2251
|
Affiliation(s)
- Jacqueline S Dron
- From the Department of Biochemistry (J.S.D., J.L., R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Robarts Research Institute (J.S.D., J.L., R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Julieta Lazarte
- From the Department of Biochemistry (J.S.D., J.L., R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Robarts Research Institute (J.S.D., J.L., R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Medicine (J.L., R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Robert A Hegele
- From the Department of Biochemistry (J.S.D., J.L., R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Robarts Research Institute (J.S.D., J.L., R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Medicine (J.L., R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
2252
|
Zhang X, Rimbert A, Balder W, Zwinderman AH, Kuivenhoven JA, Dallinga-Thie GM, Groen AK. Use of plasma metabolomics to analyze phenotype-genotype relationships in young hypercholesterolemic females. J Lipid Res 2018; 59:2174-2180. [PMID: 30266833 PMCID: PMC6210900 DOI: 10.1194/jlr.m088930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/08/2018] [Indexed: 11/20/2022] Open
Abstract
Hypercholesterolemia is characterized by high plasma LDL cholesterol and often caused by genetic mutations in LDL receptor (LDLR), APOB, or proprotein convertase subtilisin/kexin type 9 (PCSK9). However, a substantial proportion of hypercholesterolemic subjects do not have any mutations in these canonical genes, leaving the underlying pathobiology to be determined. In this study, we investigated to determine whether combining plasma metabolomics with genetic information increases insight in the biology of hypercholesterolemia. For this proof of concept study, we combined plasma metabolites from 119 hypercholesterolemic females with genetic information on the LDL canonical genes. Using hierarchical clustering, we identified four subtypes of hypercholesterolemia, which could be distinguished along two axes represented by triglyceride and large LDL particle concentration. Subjects with mutations in LDLR or APOB preferentially clustered together, suggesting that patients with defects in the LDLR pathway show a distinctive metabolomics profile. In conclusion, we show the potential of using metabolomics to segregate hypercholesterolemic subjects into different clusters, which may help in targeting genetic analysis.
Collapse
Affiliation(s)
- Xiang Zhang
- Departments of Experimental Vascular Medicine University of Amsterdam, Amsterdam, The Netherlands
| | - Antoine Rimbert
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, Groningen, The Netherlands
| | - Willem Balder
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, Groningen, The Netherlands
- Department of Cardiology, Jeroen Bosch Hospital, 's-Hertogenbosch, The Netherlands
| | - Aeilko Having Zwinderman
- Clinical Epidemiology, Biostatistics, and Bioinformatics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, Groningen, The Netherlands
| | | | - Albert Kornelis Groen
- Departments of Experimental Vascular Medicine University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
2253
|
Blackett P, George M, Wilson DP. Integrating lipid screening with ideal cardiovascular health assessment in pediatric settings. J Clin Lipidol 2018; 12:1346-1357. [DOI: 10.1016/j.jacl.2018.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/04/2018] [Accepted: 08/26/2018] [Indexed: 12/12/2022]
|
2254
|
Benito-Vicente A, Uribe KB, Jebari S, Galicia-Garcia U, Ostolaza H, Martin C. Familial Hypercholesterolemia: The Most Frequent Cholesterol Metabolism Disorder Caused Disease. Int J Mol Sci 2018; 19:ijms19113426. [PMID: 30388787 PMCID: PMC6275065 DOI: 10.3390/ijms19113426] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/21/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
Cholesterol is an essential component of cell barrier formation and signaling transduction involved in many essential physiologic processes. For this reason, cholesterol metabolism must be tightly controlled. Cell cholesterol is mainly acquired from two sources: Dietary cholesterol, which is absorbed in the intestine and, intracellularly synthesized cholesterol that is mainly synthesized in the liver. Once acquired, both are delivered to peripheral tissues in a lipoprotein dependent mechanism. Malfunctioning of cholesterol metabolism is caused by multiple hereditary diseases, including Familial Hypercholesterolemia, Sitosterolemia Type C and Niemann-Pick Type C1. Of these, familial hypercholesterolemia (FH) is a common inherited autosomal co-dominant disorder characterized by high plasma cholesterol levels. Its frequency is estimated to be 1:200 and, if untreated, increases the risk of premature cardiovascular disease. This review aims to summarize the current knowledge on cholesterol metabolism and the relation of FH to cholesterol homeostasis with special focus on the genetics, diagnosis and treatment.
Collapse
Affiliation(s)
- Asier Benito-Vicente
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Kepa B Uribe
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Shifa Jebari
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Unai Galicia-Garcia
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Helena Ostolaza
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Cesar Martin
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| |
Collapse
|
2255
|
Hyperuricemia and endothelial function: From molecular background to clinical perspectives. Atherosclerosis 2018; 278:226-231. [DOI: 10.1016/j.atherosclerosis.2018.10.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/30/2018] [Accepted: 10/05/2018] [Indexed: 12/28/2022]
|
2256
|
Stoekenbroek RM, Kallend D, Wijngaard PL, Kastelein JJ. Inclisiran for the treatment of cardiovascular disease: the ORION clinical development program. Future Cardiol 2018; 14:433-442. [PMID: 30375244 DOI: 10.2217/fca-2018-0067] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Inclisiran is a novel drug that inhibits PCSK9 synthesis specifically in the liver, harnessing the natural mechanism of RNAi. Phase I and II data show that inclisiran lowers low-density lipoprotein cholesterol levels on average by >50% with a duration of effect that enables twice-yearly dosing. Phases I, II and emerging Phase III data support inclisiran's safety, tolerability and risk-benefit profile. The ongoing ORION program includes Phase III trials that will provide robust evidence of inclisiran's safety and efficacy in individuals at high risk of atherosclerotic cardiovascular disease (ASCVD), including established ASCVD and familial hypercholesterolemia. In addition, the ORION-4 trial will assess the impact of inclisiran on cardiovascular outcomes in approximately 15,000 ASCVD subjects.
Collapse
Affiliation(s)
- Robert M Stoekenbroek
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,The Medicines Company, Parsippany, NJ 07054, USA
| | | | | | - John Jp Kastelein
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
2257
|
Behr PEB, Moriguchi EH, Castro I, Bodanese LC, Dutra OP, Leães PE, Pimentel Filho P. Indications of PCSK9 Inhibitors for Patients at High and Very High Cardiovascular Risk. Arq Bras Cardiol 2018; 111:104-108. [PMID: 30110052 PMCID: PMC6078356 DOI: 10.5935/abc.20180133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/25/2018] [Indexed: 01/14/2023] Open
Affiliation(s)
- Paulo Eduardo Ballvé Behr
- Hospital São Lucas da Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS - Brazil
| | - Emilio Hideyuki Moriguchi
- Faculdade de Medicina - Universidade Federal do Rio Grande do Sul, Porto Alegre, RS - Brazil.,Serviço de Cardiologia - Hospital de Clínicas de Porto Alegre, Porto Alegre - Brazil
| | - Iran Castro
- Instituto de Cardiologia / Fundação Universitária de Cardiologia, Porto Alegre, RS - Brazil
| | - Luiz Carlos Bodanese
- Hospital São Lucas da Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS - Brazil
| | - Oscar Pereira Dutra
- Instituto de Cardiologia / Fundação Universitária de Cardiologia, Porto Alegre, RS - Brazil
| | - Paulo Ernesto Leães
- Irmandade Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, RS - Brazil
| | | |
Collapse
|
2258
|
Association of ACE2 polymorphisms with susceptibility to essential hypertension and dyslipidemia in Xinjiang, China. Lipids Health Dis 2018; 17:241. [PMID: 30342552 PMCID: PMC6195726 DOI: 10.1186/s12944-018-0890-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/07/2018] [Indexed: 12/21/2022] Open
Abstract
Background Cardiovascular benefits by reversing environmental risks factors for essential hypertension (EH) and dyslipidemia could be weaken by high genetic risk. We investigated possible associations between ACE2 polymorphisms and dyslipidemia in patients with EH. Methods Four hundred and two hypertensive patients were enrolled in an EH group and 233 normotensive individuals were enrolled as control group from the Xinjiang region of China. Fourteen ACE2 polymorphisms were genotyped by Matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Results Participants carrying T allele (TT + CT) of rs2074192 (P = 0.006), rs4646155 (P = 0.030) and rs4646188 (P < 0.001), C allele (CT + CT or CC + CG) of rs4240157 (P = 0.012), rs4830542 (P = 0.020) and rs879922 (P < 0.001) and TT genotype of rs2106809 (P = 0.012) were associated with EH. Meanwhile,ACE2 SNPs also exhibited association with dyslipidemia but exhibited obvious heterogeneity. rs1978124 (TT + CT, P = 0.009), rs2106809 (TT, P = 0.045), rs233575 (CC + CT, P = 0.018), rs4646188 (CC, P = 0.011) and rs879922 (CC + CG, P = 0.003) were association with increased LDL-C (≥1.8 mmol/L). rs2106809 (CC + CT, P < 0.001), rs2285666(TT + CT, P = 0.017), rs4646142(CC + CG, P = 0.044), rs4646155(TT + CT, P < 0.001) and rs4646188(TT + CT, P = 0.033) were association with decreased HDL-C (< 1.0 mmol/L). rs2074192 (TT + CT, P = 0.012), rs4240157 (CC + CT, P = 0.027), rs4646156 (AA+AT, P = 0.007), rs4646188 (TT + CT, P = 0.005), rs4830542 (CC + CT, P = 0.047) and rs879922 (CC + CG, P = 0.001) were association with increased TC (≥5.2 mmol/L). rs2106809 (P = 0.034) and rs4646188 (P = 0.013) were associated with hypertriglyceridemia. Further, ischemic stroke was more prevalent with rs4240157 (CC + CT, P = 0.043), rs4646188 (CC + CT, P = 0.013) and rs4830542 (CC + CT, P = 0.037). In addition, rs2048683 and rs6632677 were not association with EH, dyslipidemia and ischemic stroke. Conclusion The ACE2 rs4646188 variant may be a potential and optimal genetic susceptibility marker for EH, dyslipidemia and its related ischemic stroke. Electronic supplementary material The online version of this article (10.1186/s12944-018-0890-6) contains supplementary material, which is available to authorized users.
Collapse
|
2259
|
Personalized Approach to Statin Selection in Primary Prevention: Genetic Risk Scores Vs Imaging Risk Scores. CURRENT CARDIOVASCULAR RISK REPORTS 2018. [DOI: 10.1007/s12170-018-0591-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
2260
|
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays an important role in regulation of LDL receptors on the hepatocyte surface and therefore is essential for effective removal of LDL particles from circulation. Genetic and biochemical studies have established that altered PCSK9 functionality influences both LDL cholesterol levels and cardiovascular risk. This has prompted development of inhibitory strategies targeting PCSK9. Study of monoclonal PCSK9 antibodies has progressed to the clinic, where they have been found to lower LDL cholesterol levels and reduce cardiovascular event rates in large, clinical outcome trials. The use of PCSK9 inhibitors in the setting of dyslipidaemia is reviewed.
Collapse
Affiliation(s)
- Stephen J Nicholls
- South Australian Health and Medical Research Institute and University of Adelaide Adelaide, SA, Australia
| |
Collapse
|
2261
|
Ravnskov U, de Lorgeril M, Diamond DM, Hama R, Hamazaki T, Hammarskjöld B, Hynes N, Kendrick M, Langsjoen PH, Mascitelli L, McCully KS, Okuyama H, Rosch PJ, Schersten T, Sultan S, Sundberg R. LDL-C does not cause cardiovascular disease: a comprehensive review of the current literature. Expert Rev Clin Pharmacol 2018; 11:959-970. [PMID: 30198808 DOI: 10.1080/17512433.2018.1519391] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION For half a century, a high level of total cholesterol (TC) or low-density lipoprotein cholesterol (LDL-C) has been considered to be the major cause of atherosclerosis and cardiovascular disease (CVD), and statin treatment has been widely promoted for cardiovascular prevention. However, there is an increasing understanding that the mechanisms are more complicated and that statin treatment, in particular when used as primary prevention, is of doubtful benefit. Areas covered: The authors of three large reviews recently published by statin advocates have attempted to validate the current dogma. This article delineates the serious errors in these three reviews as well as other obvious falsifications of the cholesterol hypothesis. Expert commentary: Our search for falsifications of the cholesterol hypothesis confirms that it is unable to satisfy any of the Bradford Hill criteria for causality and that the conclusions of the authors of the three reviews are based on misleading statistics, exclusion of unsuccessful trials and by ignoring numerous contradictory observations.
Collapse
Affiliation(s)
| | - Michel de Lorgeril
- b Laboratoire Coeur et Nutrition, TIMC-IMAG, School of Medicine , University of Grenoble-Alpes , Grenoble , France
| | - David M Diamond
- c Department of Molecular Pharmacology and Physiology, Center for Preclinical and Clinical Research on PTSD , University of South Florida , Tampa , FL , USA.,d Department of Psychology, Center for Preclinical and Clinical Research on PTSD , University of South Florida , Tampa , FL , USA
| | - Rokuro Hama
- e Japan Institute of Pharmacovigilance , Osaka , Japan
| | - Tomohito Hamazaki
- f Department of Internal Medicine, Toyama Jonan Onsen Daini Hospital , University of Toyama , Toyama , Japan
| | | | - Niamh Hynes
- h Western Vascular Institute, University Hospital Galway & Galway Clinic , National University of Ireland & Royal college of Surgeons of Ireland affiliated Hospital , Galway , Ireland
| | - Malcolm Kendrick
- i East Cheshire Trust, Macclesfield District General Hospital , Macclesfield , UK
| | | | - Luca Mascitelli
- k Medical Service , Comando Brigata Alpina "Julia"/Multinational Land Force , Udine , Italy
| | - Kilmer S McCully
- l Pathology and Laboratory Medicine Service, VA Boston Healthcare System West Roxbury , Harvard Medical School , Boston , MA , USA
| | - Harumi Okuyama
- m Faculty of Pharmaceutical Science , Nagoya City University , Mizuhoku, Nagoya , Japan
| | - Paul J Rosch
- n New York Medical College , The American Institute of Stress , New York , NY , USA
| | - Tore Schersten
- o Wallenberg Laboratory for Cardiovascluar and Metabolic Research, Sahlgren's Academy , University of Gothenburg , Gothenburg , Sweden.,p Department of Metabolism , Columbia University , New York , NY , USA
| | - Sherif Sultan
- h Western Vascular Institute, University Hospital Galway & Galway Clinic , National University of Ireland & Royal college of Surgeons of Ireland affiliated Hospital , Galway , Ireland
| | | |
Collapse
|
2262
|
Pavanello C, Parolini M, Alberti A, Carenini M, Maino P, Mombelli G, Pazzucconi F, Origgi G, Orsi F, Trivella MG, Calabresi L, De Maria R. Systematic Lab Knowledge Integration for Management of Lipid Excess in High-Risk Patients: Rationale and Design of the SKIM LEAN Project. Front Big Data 2018; 1:4. [PMID: 33693320 PMCID: PMC7931911 DOI: 10.3389/fdata.2018.00004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 09/10/2018] [Indexed: 11/29/2022] Open
Abstract
SKIM LEAN aims at exploiting Electronic Health Records (EHRs) to integrate knowledge derived from routine laboratory tests with background analysis of clinical databases, for the identification and early referral to specialist care, where appropriate, of patients with hypercholesterolemia, who may be inadequately controlled according to their cardiovascular (CV) risk level. SKIM LEAN addresses gaps in care that may occur through the lack of coordination between primary and specialist care, incomplete adherence to clinical guidelines, or poor patient's compliance to the physician's prescriptions because of comorbidities or drug side effects. Key project objectives include: (1) improved health professionals' competence and patient empowerment through a two-tiered educational website for general practitioners (GPs) and patients, and (2) implementation of a hospital-community shared care pathway to increase the proportion of patients at high/very-high CV risk (Familial Hypercholesterolemia, previous CV events) who achieve target LDL cholesterol (LDL-C) levels. Thanks to a close collaboration between clinical and information technology partners, SKIM LEAN will fully exploit the value of big data deriving from EHRs, and filter such knowledge using clinically-derived algorithms to risk-stratify patients. Alerts for GPs will be generated with interpreted test results. GPs will be able to refer patients with uncontrolled LDL-C within the shared pathway to the lipid or secondary prevention outpatient clinics of NIG hospital. Metrics to verify the project achievements include web-site visits, the number of alerts generated, numbers of patients referred by GPs, the proportion of secondary prevention patients who achieve LDL-C <100 mg/dl or a >50% decrease from baseline.
Collapse
Affiliation(s)
- Chiara Pavanello
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro E. Grossi Paoletti, Università degli Studi di Milan, Milan, Italy
| | - Marina Parolini
- Dipartimento Cardiotoracovascolare, Istituto di Fisiologia Clinica del CNR, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Antonia Alberti
- SSD Diagnosi e Cure Territoriali per Malattie Cardiache, Dipartimento Cardiotoracovascolare, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | | | - Giuliana Mombelli
- SSD Diagnosi e Cure Territoriali per Malattie Cardiache, Dipartimento Cardiotoracovascolare, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Franco Pazzucconi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro E. Grossi Paoletti, Università degli Studi di Milan, Milan, Italy
| | - Gianni Origgi
- Sistemi Informativi Aziendali, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Orsi
- Sistemi Informativi Aziendali, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Maria Giovanna Trivella
- Dipartimento Cardiotoracovascolare, Istituto di Fisiologia Clinica del CNR, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Laura Calabresi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro E. Grossi Paoletti, Università degli Studi di Milan, Milan, Italy
- *Correspondence: Laura Calabresi
| | - Renata De Maria
- Dipartimento Cardiotoracovascolare, Istituto di Fisiologia Clinica del CNR, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
2263
|
Jones PJH, Shamloo M, MacKay DS, Rideout TC, Myrie SB, Plat J, Roullet JB, Baer DJ, Calkins KL, Davis HR, Barton Duell P, Ginsberg H, Gylling H, Jenkins D, Lütjohann D, Moghadasian M, Moreau RA, Mymin D, Ostlund RE, Ras RT, Ochoa Reparaz J, Trautwein EA, Turley S, Vanmierlo T, Weingärtner O. Progress and perspectives in plant sterol and plant stanol research. Nutr Rev 2018; 76:725-746. [PMID: 30101294 PMCID: PMC6130982 DOI: 10.1093/nutrit/nuy032] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Current evidence indicates that foods with added plant sterols or stanols can lower serum levels of low-density lipoprotein cholesterol. This review summarizes the recent findings and deliberations of 31 experts in the field who participated in a scientific meeting in Winnipeg, Canada, on the health effects of plant sterols and stanols. Participants discussed issues including, but not limited to, the health benefits of plant sterols and stanols beyond cholesterol lowering, the role of plant sterols and stanols as adjuncts to diet and drugs, and the challenges involved in measuring plant sterols and stanols in biological samples. Variations in interindividual responses to plant sterols and stanols, as well as the personalization of lipid-lowering therapies, were addressed. Finally, the clinical aspects and treatment of sitosterolemia were reviewed. Although plant sterols and stanols continue to offer an efficacious and convenient dietary approach to cholesterol management, long-term clinical trials investigating the endpoints of cardiovascular disease are still lacking.
Collapse
Affiliation(s)
- Peter J H Jones
- Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Maryam Shamloo
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- George and Fay Yee Centre for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Dylan S MacKay
- George and Fay Yee Centre for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Todd C Rideout
- Department of Exercise and Nutrition Sciences, University of Buffalo, Buffalo, New York, USA
| | - Semone B Myrie
- Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jogchum Plat
- Department of Human Biology, Maastricht University, Maastricht, the Netherlands
| | - Jean-Baptiste Roullet
- Division of Metabolism, Child Development and Rehabilitation Center—Portland, Department of Pediatrics, Oregon Health & Science University, Portland, Oregon, USA
| | - David J Baer
- US Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Beltsville, Maryland, USA
| | - Kara L Calkins
- Department of Pediatrics, Division of Neonatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA; and the UCLA Mattel’s Children’s Hospital, Los Angeles, California, USA
| | | | - P Barton Duell
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Henry Ginsberg
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, USA
| | - Helena Gylling
- University of Helsinki and the Helsinki University Central Hospital, Helsinki, Finland
| | - David Jenkins
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada; and the Clinical Nutrition and Risk Factor Modification Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Mohammad Moghadasian
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robert A Moreau
- Eastern Regional Research Center, US Department of Agriculture, Agricultural Research Service, Wyndmoor, Pennsylvania, USA
| | - David Mymin
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Richard E Ostlund
- Division of Endocrinology, Metabolism and Lipid Research, Washington University, St Louis, USA
| | - Rouyanne T Ras
- Unilever Research & Development Vlaardingen, Vlaardingen, the Netherlands
| | | | - Elke A Trautwein
- Unilever Research & Development Vlaardingen, Vlaardingen, the Netherlands
| | | | - Tim Vanmierlo
- Department of Immunology and Biochemistry, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Oliver Weingärtner
- Klinik für Innere Medizin I, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany; Abteilung für Kardiologie, Klinikum Oldenburg, European Medical School Oldenburg-Groningen, Oldenburg, Germany
| |
Collapse
|
2264
|
Vallejo-Vaz AJ, Ray KK. Epidemiology of familial hypercholesterolaemia: Community and clinical. Atherosclerosis 2018; 277:289-297. [DOI: 10.1016/j.atherosclerosis.2018.06.855] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/06/2018] [Accepted: 06/14/2018] [Indexed: 01/10/2023]
|
2265
|
Affiliation(s)
- Sergio Fazio
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, Oregon, USA
| | - Michael D Shapiro
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
2266
|
Tirawanchai N, Supapornhemin S, Somkasetrin A, Suktitipat B, Ampawong S. Regulatory effect of Phikud Navakot extract on HMG-CoA reductase and LDL-R: potential and alternate agents for lowering blood cholesterol. Altern Ther Health Med 2018; 18:258. [PMID: 30249222 PMCID: PMC6154411 DOI: 10.1186/s12906-018-2327-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/17/2018] [Indexed: 11/15/2022]
Abstract
Background For decades, various cardiovascular symptoms have been relieved by the use of Ya-Hom Navakot, which is a formulation comprising 54 herbal medicines. The Thailand Ministry of Public Health listed Ya-Hom Navakot’s nine active principle and nomenclative herbal ingredients and termed them ‘Phikud Navakot’ (PN). Several reports have confirmed that PN has cardiovascular benefits similar to Ya-Hom Navakot. However, whether PN facilitates lipid-lowering activity remains unclear. Methods The present study investigated an in vitro model for examining the gene expression levels of 3-hydroxyl-3-methylglutaryl-CoA reductase (HMGCR) and low-density lipoprotein receptor (LDL-R) in HepG2 cells using qRT-PCR. The ethanol and water extractions of Ya-Hom Navakot, PN and Ya-Hom Navakot without PN were compared. Results One mg/ml of both NYEF and NYWF were found to significantly lower cholesterol by either the up-regulation of LDL-R or down-regulation of HMGCR compared with negative controls and 1 mg/ml simvastatin (p < 0.05). PNEF also up-regulated LDL-R gene expression, even more than NYEF (p < 0.05). In addition, the ethanol and water extracts of PN significantly down-regulated HMGCR gene expression compared with those of Ya-Hom Navakot without PN (p < 0.05). Conclusion The use of Ya-Hom Navakot or PN may provide an alternative treatment to lower cholesterol through HMGCR gene inhibition and LDL-R gene enhancement.
Collapse
|
2267
|
Hernández-Mijares A, Ascaso JF, Blasco M, Brea Á, Díaz Á, Mantilla T, Pedro-Botet J, Pintó X, Millán J. Residual cardiovascular risk of lipid origin. Components and pathophysiological aspects. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2018; 31:75-88. [PMID: 30262442 DOI: 10.1016/j.arteri.2018.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/10/2018] [Accepted: 06/19/2018] [Indexed: 11/24/2022]
Abstract
There is no doubt about the relationship between LDL-c and cardiovascular risk, as well as about the benefits of statin treatment. Once the objective of LDL-c has been achieved, the evidences that demonstrate the persistence of a high cardiovascular risk, a concept called residual risk, are notable. The residual risk of lipid origin is based on atherogenic dyslipidemia, characterized by an increase in triglycerides and triglyceride-rich lipoproteins, a decrease in HDL-c and qualitative alterations in LDL particles. The most commonly used measures to identify this dyslipidemia are based on the determination of total cholesterol, triglycerides, HDL, non-HDL cholesterol and remaining cholesterol, as well as apolipoprotein B100 and lipoprotein (a) in certain cases. The treatment of atherogenic dyslipidemia is based on weight loss and physical exercise. Regarding pharmacological treatment, we have no evidence of cardiovascular benefit with drugs aimed at lowering triglycerides and HDL-c, fenofibrate seems to be effective in situations of atherogenic dyslipidemia.
Collapse
Affiliation(s)
- Antonio Hernández-Mijares
- Fundación para la Investigación Sanitaria y Biomédica de la Comunidad Valenciana FISABIO, Servicio de Endocrinología y Nutrición, Hospital Universitario Dr. Peset Valencia; Departamento de Medicina, Universitat de València, Valencia, España.
| | - Juan F Ascaso
- Servicio de Endocrinología, Hospital Clínico Universitario; Departamento de Medicina, Universitat de València, Valencia, España
| | - Mariano Blasco
- Área Sanitaria de Delicias, Atención Primaria, Zaragoza, España
| | - Ángel Brea
- Servicio de Medicina Interna, Hospital San Pedro, Logroño, España
| | - Ángel Díaz
- Centro de Salud de Bembibre, Bembibre (León), España
| | - Teresa Mantilla
- Centro de Salud de Prosperidad, Atención Primaria, Madrid, España
| | - Juan Pedro-Botet
- Unidad de Lípidos y Riesgo Vascular, Servicio de Endocrinología y Nutrición, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, España
| | - Xavier Pintó
- Unidad de Lípidos, Servicio de Medicina Interna, Hospital Universitario de Bellvitge, Universitat de Barcelona, CIBERobn-ISCIII, Barcelona, España
| | - Jesús Millán
- Unidad de Lípidos, Servicio de Medicina Interna, Hospital General Universitario Gregorio Marañón, Universidad Complutense, Madrid, España.
| | | |
Collapse
|
2268
|
Priksz D, Bombicz M, Varga B, Kurucz A, Gesztelyi R, Balla J, Toth A, Papp Z, Szilvassy Z, Juhasz B. Upregulation of Myocardial and Vascular Phosphodiesterase 9A in A Model of Atherosclerotic Cardiovascular Disease. Int J Mol Sci 2018; 19:ijms19102882. [PMID: 30249014 PMCID: PMC6213954 DOI: 10.3390/ijms19102882] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/10/2018] [Accepted: 09/20/2018] [Indexed: 01/09/2023] Open
Abstract
Atherosclerosis is strongly associated with cardiac dysfunction and heart failure. Besides microvascular dysfunction and diminishment of the cardiac nitric oxide-Protein Kinase G (NO-PKG) pathway, recent evidence suggests that phosphodiesterase 9A (PDE9A) enzyme has an unfavorable role in pathological changes. Here, we characterized a rabbit model that shows cardiac dysfunction as a result of an atherogenic diet, and examined the myocardial PDE9A signaling. Rabbits were divided into Control (normal diet) and HC (atherogenic diet) groups. Cardiac function was evaluated by echocardiography. Vascular function was assessed, along with serum biomarkers. Histological stains were conducted, expression of selected proteins and cyclic guanosine monophosphate (cGMP) levels were determined. Signs of diastolic dysfunction were shown in HC animals, along with concentric hypertrophy and interstitial fibrosis. Endothelial function was diminished in HC rabbits, along with marked reduction in the aortic lumen, and increased left ventricle outflow tract (LVOT) pressures. A significant increase was shown in myocardial PDE9A levels in HC animals with unchanged vasodilator-stimulated phosphoprotein (VASP) phosphorylation and cGMP levels. Upregulation of PDE9A may be associated with early stage of cardiac dysfunction in atherosclerotic conditions. Since PDE9A is involved in cGMP degradation and in deactivation of the cardioprotective PKG signaling pathway, it may become an encouraging target for future investigations in atherosclerotic diseases.
Collapse
Affiliation(s)
- Daniel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Balazs Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Andrea Kurucz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Jozsef Balla
- Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Attila Toth
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Zoltan Papp
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Zoltan Szilvassy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Bela Juhasz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| |
Collapse
|
2269
|
Moreau F, Blanchard C, Perret C, Flet L, Douane F, Frampas E, Mirallie E, Croyal M, Aguesse A, Krempf M, Prieur X, Pichelin M, Cariou B, Le May C. In vivo evidence for transintestinal cholesterol efflux in patients with complete common bile duct obstruction. J Clin Lipidol 2018; 13:213-217.e1. [PMID: 30342919 DOI: 10.1016/j.jacl.2018.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/27/2018] [Accepted: 09/15/2018] [Indexed: 12/01/2022]
Abstract
BACKGROUND Beyond the hepatobiliary pathway, studies have demonstrated that direct transintestinal cholesterol efflux (TICE) of plasma-derived cholesterol may contribute to reverse cholesterol transport. The clinical evidence of TICE in human remains challenged because of the difficulty to discriminate the hepatobiliary and transintestinal routes in vivo. OBJECTIVE To provide the first proof of concept that TICE exists in vivo in humans by demonstrating that plasma labeled cholesterol can be excreted in the feces of patients with complete bile duct obstruction. METHODS Plasma, bile, and fecal cholesterol excretion was measured by mass spectrometry 24, 48, and 72 hours after intravenous injection of D7-cholesterol in two patients presenting cholangiocarcinomas with a total obstruction of their primary bile duct. RESULTS No trace of bile acids was detected in the feces of the two patients. Despite this, a significant amount of plasma D7-cholesterol was quantified in the feces of the two patients 48 hours and 72 hours after the intravenous injection. CONCLUSION Our data bring a direct proof that TICE is an active pathway in humans.
Collapse
Affiliation(s)
- François Moreau
- L'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Claire Blanchard
- L'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France; Service de Clinique de Chirurgie Digestive et Endocrinienne, CHU de Nantes, France
| | | | | | | | - Eric Frampas
- Department of Radiology, CHU Nantes, Nantes, France
| | - Eric Mirallie
- Service de Clinique de Chirurgie Digestive et Endocrinienne, CHU de Nantes, France
| | - Mikael Croyal
- INRA, UMR 1280, Physiologie des Adaptations Nutritionnelles, CHU Nantes, France; CRNHO, West Human Nutrition Research Center, CHU, Nantes, France
| | - Audrey Aguesse
- INRA, UMR 1280, Physiologie des Adaptations Nutritionnelles, CHU Nantes, France; CRNHO, West Human Nutrition Research Center, CHU, Nantes, France
| | - Michel Krempf
- INRA, UMR 1280, Physiologie des Adaptations Nutritionnelles, CHU Nantes, France; CRNHO, West Human Nutrition Research Center, CHU, Nantes, France
| | - Xavier Prieur
- L'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Matthieu Pichelin
- L'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France; L'institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Bertrand Cariou
- L'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France; L'institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Cédric Le May
- L'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France.
| |
Collapse
|
2270
|
Abstract
Mendelian randomization studies demonstrate that apolipoprotein B-containing lipoproteins have both causal and cumulative effects on the risk of atherosclerotic cardiovascular disease. The clinical benefit of lipid-lowering therapies depends on both the absolute reduction in circulating apolipoprotein B-containing lipoproteins and the total duration of exposure to these particles. Because atherosclerosis seems to be caused by the retention of apolipoprotein B-containing lipoproteins rather than by the cholesterol content carried by those lipoproteins, high-density lipoprotein-mediated efflux of cholesterol from the arterial wall may not reduce the risk of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Brian A Ference
- Institute for Advanced Studies, University of Bristol, 3rd Floor, Senate House, Bristol BS8 1UH, UK.
| |
Collapse
|
2271
|
Liu C, Li Y, Guan T, Lai Y, Shen Y, Zeyaweiding A, Zhao H, Li F, Maimaiti T. ACE2 polymorphisms associated with cardiovascular risk in Uygurs with type 2 diabetes mellitus. Cardiovasc Diabetol 2018; 17:127. [PMID: 30227878 PMCID: PMC6142339 DOI: 10.1186/s12933-018-0771-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 09/14/2018] [Indexed: 12/16/2022] Open
Abstract
Background Type 2 diabetes mellitus (T2D), rapidly increasing to epidemic proportions, globally escalates cardiovascular disease risk. Although intensive interventions and comprehensive management of environmental risks factors for T2D are associated with reduced cardiovascular disease, such approaches are limited for individuals with high genetic T2D risk. In this study we investigated possible associations of ACE2 polymorphisms and cardiovascular risks in Uygur patients with T2D. Methods 275 Uygur T2D patients and 272 non-diabetic Uygur individuals were enrolled as study participants. 14 ACE2 polymorphisms were genotyped by Matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Results ACE2 SNP rs1978124, rs2048683, rs2074192, rs233575, rs4240157, rs4646156, rs4646188 and rs879922 were associated with T2D (all P < 0.05). The 8 diabetic risk related ACE2 SNPs were further associated with diabetic related cardiovascular complications or events but exhibited heterogeneity as fellows: firstly, almost all diabetic risk related ACE2 SNPs (all P < 0.05) were associated with increased SBP except rs1978124 and rs2074192, while rs2074192, rs4646188 and rs879922 were associated elevated DBP (all P < 0.05). Secondly, SNP rs4646188 was not correlated with any type of dyslipidemia (TRIG, HDL-C, LDL-C or CHOL), and the other 7 diabetic risk related loci were at least correlated with one type of dyslipidemia (all P < 0.05). In particular, rs879922 were simultaneously correlated with four type of dyslipidemia (all P < 0.05). Thirdly, ACE2 SNP rs2074192 and rs879922 were associated with carotid arteriosclerosis stenosis (CAS) ≥ 50% (both P < 0.05). Fourthly, ACE2 SNP rs2074192, rs4240157, rs4646188 and 879922 were associated with increased MAU (all P < 0.05). In addition, ACE2 SNP rs2048683, rs4240157, rs4646156, rs4646188 and rs879922 were linked to heavier LVMI (all P < 0.05), but only rs4240157, rs4646156 and rs4646188 were associated with lower LVEF (all P < 0.05). Conclusion ACE2 SNP rs879922 may be a common genetic loci and optimal genetic susceptibility marker for T2D and T2D related cardiovascular risks in Uygurs. Electronic supplementary material The online version of this article (10.1186/s12933-018-0771-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cheng Liu
- Department of Cardiology, Guangzhou First People's Hospital, Medical School, South China University of Technology, #1 Panfu Road, Guangzhou, 510180, China. .,Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China. .,Department of Cardiology, Shufu People's Hospital, Kashgar Region, Xinjiang Uygur Autonomous Region (XUAR), 844100, China.
| | - Yanfang Li
- Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Tianwang Guan
- Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Yanxian Lai
- Department of Cardiology, Guangzhou First People's Hospital, Medical School, South China University of Technology, #1 Panfu Road, Guangzhou, 510180, China
| | - Yan Shen
- Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Abudurexiti Zeyaweiding
- Department of Cardiology, Shufu People's Hospital, Kashgar Region, Xinjiang Uygur Autonomous Region (XUAR), 844100, China
| | - Haiyan Zhao
- Department of Cardiology, Shufu People's Hospital, Kashgar Region, Xinjiang Uygur Autonomous Region (XUAR), 844100, China
| | - Fang Li
- Department of Cardiology, Shufu People's Hospital, Kashgar Region, Xinjiang Uygur Autonomous Region (XUAR), 844100, China
| | - Tutiguli Maimaiti
- Department of Cardiology, Shufu People's Hospital, Kashgar Region, Xinjiang Uygur Autonomous Region (XUAR), 844100, China
| |
Collapse
|
2272
|
Dyslipidemias in clinical practice. Clin Chim Acta 2018; 487:117-125. [PMID: 30201369 DOI: 10.1016/j.cca.2018.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 09/06/2018] [Accepted: 09/06/2018] [Indexed: 01/14/2023]
Abstract
Most dyslipidemic conditions have been linked to an increased risk of cardiovascular disease. Over the past few years major advances have been made regarding the genetic and metabolic basis of dyslipidemias. Detailed characterization of the genetic basis of familial lipid disorders and knowledge concerning the effects of environmental factors on the expression of dyslipidemias have increased substantially, contributing to a better diagnosis in individual patients. In addition to these developments, therapeutic options to lower cholesterol levels in clinical practice have expanded even further in patients with familial hypercholesterolemia and in subjects with cardiovascular disease. Finally, promising upcoming therapeutic lipid lowering strategies will be reviewed. All these advances will be discussed in relation to current clinical practice with special focus on common lipid disorders including familial dyslipidemias.
Collapse
|
2273
|
Affiliation(s)
- Robert DuBroff
- University of New Mexico School of Medicine, Department of Medicine, Division of Cardiology, Albuquerque.
| |
Collapse
|
2274
|
Farnier M. Nouvelles recommandations sur la prise en charge des dyslipidémies. Presse Med 2018; 47:769-774. [DOI: 10.1016/j.lpm.2018.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/14/2018] [Accepted: 09/05/2018] [Indexed: 10/28/2022] Open
|
2275
|
Korman MJ, Retterstøl K, Kristiansen IS, Wisløff T. Are PCSK9 Inhibitors Cost Effective? PHARMACOECONOMICS 2018; 36:1031-1041. [PMID: 29777433 DOI: 10.1007/s40273-018-0671-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The objective of this study was to review available health economic evaluations of PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors. These drugs reduce low-density lipid cholesterol levels and cardiovascular risk, but their cost effectiveness has been questioned. We searched Medline and Embase for economic evaluations in any language at any time. Studies were included if they analysed any PCSK9 inhibitor compared with either statin alone or in combination with ezetimibe or any other therapy considered standard prior to the introduction of PCSK9 inhibitors. We found ten full health economic evaluations of PCSK9 inhibitors, two from Europe and eight from the United States (US). Six of the eight from the US were from two different consortia that analysed PCSK9 inhibitors at different stages through the development of evidence. All studies generally reported incremental cost-effectiveness ratios above suggested thresholds for cost effectiveness, except one study from Spain. The results of this review indicate that PCSK9 inhibitors in general are not cost effective at the current prices, but lower prices may change the results.
Collapse
Affiliation(s)
- Max J Korman
- Ministry of Local Government and Modernisation, Oslo, Norway
| | - Kjetil Retterstøl
- Department of Nutrition, University of Oslo, Oslo, Norway
- Lipid Clinic, Oslo University Hospital, Oslo, Norway
| | | | - Torbjørn Wisløff
- Department of Health Management and Health Economics, University of Oslo, Oslo, Norway.
- Department of Infectious Disease Epidemiology and Modelling, Norwegian Institute of Public Health, Oslo, Norway.
| |
Collapse
|
2276
|
Impact of Lipids on Cardiovascular Health. J Am Coll Cardiol 2018; 72:1141-1156. [DOI: 10.1016/j.jacc.2018.06.046] [Citation(s) in RCA: 292] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 06/03/2018] [Accepted: 06/26/2018] [Indexed: 11/20/2022]
|
2277
|
Gaudet D, Langslet G, Gidding SS, Luirink IK, Ruzza A, Kurtz C, Lu C, Somaratne R, Raal FJ, Wiegman A. Efficacy, safety, and tolerability of evolocumab in pediatric patients with heterozygous familial hypercholesterolemia: Rationale and design of the HAUSER-RCT study. J Clin Lipidol 2018; 12:1199-1207. [DOI: 10.1016/j.jacl.2018.05.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/13/2018] [Accepted: 05/08/2018] [Indexed: 10/16/2022]
|
2278
|
Vallejo-Vaz AJ, Fayyad R, Boekholdt SM, Hovingh GK, Kastelein JJ, Melamed S, Barter P, Waters DD, Ray KK. Triglyceride-Rich Lipoprotein Cholesterol and Risk of Cardiovascular Events Among Patients Receiving Statin Therapy in the TNT Trial. Circulation 2018; 138:770-781. [DOI: 10.1161/circulationaha.117.032318] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background:
Mendelian randomization data suggest that the genetic determinants of lifetime higher triglyceride-rich lipoprotein-cholesterol (TRL-C) are causally related to cardiovascular disease and therefore a potential therapeutic target. The relevance of TRL-C among patients receiving statins is unknown. We assessed the relationship between TRL-C and cardiovascular risk, and whether this risk was modifiable among patients receiving statins in the TNT trial (Treating to New Targets).
Methods:
Patients with coronary heart disease and low-density lipoprotein cholesterol (LDL-C) 130 to 250 mg/dL entered an 8-week run-in phase with atorvastatin 10 mg/d (ATV10). After this period, participants with LDL-C <130 mg/dL entered the randomized phase with ATV10 (n=5006) versus atorvastatin 80 mg/d (ATV80, n=4995). The primary end point was coronary heart disease death, nonfatal myocardial infarction, resuscitated cardiac arrest, or stroke (major adverse cardiovascular events [MACE]). TRL-C was calculated as total cholesterol minus high-density lipoprotein cholesterol minus LDL-C. The effect of atorvastatin on TRL-C was assessed during the run-in phase (ATV10) and randomized phase (ATV80 versus ATV10). The risk of MACE was assessed across quintiles (Q) of baseline TRL-C (and, for comparison, by baseline triglycerides and non–high-density lipoprotein cholesterol) during the randomized period. Last, the association between TRL-C changes with atorvastatin and cardiovascular risk was assessed by multivariate Cox regression.
Results:
ATV10 reduced TRL-C 10.7% from an initial TRL-C of 33.9±16.6 mg/dL. ATV80 led to an additional 15.4% reduction. Cardiovascular risk factors positively correlated with TRL-C. Among patients receiving ATV10, higher TRL-C was associated with higher 5-year MACE rates (Q1=9.7%, Q5=13.8%; hazard ratio Q5-versus-Q1, 1.48; 95% confidence interval, 1.15–1.92;
P
-trend<0.0001). ATV80 (versus ATV10) did not significantly alter the risk of MACE in Q1-Q2, but significantly reduced risk in Q3-Q5 (relative risk reduction, 29%–41%; all
P
<0.0250), with evidence of effect modification (
P
-homogeneity=0.0053); results were consistent for triglycerides (
P
-homogeneity=0.0101) and directionally similar for non–high-density lipoprotein cholesterol (
P
-homogeneity=0.1387). Last, in adjusted analyses, a 1 SD percentage reduction in TRL-C with atorvastatin resulted in a significant lower risk of MACE (hazard ratio, 0.93; 95% confidence interval, 0.86–1.00;
P
=0.0482) independent of the reduction in LDL-C and of similar magnitude to that per 1 SD lowering in LDL-C (hazard ratio, 0.89; 95% confidence interval, 0.83–0.95;
P
=0.0008).
Conclusions:
The present post hoc analysis from TNT shows that increased TRL-C levels are associated with an increased cardiovascular risk and provides evidence for the cardiovascular benefit of lipid lowering with statins among patients who have coronary heart disease with high TRL-C.
Clinical Trial Registration:
URL:
https://www.clinicaltrials.gov
. Unique identifier: NCT00327691.
Collapse
Affiliation(s)
- Antonio J. Vallejo-Vaz
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, United Kingdom (A.J.V.-V., K.K.R.)
| | | | | | - G. Kees Hovingh
- Department of Vascular Medicine (G.K.H., J.J.K.), Academic Medical Center, Amsterdam, The Netherlands
| | - John J. Kastelein
- Department of Vascular Medicine (G.K.H., J.J.K.), Academic Medical Center, Amsterdam, The Netherlands
| | - Shari Melamed
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, United Kingdom (A.J.V.-V., K.K.R.)
| | - Philip Barter
- School of Medical Sciences, University of New South Wales Australia, Sydney (P.B.)
| | - David D. Waters
- Division of Cardiology, Zuckerberg San Francisco General Hospital, CA (D.D.W.)
| | - Kausik K. Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, United Kingdom (A.J.V.-V., K.K.R.)
| |
Collapse
|
2279
|
Langlois MR, Nordestgaard BG. Which Lipids Should Be Analyzed for Diagnostic Workup and Follow-up of Patients with Hyperlipidemias? Curr Cardiol Rep 2018; 20:88. [PMID: 30120626 DOI: 10.1007/s11886-018-1036-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW To summarize and discuss the clinical use of lipid and apolipoprotein tests in the settings of diagnosis and therapeutic follow-up of hyperlipidemia. RECENT FINDINGS The joint consensus panel of the European Atherosclerosis Society (EAS) and the European Federation of Clinical Chemistry and Laboratory Medicine (EFLM) recently produced recommendations on the measurement of atherogenic lipoproteins, taking into account the strengths and weaknesses of analytical and clinical performances of the tests. Total cholesterol, triglycerides, HDL cholesterol, LDL cholesterol, and calculated non-HDL cholesterol (= LDL + remnant cholesterol) constitute the primary lipid panel for hyperlipidemia diagnosis and cardiovascular risk estimation. LDL cholesterol is the primary target of lipid-lowering therapies. Non-HDL cholesterol or apolipoprotein B should be used as secondary therapeutic target in patients with mild-to-moderate hypertriglyceridemia, 2-10 mmol/l (175-880 mg/dl). Lipoprotein (a) is included in LDL cholesterol and should be measured at least once in all patients at cardiovascular risk, including to explain poor response to statin treatment.
Collapse
Affiliation(s)
- Michel R Langlois
- AZ St.-Jan Hospital, Department of Laboratory Medicine, Ruddershove 10, 8000, Brugge, Belgium. .,University of Ghent, Ghent, Belgium.
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2280
|
Rawshani A, Sattar N, Franzén S, Rawshani A, Hattersley AT, Svensson AM, Eliasson B, Gudbjörnsdottir S. Excess mortality and cardiovascular disease in young adults with type 1 diabetes in relation to age at onset: a nationwide, register-based cohort study. Lancet 2018; 392:477-486. [PMID: 30129464 PMCID: PMC6828554 DOI: 10.1016/s0140-6736(18)31506-x] [Citation(s) in RCA: 497] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/22/2018] [Accepted: 06/22/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND People with type 1 diabetes are at elevated risk of mortality and cardiovascular disease, yet current guidelines do not consider age of onset as an important risk stratifier. We aimed to examine how age at diagnosis of type 1 diabetes relates to excess mortality and cardiovascular risk. METHODS We did a nationwide, register-based cohort study of individuals with type 1 diabetes in the Swedish National Diabetes Register and matched controls from the general population. We included patients with at least one registration between Jan 1, 1998, and Dec 31, 2012. Using Cox regression, and with adjustment for diabetes duration, we estimated the excess risk of all-cause mortality, cardiovascular mortality, non-cardiovascular mortality, acute myocardial infarction, stroke, cardiovascular disease (a composite of acute myocardial infarction and stroke), coronary heart disease, heart failure, and atrial fibrillation. Individuals with type 1 diabetes were categorised into five groups, according to age at diagnosis: 0-10 years, 11-15 years, 16-20 years, 21-25 years, and 26-30 years. FINDINGS 27 195 individuals with type 1 diabetes and 135 178 matched controls were selected for this study. 959 individuals with type 1 diabetes and 1501 controls died during follow-up (median follow-up was 10 years). Patients who developed type 1 diabetes at 0-10 years of age had hazard ratios of 4·11 (95% CI 3·24-5·22) for all-cause mortality, 7·38 (3·65-14·94) for cardiovascular mortality, 3·96 (3·06-5·11) for non-cardiovascular mortality, 11·44 (7·95-16·44) for cardiovascular disease, 30·50 (19·98-46·57) for coronary heart disease, 30·95 (17·59-54·45) for acute myocardial infarction, 6·45 (4·04-10·31) for stroke, 12·90 (7·39-22·51) for heart failure, and 1·17 (0·62-2·20) for atrial fibrillation. Corresponding hazard ratios for individuals who developed type 1 diabetes aged 26-30 years were 2·83 (95% CI 2·38-3·37) for all-cause mortality, 3·64 (2·34-5·66) for cardiovascular mortality, 2·78 (2·29-3·38) for non-cardiovascular mortality, 3·85 (3·05-4·87) for cardiovascular disease, 6·08 (4·71-7·84) for coronary heart disease, 5·77 (4·08-8·16) for acute myocardial infarction, 3·22 (2·35-4·42) for stroke, 5·07 (3·55-7·22) for heart failure, and 1·18 (0·79-1·77) for atrial fibrillation; hence the excess risk differed by up to five times across the diagnosis age groups. The highest overall incidence rate, noted for all-cause mortality, was 1·9 (95% CI 1·71-2·11) per 100 000 person-years for people with type 1 diabetes. Development of type 1 diabetes before 10 years of age resulted in a loss of 17·7 life-years (95% CI 14·5-20·4) for women and 14·2 life-years (12·1-18·2) for men. INTERPRETATION Age at onset of type 1 diabetes is an important determinant of survival, as well as all cardiovascular outcomes, with highest excess risk in women. Greater focus on cardioprotection might be warranted in people with early-onset type 1 diabetes. FUNDING Swedish Heart and Lung Foundation.
Collapse
Affiliation(s)
- Araz Rawshani
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK.
| | - Stefan Franzén
- The Swedish National Diabetes Register, Västra Götalandsregionen, Gothenburg, Sweden
| | | | - Andrew T Hattersley
- Institute of Biomedical & Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Ann-Marie Svensson
- The Swedish National Diabetes Register, Västra Götalandsregionen, Gothenburg, Sweden
| | - Björn Eliasson
- Department of Internal Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Soffia Gudbjörnsdottir
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK; The Swedish National Diabetes Register, Västra Götalandsregionen, Gothenburg, Sweden
| |
Collapse
|
2281
|
Ference BA. Using Genetic Variants in the Targets of Lipid Lowering Therapies to Inform Drug Discovery and Development: Current and Future Treatment Options. Clin Pharmacol Ther 2018; 105:568-581. [PMID: 29953581 DOI: 10.1002/cpt.1163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 06/14/2018] [Indexed: 11/06/2022]
Abstract
Mendelian randomization studies and "human knock-out" studies of rare loss-of-function coding variants suggest that plasma levels of low-density lipoprotein-cholesterol LDL-C, triglycerides (TGs), and lipoprotein(a) (Lp(a)) are causally associated with the risk of cardiovascular disease, and, therefore, therapies directed against these targets should reduce the risk of cardiovascular events. However, several therapies directed against these targets have failed to reduce the risk of cardiovascular events in large-scale randomized trials, thus suggesting that causality is not sufficient evidence to establish genetic target validation. Instead, the critical question that needs to be answered to improve drug discovery and development is how much a causal biomarker needs to be changed to produce a clinically meaningful benefit in a short-term trial. This review describes how to use naturally randomized genetic evidence to accurately anticipate the results of randomized trials evaluating current and future lipid lowering therapies, inform the design of randomized trials, and transform the drug discovery and development process.
Collapse
Affiliation(s)
- Brian A Ference
- Centre for Naturally Randomized Trials, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK
| |
Collapse
|
2282
|
Vallejo-Vaz AJ. Guest Editorial: Reducing Risk in Familial Hypercholesterolaemia and Severe Dyslipidaemia: Novel Drugs Targeting PCSK9. Eur Cardiol 2018; 13:7-8. [PMID: 30310462 PMCID: PMC6159434 DOI: 10.15420/ecr.2018.13.1.ge3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Antonio J Vallejo-Vaz
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London London, UK
| |
Collapse
|
2283
|
Haghikia A, Landmesser U. Lipoproteins and Cardiovascular Redox Signaling: Role in Atherosclerosis and Coronary Disease. Antioxid Redox Signal 2018; 29:337-352. [PMID: 28817963 DOI: 10.1089/ars.2017.7052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
SIGNIFICANCE Lipoproteins, such as low-density lipoprotein, play a causal role in the development of atherosclerosis and coronary disease. Recent Advances: Lipoproteins can stimulate vascular production of reactive oxygen species, which act as important signaling molecules in the cardiovascular system contributing to the pathophysiology of endothelial dysfunction, hypertension, and atherosclerosis. CRITICAL ISSUES Modified lipoproteins have emerged as important regulators of redox signaling, such as oxidized or carbamylated low-density lipoprotein or modified high-density lipoproteins, that contain oxidized lipids, an altered protein cargo, and associated small molecules, such as symmetric dimethylarginine. FUTURE DIRECTIONS In this review, we provide an overview on signaling pathways stimulated by modified lipoproteins in the cardiovascular system and their potential role in cardiovascular disease development. Moreover, we highlight novel aspects of how gut microbiome-related mechanisms-a growing research field-may contribute to lipoprotein modification with subsequent impact on cardiovascular redox signaling. Antioxid. Redox Signal. 29, 337-352.
Collapse
Affiliation(s)
- Arash Haghikia
- 1 Department of Cardiology, Charité Universitätsmedizin Berlin , Berlin, Germany
- 2 German Center for Cardiovascular Research (DZHK) , partner site Berlin, Berlin, Germany
| | - Ulf Landmesser
- 1 Department of Cardiology, Charité Universitätsmedizin Berlin , Berlin, Germany
- 2 German Center for Cardiovascular Research (DZHK) , partner site Berlin, Berlin, Germany
- 3 Berlin Institute of Health (BIH) , Berlin, Germany
| |
Collapse
|
2284
|
Vilne B, Schunkert H. Integrating Genes Affecting Coronary Artery Disease in Functional Networks by Multi-OMICs Approach. Front Cardiovasc Med 2018; 5:89. [PMID: 30065929 PMCID: PMC6056735 DOI: 10.3389/fcvm.2018.00089] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 06/22/2018] [Indexed: 12/26/2022] Open
Abstract
Coronary artery disease (CAD) and myocardial infarction (MI) remain among the leading causes of mortality worldwide, urgently demanding a better understanding of disease etiology, and more efficient therapeutic strategies. Genetic predisposition as well as the environment and lifestyle are thought to contribute to disease risk. It is likely that non-linear and complex interactions occur between these multiple factors, involving simultaneous pathological changes in diverse cell types, tissues, and organs, at multiple molecular levels. Recent technological advances have exponentially expanded the breadth of available -omics data, from genome, epigenome, transcriptome, proteome, metabolome to even the microbiome. Integration of multiple layers of information across several -omics domains, i.e., the so-called multi-omics approach, currently holds the promise as a path toward precision medicine. Indeed, a more meaningful interpretation of genotype-phenotype relationships and the development of successful therapeutics tailored to individual patients are urgently needed. In this review, we will summarize recent findings and applications of integrative multi-omics in elucidating the etiology of CAD/MI; with a special focus on established disease susceptibility loci sequentially identified in genome-wide association studies (GWAS) over the last 10 years. Moreover, in addition to the autosomal genome, we will also consider the genetic variation in our “second genome”—the mitochondrial genome. Finally, we will summarize the current challenges in the field and point to future research directions required in order to successfully and effectively apply these approaches for precision medicine.
Collapse
Affiliation(s)
- Baiba Vilne
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| |
Collapse
|
2285
|
Mach F, Ray KK, Wiklund O, Corsini A, Catapano AL, Bruckert E, De Backer G, Hegele RA, Hovingh GK, Jacobson TA, Krauss RM, Laufs U, Leiter LA, März W, Nordestgaard BG, Raal FJ, Roden M, Santos RD, Stein EA, Stroes ES, Thompson PD, Tokgözoğlu L, Vladutiu GD, Gencer B, Stock JK, Ginsberg HN, Chapman MJ, European Atherosclerosis Society Consensus Panel. Adverse effects of statin therapy: perception vs. the evidence - focus on glucose homeostasis, cognitive, renal and hepatic function, haemorrhagic stroke and cataract. Eur Heart J 2018; 39:2526-2539. [PMID: 29718253 PMCID: PMC6047411 DOI: 10.1093/eurheartj/ehy182] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/09/2017] [Accepted: 03/22/2018] [Indexed: 12/17/2022] Open
Abstract
Aims To objectively appraise evidence for possible adverse effects of long-term statin therapy on glucose homeostasis, cognitive, renal and hepatic function, and risk for haemorrhagic stroke or cataract. Methods and results A literature search covering 2000-2017 was performed. The Panel critically appraised the data and agreed by consensus on the categorization of reported adverse effects. Randomized controlled trials (RCTs) and genetic studies show that statin therapy is associated with a modest increase in the risk of new-onset diabetes mellitus (about one per thousand patient-years), generally defined by laboratory findings (glycated haemoglobin ≥6.5); this risk is significantly higher in the metabolic syndrome or prediabetes. Statin treatment does not adversely affect cognitive function, even at very low levels of low-density lipoprotein cholesterol and is not associated with clinically significant deterioration of renal function, or development of cataract. Transient increases in liver enzymes occur in 0.5-2% of patients taking statins but are not clinically relevant; idiosyncratic liver injury due to statins is very rare and causality difficult to prove. The evidence base does not support an increased risk of haemorrhagic stroke in individuals without cerebrovascular disease; a small increase in risk was suggested by the Stroke Prevention by Aggressive Reduction of Cholesterol Levels study in subjects with prior stroke but has not been confirmed in the substantive evidence base of RCTs, cohort studies and case-control studies. Conclusion Long-term statin treatment is remarkably safe with a low risk of clinically relevant adverse effects as defined above; statin-associated muscle symptoms were discussed in a previous Consensus Statement. Importantly, the established cardiovascular benefits of statin therapy far outweigh the risk of adverse effects.
Collapse
Affiliation(s)
- François Mach
- Division of Cardiology, Department of Medical Specialties, Foundation for Medical Researches, Geneva University Hospital, Rue Gabrielle-Perret-Gentil 4 1205 Geneva, Switzerland
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
| | - Olov Wiklund
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, University of Milan and IRCCS Multimedica, Milan, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan and IRCCS Multimedica, Milan, Italy
| | - Eric Bruckert
- National Institute for Health and Medical Research (INSERM) UMRS1166, Department of Endocrinology-Metabolism, ICAN—Institute of CardioMetabolism and Nutrition, AP-HP, Hôpital de la Pitié, Paris, France
| | - Guy De Backer
- Department of Public Health, University Hospital Ghent, Ghent, Belgium
| | - Robert A Hegele
- Department of Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Ronald M Krauss
- Department of Atherosclerosis Research, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - Ulrich Laufs
- Department of Cardiology, University of Leipzig, Leipzig, Germany
| | - Lawrence A Leiter
- Li Ka Shing Knowledge Institute of St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry and The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Frederick J Raal
- Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Michael Roden
- German Center for Diabetes Research (DZD), München-Neuherberg, Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Raul D Santos
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Evan A Stein
- Metabolic and Atherosclerosis Research Center, Cincinnati, OH, USA
| | - Erik S Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Lale Tokgözoğlu
- Department of Cardiology, Hacettepe University, Ankara, Turkey
| | - Georgirene D Vladutiu
- Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, The State University of New York, New York, USA
| | - Baris Gencer
- Division of Cardiology, Department of Medical Specialties, Foundation for Medical Researches, Geneva University Hospital, Rue Gabrielle-Perret-Gentil 4 1205 Geneva, Switzerland
| | - Jane K Stock
- European Atherosclerosis Society, Gothenburg, Sweden
| | - Henry N Ginsberg
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, USA
| | - M John Chapman
- National Institute for Health and Medical Research (INSERM), and University of Pierre and Marie Curie—Paris 6, Pitié Salpêtrière, Paris, France
| | | |
Collapse
|
2286
|
Verbeek R, Hoogeveen RM, Langsted A, Stiekema LCA, Verweij SL, Hovingh GK, Wareham NJ, Khaw KT, Boekholdt SM, Nordestgaard BG, Stroes ESG. Cardiovascular disease risk associated with elevated lipoprotein(a) attenuates at low low-density lipoprotein cholesterol levels in a primary prevention setting. Eur Heart J 2018; 39:2589-2596. [PMID: 29931232 PMCID: PMC6287703 DOI: 10.1093/eurheartj/ehy334] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 05/30/2018] [Indexed: 01/10/2023] Open
Abstract
Aims Lipoprotein(a) (Lp(a)) elevation is a causal risk factor for cardiovascular disease (CVD). It has however been suggested that elevated Lp(a) causes CVD mainly in individuals with high low-density lipoprotein cholesterol (LDL-C) levels. We hypothesized that the risk associated with high Lp(a) levels would largely be attenuated at low LDL-C levels. Methods and results In 16 654 individuals from the EPIC-Norfolk prospective population study, and in 9448 individuals from the Copenhagen City Heart Study (CCHS) parallel statistical analyses were performed. Individuals were categorized according to their Lp(a) and LDL-C levels. Cut-offs were set at the 80th cohort percentile for Lp(a). Low-density lipoprotein cholesterol cut-offs were set at 2.5, 3.5, 4.5, and 5.5 mmol/L. Low-density lipoprotein cholesterol levels in the primary analyses were corrected for Lp(a)-derived LDL-C (LDL-Ccorr). Multivariable-adjusted hazard ratios were calculated for each category. The category with LDL-Ccorr <2.5 mmol/L and Lp(a) <80th cohort percentile was used as reference category. In the EPIC-Norfolk and CCHS cohorts, individuals with an Lp(a) ≥80th percentile were at increased CVD risk compared with those with Lp(a) <80th percentile for any LDL-Ccorr levels ≥2.5 mmol/L. In contrast, for LDL-Ccorr <2.5 mmol/L, the risk associated with elevated Lp(a) attenuated. However, there was no interaction between LDL-Ccorr and Lp(a) levels on CVD risk in either cohort. Conclusion Lipoprotein(a) and LDL-C are independently associated with CVD risk. At LDL-C levels below <2.5 mmol/L, the risk associated with elevated Lp(a) attenuates in a primary prevention setting.
Collapse
Affiliation(s)
- Rutger Verbeek
- Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Renate M Hoogeveen
- Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Anne Langsted
- Department of Clinical Biochemistry, Herlev Ringvej 75, 2730 Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark
| | - Lotte C A Stiekema
- Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Simone L Verweij
- Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Nicholas J Wareham
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0QQ, UK
| | | | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev Ringvej 75, 2730 Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark
| | - Erik S G Stroes
- Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
2287
|
Ofori-Asenso R, Zoungas S, Tonkin A, Liew D. LDL-Cholesterol Is the Only Clinically Relevant Biomarker for Atherosclerotic Cardiovascular Disease (ASCVD) Risk. Clin Pharmacol Ther 2018; 104:235-238. [PMID: 30004113 DOI: 10.1002/cpt.1125] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Richard Ofori-Asenso
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia.,Epidemiological Modelling Unit, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Sophia Zoungas
- Division of Metabolism, Genomics and Ageing, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Andrew Tonkin
- Cardiovascular Research Unit, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Danny Liew
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
2288
|
Bohula EA, Giugliano RP, Leiter LA, Verma S, Park JG, Sever PS, Lira Pineda A, Honarpour N, Wang H, Murphy SA, Keech A, Pedersen TR, Sabatine MS. Inflammatory and Cholesterol Risk in the FOURIER Trial. Circulation 2018. [DOI: 10.1161/circulationaha.118.034032] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Erin A. Bohula
- TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (E.A.B., R.P.G., J.-G.P., S.A.M., M.S.S.)
| | - Robert P. Giugliano
- TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (E.A.B., R.P.G., J.-G.P., S.A.M., M.S.S.)
| | - Lawrence A. Leiter
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, University of Toronto, Ontario, Canada (L.A.L., S.V.)
| | - Subodh Verma
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, University of Toronto, Ontario, Canada (L.A.L., S.V.)
| | - Jeong-Gun Park
- TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (E.A.B., R.P.G., J.-G.P., S.A.M., M.S.S.)
| | - Peter S. Sever
- International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London, United Kingdom (P.S.S.)
| | | | | | - Huei Wang
- Amgen, Thousand Oaks, CA (A.L.P., N.H., H.W.)
| | - Sabina A. Murphy
- TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (E.A.B., R.P.G., J.-G.P., S.A.M., M.S.S.)
| | - Anthony Keech
- Sydney Medical School, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Australia (A.K.)
| | - Terje R. Pedersen
- Oslo University Hospital, Ulleval and Medical Faculty, University of Oslo, Norway (T.R.P.)
| | - Marc S. Sabatine
- TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (E.A.B., R.P.G., J.-G.P., S.A.M., M.S.S.)
| |
Collapse
|
2289
|
Kereiakes DJ, Lepor NE, Gerber R, Veronica Lee L, Elassal J, Thompson D, Michael Gibson C. Efficacy and safety of alirocumab in patients with or without prior coronary revascularization: Pooled analysis of eight ODYSSEY phase 3 trials. Atherosclerosis 2018; 277:211-218. [PMID: 30025648 DOI: 10.1016/j.atherosclerosis.2018.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/29/2018] [Accepted: 07/06/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Patients with atherosclerotic cardiovascular disease (ASCVD) and prior revascularization are at high risk of further cardiovascular events and may require additional lipid-lowering therapies beyond maximally tolerated statin therapy. We assessed the efficacy and safety of alirocumab, a proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor, in patients with ASCVD, with or without prior coronary revascularization (percutaneous coronary intervention [PCI] or coronary artery bypass graft [CABG]). METHODS Data from eight controlled (placebo/ezetimibe) phase 3 ODYSSEY trials were pooled and stratified by trial design: alirocumab 150 mg or 75 mg with possible dose increase to 150 mg (75/150 mg) every 2 weeks (Q2W) versus placebo, and alirocumab 75/150 mg Q2W versus ezetimibe. Most patients received background maximally tolerated statin therapy. RESULTS Among 4629 randomized patients with hypercholesterolemia, 3382 had ASCVD including 2191 with prior revascularization. Although baseline characteristics were comparable between alirocumab and control groups, revascularized patients were more likely to be male, have had prior myocardial infarction/stroke, have higher lipoprotein (a) and PCSK9 levels, and were more often treated with high-intensity statin therapy. Alirocumab significantly reduced low-density lipoprotein cholesterol (LDL-C; primary endpoint; p < 0.0001), lipoprotein (a), non-high-density lipoprotein cholesterol, and apolipoprotein B levels from baseline to week 24 (vs. control), regardless of stratified treatment group or revascularization status. On-treatment LDL-C levels with alirocumab ranged from 45.6 to 64.8 mg/dL. Alirocumab had a similar safety profile regardless of revascularization status, and higher rates of injection-site reactions versus controls. CONCLUSIONS Alirocumab is generally well-tolerated and effective with a similar safety profile in high-risk patients with or without prior revascularization (PCI/CABG).
Collapse
Affiliation(s)
- Dean J Kereiakes
- The Christ Hospital, Heart and Vascular Center/The Lindner Research Center, Cincinnati, OH, USA.
| | - Norman E Lepor
- Cedars-Sinai Medical Center/Westside Medical Associates of Los Angeles, Beverly Hills, CA, USA
| | - Robert Gerber
- Department of Cardiology, East Sussex Healthcare NHS Trust, Hastings, UK
| | | | - Joe Elassal
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | |
Collapse
|
2290
|
Affiliation(s)
- Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig; Leipzig, Germany
| | | |
Collapse
|
2291
|
|
2292
|
Mortensen MB, Nordestgaard B. Comparison of Five Major Guidelines for Statin Use in Primary Prevention. Ann Intern Med 2018; 169:67-68. [PMID: 29971420 DOI: 10.7326/l18-0182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | - Børge Nordestgaard
- Herlev and Gentofte Hospital and Copenhagen University Hospital, Herlev, Denmark (M.B.M., B.N.)
| |
Collapse
|
2293
|
ApoB-100 Lipoprotein Complex Formation with Intima Proteoglycans as a Cause of Atherosclerosis and Its Possible Ex Vivo Evaluation as a Disease Biomarker. J Cardiovasc Dev Dis 2018; 5:jcdd5030036. [PMID: 29966388 PMCID: PMC6162553 DOI: 10.3390/jcdd5030036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 06/27/2018] [Accepted: 06/28/2018] [Indexed: 12/22/2022] Open
Abstract
Experimental and clinical data indicate that the initiation and progress of atherosclerosis and its clinical manifestations are first caused by circulating apoB-100 lipoproteins that enter and are retained in the arterial intima. Extracellular sulfated proteoglycans (PGs) of the intima are the retention agents. The PGs also initiate physical and biochemical lipoprotein degradation with the production of bioactive, lipid products that trigger an inflammatory response that leads to atherosclerosis. There are many simple methods for measuring abnormalities of circulating lipoproteins and their relation to atherosclerotic cardiovascular disease (ACVD). However, limited research aims to evaluate procedures that could report quantitatively about the contribution of the interaction of apoB-100 lipoprotein-arterial intima PGs to clinical manifestation of ACVD. In the present review we discuss observations indicating that simple ex vivo evaluation of the affinity of apoB-100 lipoproteins for arterial PGs and glycosaminoglycans (GAGs) can give an indication of its association with clinical manifestations of atherosclerosis. In addition, we discuss molecular and cellular aspects of the apoB-100 lipoproteins association with arterial PGs that are related to atherogenesis and that support the experimental framework behind the current “Response-to-Retention” hypothesis of atherosclerosis.
Collapse
|
2294
|
Griffin BA, Lovegrove JA. Butter Increases High-Density Lipoprotein Functional Capacity: Is This Compensation for Its Adverse Effect on Serum Low-Density Lipoprotein Cholesterol? J Nutr 2018; 148:1069-1070. [PMID: 29901721 DOI: 10.1093/jn/nxy086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/16/2018] [Indexed: 11/12/2022] Open
Affiliation(s)
- Bruce A Griffin
- Department of Nutritional Sciences, University of Surrey, Surrey, United Kingdom
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, United Kingdom
| |
Collapse
|
2295
|
Civeira F, Pocoví M. Familial hypercholesterolemia in pediatric patients. The success begins here. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2018; 30:179-180. [PMID: 29986810 DOI: 10.1016/j.arteri.2018.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Affiliation(s)
- Fernando Civeira
- Instituto De Investigación Sanitaria Aragón, CIBERCV, Universidad de Zaragoza, Zaragoza, España.
| | - Miguel Pocoví
- Instituto De Investigación Sanitaria Aragón, CIBERCV, Universidad de Zaragoza, Zaragoza, España
| |
Collapse
|
2296
|
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: 177] [Impact Index Per Article: 25.3] [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]
|
2297
|
Zafrir B, Jubran A. Lipid-lowering therapy with PCSK9-inhibitors in the real-world setting: Two-year experience of a regional lipid clinic. Cardiovasc Ther 2018; 36:e12439. [PMID: 29863817 DOI: 10.1111/1755-5922.12439] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/26/2018] [Accepted: 05/30/2018] [Indexed: 01/13/2023] Open
Abstract
AIM PCSK9 inhibitors (PCSK9i) effectively lower cholesterol levels in randomized trials with reduction in cardiovascular outcomes and favorable safety profile. However, the access to PCSK9i is limited due to high cost and data regarding the use of PCSK9i in real-world practice is limited. METHODS Data on all patients submitted for approval of PCSK9i at a regional lipid clinic, outside of clinical trials. Patients' profile, approval rates, low-density lipoprotein cholesterol (LDL-C) reduction rates, and adverse events were evaluated. RESULTS Recommendation for PCSK9i was given to 133 patients; 16 did not receive insurance approval and additional 16 were approved but did not initiate therapy. Of the 101 treated patients (47% females; mean age 61 ± 11 years), 52 had probable/definite familial hypercholesterolemia (FH) (peak LDL-C level 305 ± 87 mg/dL vs non-FH 204 ± 39 mg/dL) and 62% had an established cardiovascular disease. Statin intolerance was reported by 77%. Follow-up lipid panel was available in 66/101 patients: mean LDL-C reduction was 59% ± 19. Subjects with heterozygous FH had similar LDL-C decrease than those with non-FH (59% ± 22 vs 60% ± 14, P = .792). LDL-C < 100 mg/dL was achieved by 76%, LDL-C < 70 mg/dL by 58% and LDL-C < 40 mg/dL by 18% of those with follow-up data. Side effects were reported by 10%, mainly musculoskeletal complaints and flu-like symptoms, and 15% have discontinued treatment. CONCLUSIONS Patient selection by a regional lipid clinic resulted in a high real-world PCSK9i insurance approval, with efficacy and safety comparable to randomized clinical trials. Cost and medication nonadherence are potential barriers to successful implementation of therapy in routine clinical care.
Collapse
Affiliation(s)
- Barak Zafrir
- Cardiovascular Department, Lady Davis Carmel Medical Center, Haifa, Israel.,Clalit Health Services, Haifa, Israel
| | - Ayman Jubran
- Cardiovascular Department, Lady Davis Carmel Medical Center, Haifa, Israel.,Clalit Health Services, Haifa, Israel
| |
Collapse
|
2298
|
PCSK9 monoclonal antibody on a knife-edge: An article of faith in FH? J Clin Lipidol 2018; 12:844-848. [PMID: 29945779 DOI: 10.1016/j.jacl.2018.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/21/2018] [Accepted: 05/23/2018] [Indexed: 11/23/2022]
|
2299
|
Rhainds D, Brodeur MR, Tardif JC. Lipids, Apolipoproteins, and Inflammatory Biomarkers of Cardiovascular Risk: What Have We Learned? Clin Pharmacol Ther 2018; 104:244-256. [DOI: 10.1002/cpt.1114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/19/2018] [Accepted: 05/09/2018] [Indexed: 11/11/2022]
Affiliation(s)
| | | | - Jean-Claude Tardif
- Montreal Heart Institute; Montreal Quebec Canada
- Faculty of Medicine; Université de Montréal; Montreal Quebec Canada
| |
Collapse
|
2300
|
Zanoni P, Velagapudi S, Yalcinkaya M, Rohrer L, von Eckardstein A. Endocytosis of lipoproteins. Atherosclerosis 2018; 275:273-295. [PMID: 29980055 DOI: 10.1016/j.atherosclerosis.2018.06.881] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/04/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
During their metabolism, all lipoproteins undergo endocytosis, either to be degraded intracellularly, for example in hepatocytes or macrophages, or to be re-secreted, for example in the course of transcytosis by endothelial cells. Moreover, there are several examples of internalized lipoproteins sequestered intracellularly, possibly to exert intracellular functions, for example the cytolysis of trypanosoma. Endocytosis and the subsequent intracellular itinerary of lipoproteins hence are key areas for understanding the regulation of plasma lipid levels as well as the biological functions of lipoproteins. Indeed, the identification of the low-density lipoprotein (LDL)-receptor and the unraveling of its transcriptional regulation led to the elucidation of familial hypercholesterolemia as well as to the development of statins, the most successful therapeutics for lowering of cholesterol levels and risk of atherosclerotic cardiovascular diseases. Novel limiting factors of intracellular trafficking of LDL and the LDL receptor continue to be discovered and to provide drug targets such as PCSK9. Surprisingly, the receptors mediating endocytosis of high-density lipoproteins or lipoprotein(a) are still a matter of controversy or even new discovery. Finally, the receptors and mechanisms, which mediate the uptake of lipoproteins into non-degrading intracellular itineraries for re-secretion (transcytosis, retroendocytosis), storage, or execution of intracellular functions, are largely unknown.
Collapse
Affiliation(s)
- Paolo Zanoni
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Srividya Velagapudi
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Mustafa Yalcinkaya
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Lucia Rohrer
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|