disease, identifying PCSK9 inhibition as a valid therapeutic approach to manage hypercholesterolaemia and related diseases. Although PCSK9 is expressed mainly in the liver, it is present also in other tissues and organs with specific functions, raising the question of whether a pharmacological inhibition of PCSK9 to treat hypercholesterolaemia and associated cardiovascular diseases might be helpful or deleterious in non-hepatic tissues. For example, PCSK9 is expressed in the vascular wall, in the kidneys, and in the brain, where it was proposed to play a role in development, neurocognitive process, and neuronal apoptosis. A link between PCSK9 and immunity was also proposed as both sepsis and viral infections are differentially affected in the presence or absence of PCSK9. Despite the increasing number of observations, the debate on the exact roles of PCSK9 in extrahepatic tissues is still ongoing, and as very effective drugs that inhibit PCSK9 have become available to the clinician, a better understanding of the biological roles of PCSK9 is warranted.
Collapse
Affiliation(s)
- Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy Center for the Study of Atherosclerosis, Ospedale Bassini, Cinisello Balsamo, Italy
| | - Hagai Tavori
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Angela Pirillo
- Center for the Study of Atherosclerosis, Ospedale Bassini, Cinisello Balsamo, Italy IRCCS Multimedica, Milan, Italy
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy IRCCS Multimedica, Milan, Italy
| |
Collapse
|
43
|
Affiliation(s)
- Peter P. Toth
- From the CGH Medical Center, Sterling, IL; and Cicccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
44
|
Tavori H, Christian D, Minnier J, Plubell D, Shapiro MD, Yeang C, Giunzioni I, Croyal M, Duell PB, Lambert G, Tsimikas S, Fazio S. PCSK9 Association With Lipoprotein(a). Circ Res 2016; 119:29-35. [PMID: 27121620 DOI: 10.1161/circresaha.116.308811] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 04/26/2016] [Indexed: 12/12/2022]
Abstract
RATIONALE Lipoprotein(a) [Lp(a)] is a highly atherogenic low-density lipoprotein-like particle characterized by the presence of apoprotein(a) [apo(a)] bound to apolipoprotein B. Proprotein convertase subtilisin/kexin type 9 (PCSK9) selectively binds low-density lipoprotein; we hypothesized that it can also be associated with Lp(a) in plasma. OBJECTIVE Characterize the association of PCSK9 and Lp(a) in 39 subjects with high Lp(a) levels (range 39-320 mg/dL) and in transgenic mice expressing either human apo(a) only or human Lp(a) (via coexpression of human apo(a) and human apolipoprotein B). METHODS AND RESULTS We show that PCSK9 is physically associated with Lp(a) in vivo using 3 different approaches: (1) analysis of Lp(a) fractions isolated by ultracentrifugation; (2) immunoprecipitation of plasma using antibodies to PCSK9 and immunodetection of apo(a); (3) ELISA quantification of Lp(a)-associated PCSK9. Plasma PCSK9 levels correlated with Lp(a) levels, but not with the number of kringle IV-2 repeats. PCSK9 did not bind to apo(a) only, and the association of PCSK9 with Lp(a) was not affected by the loss of the apo(a) region responsible for binding oxidized phospholipids. Preferential association of PCSK9 with Lp(a) versus low-density lipoprotein (1.7-fold increase) was seen in subjects with high Lp(a) and normal low-density lipoprotein. Finally, Lp(a)-associated PCSK9 levels directly correlated with plasma Lp(a) levels but not with total plasma PCSK9 levels. CONCLUSIONS Our results show, for the first time, that plasma PCSK9 is found in association with Lp(a) particles in humans with high Lp(a) levels and in mice carrying human Lp(a). Lp(a)-bound PCSK9 may be pursued as a biomarker for cardiovascular risk.
Collapse
Affiliation(s)
- Hagai Tavori
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.).
| | - Devon Christian
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Jessica Minnier
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Deanna Plubell
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Michael D Shapiro
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Calvin Yeang
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Ilaria Giunzioni
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Mikael Croyal
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - P Barton Duell
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Gilles Lambert
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Sotirios Tsimikas
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Sergio Fazio
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.).
| |
Collapse
|
45
|
Walley KR, Francis GA, Opal SM, Stein EA, Russell JA, Boyd JH. The Central Role of Proprotein Convertase Subtilisin/Kexin Type 9 in Septic Pathogen Lipid Transport and Clearance. Am J Respir Crit Care Med 2016; 192:1275-86. [PMID: 26252194 DOI: 10.1164/rccm.201505-0876ci] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microbial cell walls contain pathogenic lipids, including LPS in gram-negative bacteria, lipoteichoic acid in gram-positive bacteria, and phospholipomannan in fungi. These pathogen lipids are major ligands for innate immune receptors and figure prominently in triggering the septic inflammatory response. Alternatively, pathogen lipids can be cleared and inactivated, thus limiting the inflammatory response. Accordingly, biological mechanisms for sequestering and clearing pathogen lipids from the circulation have evolved. Pathogen lipids released into the circulation are initially bound by transfer proteins, notably LPS binding protein and phospholipid transfer protein, and incorporated into high-density lipoprotein particles. Next, LPS binding protein, phospholipid transfer protein, and other transfer proteins transfer these lipids to ApoB-containing lipoproteins, including low-density (LDL) and very-low-density lipoproteins and chylomicrons. Pathogen lipids within these lipoproteins and their remnants are then cleared from the circulation by the liver. Hepatic clearance involves the LDL receptor (LDLR) and possibly other receptors. Once absorbed by the liver, these lipids are then excreted in the bile. Recent evidence suggests pathogen lipid clearance can be modulated. Importantly, reduced proprotein convertase subtilisin/kexin type 9 activity increases recycling of the LDLR and thereby increases LDLR on the surface of hepatocytes, which increases clearance by the liver of pathogen lipids transported in LDL. Increased pathogen lipid clearance, which can be achieved by inhibiting proprotein convertase subtilisin/kexin type 9, may decrease the systemic inflammatory response to sepsis and improve clinical outcomes.
Collapse
Affiliation(s)
- Keith R Walley
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gordon A Francis
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Steven M Opal
- 2 Infectious Disease Division, Memorial Hospital of Rhode Island and Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - Evan A Stein
- 3 Metabolic and Atherosclerosis Research Center, Cincinnati, Ohio
| | - James A Russell
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - John H Boyd
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
46
|
Tavori H, Giunzioni I, Predazzi IM, Plubell D, Shivinsky A, Miles J, Devay RM, Liang H, Rashid S, Linton MF, Fazio S. Human PCSK9 promotes hepatic lipogenesis and atherosclerosis development via apoE- and LDLR-mediated mechanisms. Cardiovasc Res 2016; 110:268-78. [PMID: 26980204 DOI: 10.1093/cvr/cvw053] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 03/08/2016] [Indexed: 01/07/2023] Open
Abstract
AIMS Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes the degradation of hepatic low-density lipoprotein (LDL) receptors (LDLR), thereby, decreasing hepatocyte LDL-cholesterol (LDL-C) uptake. However, it is unknown whether PCSK9 has effects on atherogenesis that are independent of lipid changes. The present study investigated the effect of human (h) PCSK9 on plasma lipids, hepatic lipogenesis, and atherosclerotic lesion size and composition in transgenic mice expressing hPCSK9 (hPCSK9tg) on wild-type (WT), LDLR⁻/⁻, or apoE⁻/⁻ background. METHODS AND RESULTS hPCSK9 expression significantly increased plasma cholesterol (+91%), triglycerides (+18%), and apoB (+57%) levels only in WT mice. The increase in plasma lipids was a consequence of both decreased hepatic LDLR and increased hepatic lipid production, mediated transcriptionally and post-transcriptionally by PCSK9 and dependent on both LDLR and apoE. Despite the lack of changes in plasma lipids in mice expressing hPCSK9 and lacking LDLR (the main target for PCSK9) or apoE (a canonical ligand for the LDLR), hPCSK9 expression increased aortic lesion size in the absence of apoE (268 655 ± 97 972 µm² in hPCSK9tg/apoE⁻/⁻ vs. 189 423 ± 65 700 µm(2) in apoE⁻/⁻) but not in the absence of LDLR. Additionally, hPCSK9 accumulated in the atheroma and increased lesion Ly6C(hi) monocytes (by 21%) in apoE⁻/⁻ mice, but not in LDLR⁻/⁻ mice. CONCLUSIONS PCSK9 increases hepatic lipid and lipoprotein production via apoE- and LDLR-dependent mechanisms. However, hPCSK9 also accumulate in the artery wall and directly affects atherosclerosis lesion size and composition independently of such plasma lipid and lipoprotein changes. These effects of hPCSK9 are dependent on LDLR but are independent of apoE.
Collapse
Affiliation(s)
- Hagai Tavori
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Ilaria Giunzioni
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Irene M Predazzi
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Deanna Plubell
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Anna Shivinsky
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Joshua Miles
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Rachel M Devay
- Rinat Laboratory, Pfizer Inc., South San Francisco, CA, USA
| | - Hong Liang
- Rinat Laboratory, Pfizer Inc., South San Francisco, CA, USA
| | - Shirya Rashid
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada Dalhousie Medicine New Brunswick University, Saint John, New Brunswick, Canada
| | - MacRae F Linton
- Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sergio Fazio
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
47
|
Banerjee Y, Santos RD, Al-Rasadi K, Rizzo M. Targeting PCSK9 for therapeutic gains: Have we addressed all the concerns? Atherosclerosis 2016; 248:62-75. [PMID: 26987067 DOI: 10.1016/j.atherosclerosis.2016.02.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 01/28/2016] [Accepted: 02/16/2016] [Indexed: 02/08/2023]
Abstract
Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) regulates the expression of low-density lipoprotein (LDL)-receptors, through reducing their recycling by binding to the receptor along with LDL and targeting it for lysosomal destruction. PCSK9 also enhances the degradation of very-low-density-lipoprotein receptor (VLDLR) and lipoprotein receptor-related protein 1 (LRP-1) in a LDL-receptor independent manner. This role in lipid homeostasis presents PCSK9 as an attractive target for the therapeutic management of familial hypercholesterolemia as well as other refractory dyslipidaemias. However, PCSK9 mediates multifarious functions independent of its role in lipid homeostasis, which can be grouped under "pleiotropic functions" of the protein. This includes PCSK9's role in: trafficking of epithelial sodium channel; hepatic regeneration; pancreatic integrity and glucose homeostasis; antiviral activity; antimalarial activity; regulation of different cell signalling pathways; cortical neural differentiation; neuronal apoptosis and Alzheimer's disease. The question that needs to be investigated in depth is "How will the pleotropic functions of PCSK9, be affected by the therapeutic intervention of the protease's LDL-receptor lowering activity?" In this review, we appraise the different lipid lowering strategies targeting PCSK9 in light of the protein's different pleiotropic functions. Additionally, we delineate the key areas that require further examination, to ensure the long-term safety of the above lipid-lowering strategies.
Collapse
Affiliation(s)
- Yajnavalka Banerjee
- Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman.
| | - Raul D Santos
- Lipid Clinic Heart Institute (InCor), University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Khalid Al-Rasadi
- Department of Clinical Biochemistry, Sultan Qaboos University Hospital, Muscat, Oman
| | - Manfredi Rizzo
- Department of Internal Medicine and Medical Specialties, University of Palermo, Italy; Euro-Mediterranean Institute of Science and Technology, Italy
| |
Collapse
|
48
|
Fazio S. The role of PCSK9 in intestinal lipoprotein metabolism: synergism of statin and ezetimibe. ATHEROSCLEROSIS SUPP 2015; 17:23-6. [PMID: 25659873 DOI: 10.1016/s1567-5688(15)50006-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a major role in the regulation of lipoprotein metabolism, mostly through control of low-density lipoprotein receptor degradation. Depletion of cellular cholesterol causes a compensatory increase in plasma PCSK9 levels, which can diminish the cholesterol-lowering power of statins and may lead to the overproduction of intestinal lipoproteins, mainly thorough the up regulation of microsomal triglyceride transfer protein and the Niemann-Pick C1-like 1 protein, the target of ezetimibe. Thus, ezetimibe therapy may counter this unwanted effect of statins, providing an additional theoretical rationale for combining the effect of ezetimibe on intestinal cholesterol absorption and that of statins on cholesterol synthesis.
Collapse
Affiliation(s)
- Sergio Fazio
- Cornelius Vanderbilt Professor of Medicine Professor of Pathology, Immunology, and Microbiology Chief, Section of CVD Prevention Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| |
Collapse
|
49
|
Giunzioni I, Tavori H. New developments in atherosclerosis: clinical potential of PCSK9 inhibition. Vasc Health Risk Manag 2015; 11:493-501. [PMID: 26345307 PMCID: PMC4554462 DOI: 10.2147/vhrm.s74692] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pro-protein convertase subtilisin/kexin type 9 (PCSK9) is a secreted 692-amino acid protein that binds surface low-density lipoprotein (LDL) receptor (LDLR) and targets it toward lysosomal degradation. As a consequence, the number of LDLRs at the cell surface is decreased, and LDL-cholesterol (LDL-C) clearance is reduced, a phenomenon that is magnified by gain-of-function mutations of PCSK9. In contrast, loss-of-function mutations of PCSK9 result in increased surface LDLR and improved LDL-C clearance. This provides the rationale for targeting PCSK9 in hypercholesterolemic subjects as a means to lower LDL-C levels. Monoclonal antibodies (mAbs) against PCSK9 that block its interaction with the LDLR have been developed in the past decade. Two companies have recently received the approval for their anti-PCSK9 mAbs by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) Regeneron/Sanofi, with alirocumab (commercial name – PRALUENT®) and, Amgen with evolocumab (commercial name – Repatha™). The introduction of anti-PCSK9 mAbs will provide an alternative therapeutic strategy to address many of the unmet needs of current lipid-lowering therapies, such as inability to achieve goal LDL-C level, or intolerance and aversion to statins. This review will focus on the kinetics of PCSK9, pharmacokinetics and pharmacodynamics of anti-PCSK9 mAbs, and recent data linking PCSK9 and anti-PCSK9 mAbs to cardiovascular events. Moreover, it will highlight the unanswered questions that still need to be addressed in order to understand the physiologic function, kinetics, and dynamics of PCSK9.
Collapse
Affiliation(s)
- Ilaria Giunzioni
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Hagai Tavori
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
50
|
Abstract
The proof of concept that proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition affects cholesterol levels was first established after the demonstration that PCSK9 loss-of-function mutations result in a significant drop in circulating LDL cholesterol levels. Subsequent studies revealed that PCSK9 binds the epidermal growth factor precursor homology domain-A on the surface LDL Receptor (LDLR) and directs LDLR and PCSK9 for lysosomal degradation. Alirocumab (also known as SAR236553/REGN727) is a monoclonal antibody that binds circulating PCSK9 and blocks its interactions with surface LDLR. Alirocumab clinical trials with different doses on different administration schedules were shown to significantly reduce LDL cholesterol both as a mono-therapy and in combination with statins or ezetimibe. Although there is great potential for anti-PCSK9 therapies in the management of cholesterol metabolism, there is no clear evidence yet that blocking PCSK9 reduces cardiovascular disease outcome. This is being investigated in ongoing Phase III clinical trials with alirocumab.
Collapse
Affiliation(s)
- Hagai Tavori
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA.
| | | | | |
Collapse
|