1
|
Lian PA, Zhu WQ, Zhao WX, Huang PP, Ran JL, Tang YX, Huang XS, Li R. Lipoprotein(a) in atherosclerotic cardiovascular disease and proprotein convertase subtilisin/kexin-type 9 inhibitors. Clin Chim Acta 2024; 565:119982. [PMID: 39366516 DOI: 10.1016/j.cca.2024.119982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
High plasma lipoprotein(a) (Lp(a)) levels increase the cardiovascular risk in populations with atherosclerotic cardiovascular disease (ASCVD). Apolipoprotein (a) [apo(a)], a unique protein component of Lp(a), plays an important role in the pathogenesis of atherosclerosis. Statins, the primary medication in managing ASCVD, lower low-density lipoprotein cholesterol (LDL-C) but concurrently elevate plasma Lp(a) levels, contributing to an increased residual cardiovascular risk. In turn, proprotein convertase subtilisin/kexin-type 9 (PCSK9) inhibitors, a novel class of LDL-C lowering drugs, effectively reduce plasma Lp(a) levels, which is believed to decrease residual cardiovascular risk. However, the mechanism by which PCSK9 inhibitors reduce Lp(a) levels remains unknown. In addition, there are some clinical limitations of PCSK9 inhibitors. Here, we systematically review the past, present, and prospects of studies pertaining to Lp(a), PCSK9 inhibitors, and ASCVD.
Collapse
Affiliation(s)
- Ping-An Lian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen-Qiang Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei-Xin Zhao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Piao-Piao Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan-Li Ran
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya-Xin Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xian-Sheng Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cardiovascular Medicine, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China
| | - Rong Li
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Stomatology, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China.
| |
Collapse
|
2
|
Thau H, Neuber S, Emmert MY, Nazari-Shafti TZ. Targeting Lipoprotein(a): Can RNA Therapeutics Provide the Next Step in the Prevention of Cardiovascular Disease? Cardiol Ther 2024; 13:39-67. [PMID: 38381282 PMCID: PMC10899152 DOI: 10.1007/s40119-024-00353-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/12/2024] [Indexed: 02/22/2024] Open
Abstract
Numerous genetic and epidemiologic studies have demonstrated an association between elevated levels of lipoprotein(a) (Lp[a]) and cardiovascular disease. As a result, lowering Lp(a) levels is widely recognized as a promising strategy for reducing the risk of new-onset coronary heart disease, stroke, and heart failure. Lp(a) consists of a low-density lipoprotein-like particle with covalently linked apolipoprotein A (apo[a]) and apolipoprotein B-100, which explains its pro-thrombotic, pro-inflammatory, and pro-atherogenic properties. Lp(a) serum concentrations are genetically determined by the apo(a) isoform, with shorter isoforms having a higher rate of particle synthesis. To date, there are no approved pharmacological therapies that effectively reduce Lp(a) levels. Promising treatment approaches targeting apo(a) expression include RNA-based drugs such as pelacarsen, olpasiran, SLN360, and lepodisiran, which are currently in clinical trials. In this comprehensive review, we provide a detailed overview of RNA-based therapeutic approaches and discuss the recent advances and challenges of RNA therapeutics specifically designed to reduce Lp(a) levels and thus the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Henriette Thau
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Sebastian Neuber
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Maximilian Y Emmert
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353, Berlin, Germany.
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353, Berlin, Germany.
- Institute for Regenerative Medicine, University of Zurich, 8044, Zurich, Switzerland.
| | - Timo Z Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353, Berlin, Germany
- BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353, Berlin, Germany
| |
Collapse
|
3
|
Sulu C, Dedeoglu SE, Gonen B, Hepokur M, Guzel AN, Sahin S, Demir AN, Kara Z, Konukoglu D, Damci T, Gonen MS. Serum Lipoprotein(a) Is Not Associated with Graves' Ophthalmopathy. Metab Syndr Relat Disord 2024; 22:69-76. [PMID: 37883657 DOI: 10.1089/met.2023.0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023] Open
Abstract
Aim: To investigate the relationship of serum lipoprotein(a) [Lp(a)] and other serum lipids with presence of Graves' ophthalmopathy (GO). Methods: A total of 99 consecutive patients diagnosed with Graves' disease (GD), aged 18-65 years, who had not received prior treatment for GO, thyroid surgery, or radioactive iodine therapy, were recruited between June 2020 and July 2022. In addition, 56 healthy controls (HCs) were included as the control group. All patients underwent an ophthalmological examination, and were classified based on the presence of GO into the GO group (n = 45) and no GO group (n = 54). Fasting blood samples were collected from all participants to analyze serum lipid parameters, including Lp(a), total cholesterol, low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, and triglycerides. Results: The median serum levels of Lp(a) were 5.7 [4.3-9.2] in the GO group, 6.7 [3.7-9.9] in the no GO group, and 4.7 [3-7.6] in the HC group. The intergroup comparisons of serum Lp(a) levels showed no significant result. The serum levels of total cholesterol, LDL cholesterol, HDL cholesterol, and triglycerides were also similar between the groups (P > 0.05 for all). However, when analyzing only euthyroid GD patients and the control group, the serum LDL cholesterol levels were found to be significantly higher in the euthyroid GO group [median: 132 interquartile range (IQR) (110-148) mg/dL] than in the HCs [median: 96 IQR (94-118) mg/dL] (P = 0.002). Conclusion: The findings of our study did not support the association between serum Lp(a) levels and GO.
Collapse
Affiliation(s)
- Cem Sulu
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Selin Ece Dedeoglu
- Department of Internal Medicine, Eyupsultan State Hospital, Istanbul, Turkiye
| | - Busenur Gonen
- Department of Ophthalmology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Mustafa Hepokur
- Department of Ophthalmology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Adnan Nuri Guzel
- Department of Internal Medicine, and Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Serdar Sahin
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Ahmet Numan Demir
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Zehra Kara
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Dildar Konukoglu
- Department of Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Taner Damci
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Mustafa Sait Gonen
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| |
Collapse
|
4
|
Kumar R, Chhillar N, Gupta DS, Kaur G, Singhal S, Chauhan T. Cholesterol Homeostasis, Mechanisms of Molecular Pathways, and Cardiac Health: A Current Outlook. Curr Probl Cardiol 2024; 49:102081. [PMID: 37716543 DOI: 10.1016/j.cpcardiol.2023.102081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
The metabolism of lipoproteins, which regulate the transit of the lipid to and from tissues, is crucial to maintaining cholesterol homeostasis. Cardiac remodeling is referred to as a set of molecular, cellular, and interstitial changes that, following injury, affect the size, shape, function, mass, and geometry of the heart. Acetyl coenzyme A (acetyl CoA), which can be made from glucose, amino acids, or fatty acids, is the precursor for the synthesis of cholesterol. In this article, the authors explain concepts behind cardiac remodeling, its clinical ramifications, and the pathophysiological roles played by numerous various components, such as cell death, neurohormonal activation, oxidative stress, contractile proteins, energy metabolism, collagen, calcium transport, inflammation, and geometry. The levels of cholesterol are traditionally regulated by 2 biological mechanisms at the transcriptional stage. First, the SREBP transcription factor family regulates the transcription of crucial rate-limiting cholesterogenic and lipogenic proteins, which in turn limits cholesterol production. Immune cells become activated, differentiated, and divided, during an immune response with the objective of eradicating the danger signal. In addition to creating ATP, which is used as energy, this process relies on metabolic reprogramming of both catabolic and anabolic pathways to create metabolites that play a crucial role in regulating the response. Because of changes in signal transduction, malfunction of the sarcoplasmic reticulum and sarcolemma, impairment of calcium handling, increases in cardiac fibrosis, and progressive loss of cardiomyocytes, oxidative stress appears to be the primary mechanism that causes the transition from cardiac hypertrophy to heart failure. De novo cholesterol production, intestinal cholesterol absorption, and biliary cholesterol output are consequently crucial processes in cholesterol homeostasis. In the article's final section, the pharmacological management of cardiac remodeling is explored. The route of treatment is explained in different steps: including, promising, and potential strategies. This chapter offers a brief overview of the history of the study of cholesterol absorption as well as the different potential therapeutic targets.
Collapse
Affiliation(s)
| | - Neelam Chhillar
- Deparetment of Biochemistry, School of Medicine, DY Patil University, Navi Mumbai, India
| | - Dhruv Sanjay Gupta
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Ginpreet Kaur
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Shailey Singhal
- Cluster of Applied Sciences, University of Petroleum and Energy Studies, Dehradun, India
| | - Tanya Chauhan
- Division of Forensic Biology, National Forensic Sciences University, Delhi Campus (LNJN NICFS) Delhi, India
| |
Collapse
|
5
|
Hiraishi C, Matsui S, Kojima T, Sato R, Ando K, Fujimoto K, Yoshida H. Association of Renal Function and Statin Therapy with Lipoprotein(a) in Patients with Type 2 Diabetes. J Atheroscler Thromb 2024; 31:81-89. [PMID: 37558461 PMCID: PMC10776332 DOI: 10.5551/jat.64261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 06/11/2023] [Indexed: 08/11/2023] Open
Abstract
AIM A high level of serum lipoprotein(a) [Lp(a)] is associated with kidney disease development in patients with type 2 diabetes (T2DM). Recent studies have suggested that statins may affect serum levels of Lp(a). However, the statin effect is not well-defined in patients with T2DM with kidney dysfunction. This retrospective study aimed to investigate the relevance of kidney dysfunction and statin therapy to Lp(a) in patients with T2DM. METHODS Japanese patients with T2DM (n=149, 96 men and 53 women) were divided into two groups: statin users (n=79) and non-statin users (n=70). Multiple logistic regression analyses were performed with Lp(a) as the objective variable and estimated glomerular filtration rate (eGFR), hemoglobin A1c, age, gender, and body mass index as the explanatory variables. RESULTS Lp(a) serum levels were higher in statin users than in non-statin users (P=0.022). Multivariate regression analysis results showed an inverse correlation of eGFR to log Lp(a) in all patients (P=0.009) and in non-statin users (P=0.025), but not in statin users. In a multiple logistic regression analysis for median Lp(a), there was an inverse association between eGFR and Lp(a) level (odds ratio, 0.965; 95% confidence interval, 0.935-0.997; P=0.030) in non-statin users as well as in all participants, but not in statin users. CONCLUSIONS The present study suggests that a high Lp(a) level in patients with T2DM, except in statin users, is significantly associated with decreased eGFR, indicating that the increased Lp(a) levels under statin therapy might diminish the relationship between Lp(a) and eGFR.
Collapse
Affiliation(s)
- Chika Hiraishi
- Section of Internal Medicine of Metabolism and Nutrition, The Jikei University Graduate School of Medicine, Tokyo, Japan
- Department of General Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Chiba, Japan
| | - Sadako Matsui
- Food and Nutrition, Faculty of Human Science and Design, Japan Women’s University, Tokyo, Japan
| | - Takai Kojima
- Department of Laboratory Medicine, The Jikei University Kashiwa Hospital, Chiba, Japan
| | - Ryo Sato
- Department of Laboratory Medicine, The Jikei University Kashiwa Hospital, Chiba, Japan
| | - Kiyotaka Ando
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Tokyo, Japan
| | - Kei Fujimoto
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, The Jikei University Daisan Hospital, Tokyo, Japan
| | - Hiroshi Yoshida
- Section of Internal Medicine of Metabolism and Nutrition, The Jikei University Graduate School of Medicine, Tokyo, Japan
- Department of General Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Chiba, Japan
- Department of Laboratory Medicine, The Jikei University Kashiwa Hospital, Chiba, Japan
| |
Collapse
|
6
|
Chang YC, Hsu LA, Ko YL. Exploring PCSK9 Genetic Impact on Lipoprotein(a) via Dual Approaches: Association and Mendelian Randomization. Int J Mol Sci 2023; 24:14668. [PMID: 37834124 PMCID: PMC10572552 DOI: 10.3390/ijms241914668] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Previous investigations have suggested an association between the PCSK9 common polymorphism E670G and Lipoprotein(a) (Lp(a)) levels, as well as a link between plasma PCSK9 levels and Lp(a) concentrations. However, the causal relationship between plasma PCSK9 and Lp(a) levels remains uncertain. In this study, we explored the association between PCSK9 E670G polymorphism and Lp(a) levels in 614 healthy Taiwanese individuals. Employing a two-sample Mendelian randomization (MR) analysis using openly accessible PCSK9 and Lp(a) summary statistics from the genome-wide association studies (GWAS) and UK Biobank, we aimed to determine if a causal link exists between plasma PCSK9 levels and Lp(a) concentrations. Our findings reveal that the E670G G allele is independently associated with a decreased likelihood of developing elevated Lp(a) levels. This association persists even after adjusting for common cardiovascular risk factors and irrespective of lipid profile variations. The MR analysis, utilizing six PCSK9 GWAS-associated variants as instrumental variables to predict plasma PCSK9 levels, provides compelling evidence of a causal relationship between plasma PCSK9 levels and Lp(a) concentration. In conclusion, our study not only replicates the association between the PCSK9 E670G polymorphism and Lp(a) levels but also confirms a causative relationship between PCSK9 levels and Lp(a) concentrations through MR analysis.
Collapse
Affiliation(s)
- Ya-Ching Chang
- Department of Dermatology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Tao-Yuan 33305, Taiwan;
| | - Lung-An Hsu
- Cardiovascular Division, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Tao-Yuan 33305, Taiwan
| | - Yu-Lin Ko
- Department of Research, Division of Cardiology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 23142, Taiwan;
| |
Collapse
|
7
|
Feng T, Li Y, Xue X, Yang W, Li Q, Huang Y, Zhu T, Wang J, Xu L, Li X, Gao J, Sun S, Zhu B, Zhang S, Cao B, Xuan J, Zhao Z, Qian B. Association of statin use and increase in lipoprotein(a): a real-world database research. Eur J Med Res 2023; 28:212. [PMID: 37393291 DOI: 10.1186/s40001-023-01155-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 06/03/2023] [Indexed: 07/03/2023] Open
Abstract
BACKGROUND There is an increased concern that statins may have an unintended effect of elevated lipoprotein(a) [Lp(a)]. We conducted a large sample real-world study to test the association. METHODS This retrospective cohort study was conducted using data from an integrated SuValue database, which includes 221 hospitals across China covering more than 200,000 of population with longitudinal follow-up to 10 years. Propensity score matching was applied to identify two comparable cohorts with statin users and non-statin users. Detailed follow-up information such as Lp(a) levels were extracted. The hazard ratio was calculated on Lp(a) changes based on the statin usage cohorts. Detailed subgroup and different characteristic cohorts' analyses were also conducted. RESULTS After baseline propensity score matching, a total of 42,166 patients were included in a 1:1 matched ratio between statin users and non-statin users. In the case of no difference in low density lipoprotein (LDL-C), Lp(a) was increased significantly with the use of statins (adjusted HR 1.47; 95% confidence interval [CI] 1.43-1.50). Lp(a) increase was observed in various subgroup analyses and different cohorts. The dose intensity of statin was positively associated with the evaluated Lp(a) level. CONCLUSION The use of statins was associated with an increased risk of Lp(a) elevation compared with non-statin use counterparts. The clinical relevance of these increases needs to be addressed in surrogate marker trials and/or large, cardiovascular outcomes trials.
Collapse
Affiliation(s)
- Tienan Feng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Yao Li
- Clinical Center for Intelligent Rehabilitation Research, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | | | - Wei Yang
- SuValue Health Ltd, Shanghai, China
| | - Qiang Li
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yushi Huang
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Tengteng Zhu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jue Wang
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Limin Xu
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xianchen Li
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Gao
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Bin Zhu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - ShuYu Zhang
- Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Beibei Cao
- Department of Printing Equipment Engineering, Shanghai Publishing and Printing College, Shanghai, China
| | - Jianwei Xuan
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| | - Zhigang Zhao
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Biyun Qian
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
Zhang G, Ye X, Wang X, Lin Y, Zhu C, Pan J, Yin X, Ye M, Lv W, Tang W, Liu J, Yang X, Hui L, Zheng K. Serum total cholesterol levels associated with immediate memory performance in patients with chronic schizophrenia. Schizophr Res 2023; 255:256-260. [PMID: 37060796 DOI: 10.1016/j.schres.2023.03.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/13/2023] [Accepted: 03/26/2023] [Indexed: 04/17/2023]
Abstract
Cognitive impairments are common in patients with schizophrenia. Changes in total cholesterol (TC) may be involved in the development of schizophrenia and associated with cognitive function. This study aimed to investigate differences in serum TC level and cognitive function between schizophrenia patients and healthy controls and explore the relationship between serum TC level and cognitive function in patients with schizophrenia. A total of 105 schizophrenia patients and 105 healthy controls were recruited. Results showed that patients with schizophrenia had significantly lower scores on the overall RBANS scale and subscales (i.e., immediate memory, language, attention, and delayed memory) than those of healthy controls. Pearson's correlation analyses showed that in patients with schizophrenia, serum TC levels were positively associated with RBANS subscale scores of immediate memory and language. Furthermore, multivariate regression analyses showed that serum TC level was positively associated with the immediate memory index in patients with schizophrenia. However, no significant association was found between serum TC level and RBANS score in the healthy control group. Our results suggest that elevated serum TC level may be related to improved cognitive function in patients with schizophrenia, especially that of immediate memory.
Collapse
Affiliation(s)
- Guohua Zhang
- School of Mental Health, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou 325035, PR China.
| | - Xiaodan Ye
- The affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Xin Wang
- Center for Health Behaviours Research, JC School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Yixuan Lin
- School of Psychology and Cognitive Science, East China Normal University, Shanghai, PR China
| | - Cheng Zhu
- The affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Jianshe Pan
- The affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Xiaoli Yin
- The affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Minjie Ye
- The affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Wei Lv
- The affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Wei Tang
- The affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Jiahong Liu
- The affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Xue Yang
- Center for Health Behaviours Research, JC School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Li Hui
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou 215137, PR China.
| | - Ke Zheng
- The affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou 325000, PR China.
| |
Collapse
|
9
|
Zhang S, Kou X, Zhao H, Mak KK, Balijepalli MK, Pichika MR. Zingiber officinale var. rubrum: Red Ginger's Medicinal Uses. Molecules 2022; 27:775. [PMID: 35164040 PMCID: PMC8840670 DOI: 10.3390/molecules27030775] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 11/17/2022] Open
Abstract
Zingiber officinale var. rubrum (red ginger) is widely used in traditional medicine in Asia. Unlike other gingers, it is not used as a spice in cuisines. To date, a total of 169 chemical constituents have been reported from red ginger. The constituents include vanilloids, monoterpenes, sesquiterpenes, diterpenes, flavonoids, amino acids, etc. Red ginger has many therapeutic roles in various diseases, including inflammatory diseases, vomiting, rubella, atherosclerosis, tuberculosis, growth disorders, and cancer. Scientific evidence suggests that red ginger exhibits immunomodulatory, antihypertensive, antihyperlipidemic, antihyperuricemic, antimicrobial, and cytotoxic activities. These biological activities are the underlying causes of red ginger's therapeutic benefits. In addition, there have been few reports on adverse side effects of red ginger. This review aims to provide insights in terms the bioactive constituents and their biosynthesis, biological activities, molecular mechanisms, pharmacokinetics, and qualitative and quantitative analysis of red ginger.
Collapse
Affiliation(s)
- Shiming Zhang
- School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia; (S.Z.); (K.-K.M.)
| | - Xuefang Kou
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Hui Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Kit-Kay Mak
- School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia; (S.Z.); (K.-K.M.)
- Pharmaceutical Chemistry Department, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development & Innovation (IRDI), International Medical University, Kuala Lumpur 57000, Malaysia
| | - Madhu Katyayani Balijepalli
- Department of Pharmacology, Faculty of Medicine and Health Sciences, MAHSA University, Selangor 42610, Malaysia;
| | - Mallikarjuna Rao Pichika
- Pharmaceutical Chemistry Department, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development & Innovation (IRDI), International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
10
|
Abstract
Lipoprotein(a) [Lp(a)] is an atherogenic lipoprotein with a strong genetic regulation. Up to 90% of the concentrations are explained by a single gene, the LPA gene. The concentrations show a several-hundred-fold interindividual variability ranging from less than 0.1 mg/dL to more than 300 mg/dL. Lp(a) plasma concentrations above 30 mg/dL and even more above 50 mg/dL are associated with an increased risk for cardiovascular disease including myocardial infarction, stroke, aortic valve stenosis, heart failure, peripheral arterial disease, and all-cause mortality. Since concentrations above 50 mg/dL are observed in roughly 20% of the Caucasian population and in an even higher frequency in African-American and Asian-Indian ethnicities, it can be assumed that Lp(a) is one of the most important genetically determined risk factors for cardiovascular disease.Carriers of genetic variants that are associated with high Lp(a) concentrations have a markedly increased risk for cardiovascular events. Studies that used these genetic variants as a genetic instrument to support a causal role for Lp(a) as a cardiovascular risk factor are called Mendelian randomization studies. The principle of this type of studies has been introduced and tested for the first time ever with Lp(a) and its genetic determinants.There are currently no approved pharmacologic therapies that specifically target Lp(a) concentrations. However, some therapies that target primarily LDL cholesterol have also an influence on Lp(a) concentrations. These are mainly PCSK9 inhibitors that lower LDL cholesterol by 60% and Lp(a) by 25-30%. Furthermore, lipoprotein apheresis lowers both, Lp(a) and LDL cholesterol, by about 60-70%. Some sophisticated study designs and statistical analyses provided support that lowering Lp(a) by these therapies also lowers cardiovascular events on top of the effect caused by lowering LDL cholesterol, although this was not the main target of the therapy. Currently, new therapies targeting RNA such as antisense oligonucleotides (ASO) or small interfering RNA (siRNA) against apolipoprotein(a), the main protein of the Lp(a) particle, are under examination and lower Lp(a) concentrations up to 90%. Since these therapies specifically lower Lp(a) concentrations without influencing other lipoproteins, they will serve the last piece of the puzzle whether a decrease of Lp(a) results also in a decrease of cardiovascular events.
Collapse
|
11
|
Abstract
The study of lipoprotein(a) [Lp(a)] over the years has been a source of both enlightenment and frustration for the medical community. Accumulating evidence from large sample observational studies, Mendelian randomization studies, and genome-wide association studies has strengthened the association between Lp(a) and the development of atherosclerotic cardiovascular disease. This evidence supports the testing of Lp(a) in certain high-risk populations in order for clinicians to improve the risk profile of patients. Despite a variety of medical therapies that have been proven to reduce Lp(a) levels, the connection between the medical management of serum Lp(a) and improved cardiovascular outcomes remains elusive, due to the lack of specificity that current therapies have in targeting the Lp(a) production pathway. A new frontier in Lp(a) research has emerged with antisense-oligonucleotide therapy and RNA interference therapy, both of which target Lp(a) production at the level of mRNA translation. These therapies provide a pathway for investigating the effect of medical management of serum Lp(a) on cardiovascular outcomes.
Collapse
|
12
|
The Role of Structure and Biophysical Properties in the Pleiotropic Effects of Statins. Int J Mol Sci 2020; 21:ijms21228745. [PMID: 33228116 PMCID: PMC7699354 DOI: 10.3390/ijms21228745] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Statins are a class of drugs used to lower low-density lipoprotein cholesterol and are amongst the most prescribed medications worldwide. Most statins work as a competitive inhibitor of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGR), but statin intolerance from pleiotropic effects have been proposed to arise from non-specific binding due to poor enzyme-ligand sensitivity. Yet, research into the physicochemical properties of statins, and their interactions with off-target sites, has not progressed much over the past few decades. Here, we present a concise perspective on the role of statins in lowering serum cholesterol levels, and how their reported interactions with phospholipid membranes offer a crucial insight into the mechanism of some of the more commonly observed pleiotropic effects of statin administration. Lipophilicity, which governs hepatoselectivity, is directly related to the molecular structure of statins, which dictates interaction with and transport through membranes. The structure of statins is therefore a clinically important consideration in the treatment of hypercholesterolaemia. This review integrates the recent biophysical studies of statins with the literature on the physiological effects and provides new insights into the mechanistic cause of statin pleiotropy, and prospective means of understanding the cholesterol-independent effects of statins.
Collapse
|
13
|
Increased cardiovascular risk associated with hyperlipoproteinemia (a) and the challenges of current and future therapeutic possibilities. Anatol J Cardiol 2020; 23:60-69. [PMID: 32011323 PMCID: PMC7040869 DOI: 10.14744/anatoljcardiol.2019.56068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Population, genetic, and clinical studies demonstrated a causative and continuous, from other plasma lipoproteins independent relationship between elevated plasma lipoprotein (a) [Lp(a)] concentration and the development of cardiovascular disease (CVD), mainly those related to athe-rosclerotic CVD, and calcific aortic stenosis. Currently, a strong international consensus is still lacking regarding the single value which would be commonly used to define hyperlipoproteinemia (a). Its prevalence in the general population is estimated to be in the range of 10%–35% in accordance with the most commonly used threshold levels (>30 or >50 mg/dL). Since elevated Lp(a) can be of special importance in patients with some genetic disorders, as well as in individuals with otherwise controlled major risk factors, the identification and establishment of the proper therapeutic interventions that would lower Lp(a) levels and lead to CVD risk reduction could be very important. The majority of the classical lipid-lowering agents (statins, ezetimibe, and fibrates), as well as nutraceuticals (CoQ10 and garlic), appear to have no significant effect on its plasma levels, whereas for the drugs with the demonstrated Lp(a)-lowering effects (aspirin, niacin, and estrogens), their clinical efficacy in reducing cardiovascular (CV) events has not been unequivocally proven yet. Both Lp(a) apheresis and proprotein convertase subtilisin/kexin type 9 inhibitors can reduce the plasma Lp(a) by approximately 20%–30% on average, in parallel with much larger reduction of low-density lipoprotein cholesterol (up to 70%), what puts us in a difficulty to conclude about the true contribution of lowered Lp(a) to the reduction of CV events. The most recent advancement in the field is the introduction of the novel apolipoprotein (a) [apo(a)] antisense oligonucleotide therapy targeting apo(a), which has already proven itself as being very effective in decreasing plasma Lp(a) (by even >90%), but should be further tested in clinical trials. The aim of this review was to present some of the most important accessible scientific data, as well as dilemmas related to the currently and potentially in the near future more widely available therapeutic options for the management of hyperlipoproteinemia (a).
Collapse
|
14
|
Wang X, Li J, Ju J, Fan Y, Xu H. Effect of different types and dosages of statins on plasma lipoprotein(a) levels: A network meta-analysis. Pharmacol Res 2020; 163:105275. [PMID: 33166736 DOI: 10.1016/j.phrs.2020.105275] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/06/2020] [Accepted: 10/22/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIM Studies differ with respect to the effects of statins and their on lipoprotein(a)[Lp(a)] levels. The aim of the present study was to resolve these differences by determining the effect of various types and dosages of statins on Lp(a) levels. METHODS We searched PubMed, Embase and the Cochrane library for randomized controlled trials (RCTs) investigating the efficacy of statins on plasma Lp(a) levels. Study selection, data extraction and risk of bias assessment were conducted independently by four authors. We conducted pairwise meta-analysis and network meta-analysis (NMA). Consistency models were applied to NMA and the ranking probabilities for each treatment's efficacy were calculated. Node-splitting analysis was used to test inconsistency. This study was registered with PROSPERO, number CRD42020167612. RESULTS Twenty RCTs with 23,605 participants were included, involving 11 interventions. Most of the included studies presented some risks of bias, especially risks of performance and detection bias. In the pairwise meta-analysis, pooled results showed a small but statistically significant difference between high-intensity rosuvastatin and placebo on Lp(a) levels (MD = 1.81, 95 % CI [0.43, 3.19], P = 0.01). In the NMA, different types and dosages of statins showed no significant effect on the level of Lp(a), and there was no obvious difference between them. Subgroup analysis based on different populations and treatment durations did not provide any statistically significant findings about different statins on Lp(a) levels. Node-splitting analysis showed that no significant inconsistency existed (P > 0.05). CONCLUSIONS Statins have no clinically significant effect on Lp(a) levels, and there is no significant difference in the effect on Lp(a) levels between different types and dosages of statins. Moderate-intensity pitavastatin tended to have the best effect on reducing Lp(a) levels; nevertheless, it was insignificant. Our findings highlight the necessity for further study of the effect of statins on Lp(a) levels in future studies.
Collapse
Affiliation(s)
- Xinyi Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Jingen Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
| | - Jianqing Ju
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yixuan Fan
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Hao Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
15
|
Koushki K, Shahbaz SK, Mashayekhi K, Sadeghi M, Zayeri ZD, Taba MY, Banach M, Al-Rasadi K, Johnston TP, Sahebkar A. Anti-inflammatory Action of Statins in Cardiovascular Disease: the Role of Inflammasome and Toll-Like Receptor Pathways. Clin Rev Allergy Immunol 2020; 60:175-199. [PMID: 32378144 PMCID: PMC7985098 DOI: 10.1007/s12016-020-08791-9] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is one type of cardiovascular disease (CVD) in which activation of the NLRP3 inflammasome and toll-like receptor (TLR) pathways is implicated. One of the most effective treatments for atherosclerosis is the use of statin medications. Recent studies have indicated that statins, in addition to their lipid-lowering effects, exert inhibitory and/or stimulatory effects on the NLRP3 inflammasome and TLRs. Some of the statins lead to activation of the inflammasome and subsequently cause secretion of IL-1β and IL-18. Thus, these actions may further aggravate the disease. On the other hand, some statins cause inhibition of the inflammasome or TLRs and along with lipid-lowering, help to improve the disease by reducing inflammation. In this article, we discuss these contradictory studies and the mechanisms of action of statins on the NLRP3 inflammasome and TLR pathways. The dose-dependent effects of statins on the NLRP3 complex are related to their chemistry, pharmacokinetic properties, and danger signals. Lipophilic statins have more pleiotropic effects on the NLRP3 complex in comparison to hydrophilic statins. Statins can suppress TLR4/MyD88/NF-ĸB signaling and cause an immune response shift to an anti-inflammatory response. Furthermore, statins inhibit the NF-ĸB pathway by decreasing the expression of TLRs 2 and 4. Statins are cost-effective drugs, which should have a continued future in the treatment of atherosclerosis due to both their immune-modulating and lipid-lowering effects.
Collapse
Affiliation(s)
- Khadijeh Koushki
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sanaz Keshavarz Shahbaz
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kazem Mashayekhi
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeinab Deris Zayeri
- Golestan Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Khalid Al-Rasadi
- Medical Research Centre, Sultan Qaboos University, Muscat, Oman
- Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
16
|
|
17
|
Farrah TE, Anand A, Gallacher PJ, Kimmitt R, Carter E, Dear JW, Mills NL, Webb DJ, Dhaun N. Endothelin Receptor Antagonism Improves Lipid Profiles and Lowers PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) in Patients With Chronic Kidney Disease. Hypertension 2019; 74:323-330. [PMID: 31177906 PMCID: PMC6635059 DOI: 10.1161/hypertensionaha.119.12919] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Dyslipidemia is common in chronic kidney disease (CKD). Despite statins, many patients fail to adequately lower lipids and remain at increased risk of cardiovascular disease. Selective ETA (endothelin-A) receptor antagonists reduce cardiovascular disease risk factors. Preclinical data suggest that ETA antagonism has beneficial effects on circulating lipids. We assessed the effects of selective ETA antagonism on circulating lipids and PCSK9 (proprotein convertase subtilisin/kexin type 9) in CKD. This was a secondary analysis of a fully randomized, double-blind, 3-phase crossover study. Twenty-seven subjects with predialysis CKD on optimal cardio- and renoprotective treatment were randomly assigned to receive 6 weeks dosing with placebo, the selective ETA receptor antagonist, sitaxentan, or long-acting nifedipine. We measured circulating lipids and PCSK9 at baseline and then after 3 and 6 weeks. Baseline lipids and PCSK9 did not differ before each study phase. Whereas placebo and nifedipine had no effect on lipids, 6 weeks of ETA antagonism significantly reduced total (-11±1%) and low-density lipoprotein-associated (-20±3%) cholesterol, lipoprotein (a) (-16±2%) and triglycerides (-20±4%); high-density lipoprotein-associated cholesterol increased (+14±2%), P<0.05 versus baseline for all. Additionally, ETA receptor antagonism, but neither placebo nor nifedipine, reduced circulating PCSK9 (-19±2%; P<0.001 versus baseline; P<0.05 versus nifedipine and placebo). These effects were independent of statin use and changes in blood pressure or proteinuria. Selective ETA antagonism improves lipid profiles in optimally-managed patients with CKD, effects that may occur through a reduction in circulating PCSK9. ETA receptor antagonism offers a potentially novel strategy to reduce cardiovascular disease risk in CKD. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT00810732.
Collapse
Affiliation(s)
- Tariq E. Farrah
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute (T.E.F., A.A., P.J.G., R.K., E.C., J.W.D., N.L.M., D.J.W., N.D.),Department of Renal Medicine, Royal Infirmary of Edinburgh (T.E.F., P.J.G., N.D.)
| | - Atul Anand
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute (T.E.F., A.A., P.J.G., R.K., E.C., J.W.D., N.L.M., D.J.W., N.D.)
| | - Peter J. Gallacher
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute (T.E.F., A.A., P.J.G., R.K., E.C., J.W.D., N.L.M., D.J.W., N.D.),Department of Renal Medicine, Royal Infirmary of Edinburgh (T.E.F., P.J.G., N.D.)
| | - Robert Kimmitt
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute (T.E.F., A.A., P.J.G., R.K., E.C., J.W.D., N.L.M., D.J.W., N.D.)
| | - Edwin Carter
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute (T.E.F., A.A., P.J.G., R.K., E.C., J.W.D., N.L.M., D.J.W., N.D.)
| | - James W. Dear
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute (T.E.F., A.A., P.J.G., R.K., E.C., J.W.D., N.L.M., D.J.W., N.D.)
| | - Nicholas L. Mills
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute (T.E.F., A.A., P.J.G., R.K., E.C., J.W.D., N.L.M., D.J.W., N.D.)
| | - David J. Webb
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute (T.E.F., A.A., P.J.G., R.K., E.C., J.W.D., N.L.M., D.J.W., N.D.)
| | - Neeraj Dhaun
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute (T.E.F., A.A., P.J.G., R.K., E.C., J.W.D., N.L.M., D.J.W., N.D.),Department of Renal Medicine, Royal Infirmary of Edinburgh (T.E.F., P.J.G., N.D.)
| |
Collapse
|
18
|
Yan J, Pan Y, Xiao J, Ma W, Li L, Zhong M, Long H, Kong F, Shao W. High Level of Lipoprotein(a) as Predictor for Recurrent Heart Failure in Patients with Chronic Heart Failure: a Cohort Study. Arq Bras Cardiol 2019; 113:197-204. [PMID: 31340235 PMCID: PMC6777886 DOI: 10.5935/abc.20190120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/14/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Elevated plasma levels of Lipoprotein(a) [Lp(a)] are recognized as a significant risk factor for atherosclerotic vascular disease. However, there are limited data regarding association between Lp(a) and recurrent heart failure (HF) in patients with chronic HF caused by coronary heart disease (CHD). OBJECTIVE Elevated levels of Lp(a) might have a prognostic impact on recurrent HF in patients with chronic HF caused by CHD. METHODS A total of 309 patients with chronic HF caused by CHD were consecutively enrolled in this study. The patients were divided into 2 groups according to whether Lp(a) levels were above or below the median level for the entire cohort (20.6 mg/dL): the high Lp(a) group (n = 155) and the low Lp(a) group (n = 154). A 2-sided p < 0.05 was statistically considered significant. RESULTS During the median follow-up period of 186 days, 31 cases out of a total of 309 patients (10.03%) could not be reached during follow-up. A Kaplan-Meier analysis demonstrated that patients with higher Lp(a) levels had a higher incidence of recurrent HF than those with lower Lp(a) levels (log-rank < 0.0001). A multivariate Cox regression analysis revealed that Lp(a) levels were independently correlated with the incidence of recurrent HF after adjustment of potential confounders (hazard ratio: 2.720, 95 % confidence interval: 1.730-4.277, p < 0.0001). CONCLUSIONS In Chinese patients with chronic HF caused by CHD, elevated levels of Lp(a) are independently associated with recurrent HF.
Collapse
Affiliation(s)
- Jianlong Yan
- Huadu District People's Hospital, Southern Medical University - Cardiology, Guangzhou - China
| | - Yanbin Pan
- Huadu District People's Hospital, Southern Medical University - Intensive Care Unit., Guangzhou - China
| | - Junhui Xiao
- Huadu District People's Hospital, Southern Medical University - Cardiology, Guangzhou - China
| | - Wenxue Ma
- Huadu District People's Hospital, Southern Medical University - Cardiology, Guangzhou - China
| | - Li Li
- Huadu District People's Hospital, Southern Medical University - Cardiology, Guangzhou - China
| | - Mingjiang Zhong
- Huadu District People's Hospital, Southern Medical University - Cardiology, Guangzhou - China
| | - Haiquan Long
- Huadu District People's Hospital, Southern Medical University - Cardiology, Guangzhou - China
| | - Fanliang Kong
- Huadu District People's Hospital, Southern Medical University - Cardiology, Guangzhou - China
| | - Wenming Shao
- The First Affiliated Hospital of Jinan University - Emergency, Guangzhou - China
| |
Collapse
|
19
|
Multilayer-Coated Tablet of Clopidogrel and Rosuvastatin: Preparation and In Vitro/In vivo Characterization. Pharmaceutics 2019; 11:pharmaceutics11070313. [PMID: 31277408 PMCID: PMC6680394 DOI: 10.3390/pharmaceutics11070313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 11/21/2022] Open
Abstract
The acid lability of rosuvastatin hinders the preparation of mixed combination formulations of rosuvastatin with acidic drugs such as clopidogrel. Therefore, the purpose of this study was to develop a multilayer-coated tablet that avoids physicochemical interactions between rosuvastatin and clopidogrel. Among the tested hydrophobic materials, glyceryl behenate was most effective at inhibiting the production of lactone, the acid degradation product of rosuvastatin. Therefore, the multilayer-coated tablet included a hydrophobic separation layer consisting of glyceryl behenate between the clopidogrel core tablet and the rosuvastatin coating layer. In order to prevent delayed dissolution by the stable hydrophobic separation layer, crospovidone was added into the clopidogrel core tablet as an effective disintegrant. Copovidone was also added to the coating layer of rosuvastatin, achieving a dissolution profile comparable to that of the reference drug, Crestor®. The resulting multilayer-coated tablet exhibited similar pharmacokinetic profiles to those of reference drugs (Plavix® and Crestor®) in beagle dogs, and there was no statistically significant difference in the maximum plasma concentration (Cmax), the time to reach the maximum plasma concentration (Tmax), or the area under the plasma-concentration time curve (AUC) between the test and reference formulations. The storage stability tests showed that the amounts of acid degradation products and total impurities were comparable to that of the reference drug. In conclusion, the present study successfully developed a stable multilayer-coated tablet containing both clopidogrel and rosuvastatin that may improve the patient compliance in combination therapy for cardiovascular diseases.
Collapse
|
20
|
Yahya R, Berk K, Verhoeven A, Bos S, van der Zee L, Touw J, Erhart G, Kronenberg F, Timman R, Sijbrands E, Roeters van Lennep J, Mulder M. Statin treatment increases lipoprotein(a) levels in subjects with low molecular weight apolipoprotein(a) phenotype. Atherosclerosis 2019; 289:201-205. [PMID: 31327478 DOI: 10.1016/j.atherosclerosis.2019.07.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 05/20/2019] [Accepted: 07/02/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND AND AIMS We aimed to evaluate the effect of statin treatment initiation on lipoprotein(a) [Lp(a)] levels in patients with dyslipidemia, and the interactions with the apolipoprotein(a) [apo(a)] phenotype, LPA single nucleotide polymorphisms (SNPs) and change in LDL cholesterol. METHODS The study population consisted of patients with dyslipidemia, predominantly familial hypercholesterolemia, who first initiated statin treatment (initiation group; n = 39) or were already on stable statin treatment for at least 4 months (control group; n = 42). Plasma Lp(a) levels were determined with a particle-enhanced immunoturbidimetric assay before and at least 2 months after start of statin treatment in individuals of the initiation group, and at two time points with an interval of at least 2 months in the control group. High and low molecular weight (HMW and LMW, respectively) apo(a) phenotype was determined by immunoblotting, and the common LPA SNPs rs10455872, rs3798220 and rs41272110 by Taqman assay. RESULTS Plasma Lp(a) levels did not increase significantly in the initiation group (median 20.5 (IQR 10.9-80.7) to 23.3 (10.8-71.8) mg/dL; p = 0.09) nor in the control group (30.9 (IQR 9.2-147.0) to 31.7 (IQR 10.9-164.0) mg/dL; p = 0.61). In patients with the LMW apo(a) phenotype, Lp(a) levels increased significantly from 66.4 (IQR 23.5-148.3) to 97.4 (IQR 24.9-160.4) mg/dL (p = 0.026) in the initiation group, but not in the control group and not in patients characterized by the HMW apo(a) phenotype. Interactions with common LPA SNPs and change in LDL cholesterol were not significant. CONCLUSIONS Statins affect Lp(a) levels differently in patients with dyslipidemia depending on the apo(a) phenotype. Statins increase Lp(a) levels exclusively in patients with the LMW apo(a) phenotype.
Collapse
Affiliation(s)
- Reyhana Yahya
- Department of Internal Medicine, division of Vascular Medicine and Pharmacology, Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Kirsten Berk
- Department of Internal Medicine, division of Vascular Medicine and Pharmacology, Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands; Department of Dietetics and Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Adrie Verhoeven
- Department of Internal Medicine, division of Vascular Medicine and Pharmacology, Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Sven Bos
- Department of Internal Medicine, division of Vascular Medicine and Pharmacology, Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Leonie van der Zee
- Department of Internal Medicine, division of Vascular Medicine and Pharmacology, Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jeanette Touw
- Department of Internal Medicine, division of Vascular Medicine and Pharmacology, Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Gertraud Erhart
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Reinier Timman
- Department of Psychiatry, Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eric Sijbrands
- Department of Internal Medicine, division of Vascular Medicine and Pharmacology, Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jeanine Roeters van Lennep
- Department of Internal Medicine, division of Vascular Medicine and Pharmacology, Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Monique Mulder
- Department of Internal Medicine, division of Vascular Medicine and Pharmacology, Division of Medical Psychology and Psychotherapy, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
21
|
Rawther T, Tabet F. Biology, pathophysiology and current therapies that affect lipoprotein (a) levels. J Mol Cell Cardiol 2019; 131:1-11. [DOI: 10.1016/j.yjmcc.2019.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/22/2019] [Accepted: 04/09/2019] [Indexed: 12/11/2022]
|
22
|
Tsimikas S, Gordts PLSM, Nora C, Yeang C, Witztum JL. Statin therapy increases lipoprotein(a) levels. Eur Heart J 2019; 41:2275-2284. [DOI: 10.1093/eurheartj/ehz310] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/18/2018] [Accepted: 04/27/2019] [Indexed: 01/03/2023] Open
Abstract
Abstract
Aims
Lipoprotein(a) [Lp(a)] is elevated in 20–30% of people. This study aimed to assess the effect of statins on Lp(a) levels.
Methods and results
This subject-level meta-analysis includes 5256 patients (1371 on placebo and 3885 on statin) from six randomized trials, three statin-vs.-placebo trials, and three statin-vs.-statin trials, with pre- and on-treatment (4–104 weeks) Lp(a) levels. Statins included atorvastatin 10 mg/day and 80 mg/day, pravastatin 40 mg/day, rosuvastatin 40 mg/day, and pitavastatin 2 mg/day. Lipoprotein(a) levels were measured with the same validated assay. The primary analysis of Lp(a) is based on the log-transformed data. In the statin-vs.-placebo pooled analysis, the ratio of geometric means [95% confidence interval (CI)] for statin to placebo is 1.11 (1.07–1.14) (P < 0.0001), with ratio >1 indicating a higher increase in Lp(a) from baseline in statin vs. placebo. The mean percent change from baseline ranged from 8.5% to 19.6% in the statin groups and −0.4% to −2.3% in the placebo groups. In the statin-vs.-statin pooled analysis, the ratio of geometric means (95% CI) for atorvastatin to pravastatin is 1.09 (1.05–1.14) (P < 0.0001). The mean percent change from baseline ranged from 11.6% to 20.4% in the pravastatin group and 18.7% to 24.2% in the atorvastatin group. Incubation of HepG2 hepatocytes with atorvastatin showed an increase in expression of LPA mRNA and apolipoprotein(a) protein.
Conclusion
This meta-analysis reveals that statins significantly increase plasma Lp(a) levels. Elevations of Lp(a) post-statin therapy should be studied for effects on residual cardiovascular risk.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Vascular Medicine Program, Sulpizio Cardiovascular Center, Division of Cardiovascular Diseases, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
| | - Philip L S M Gordts
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
| | - Chelsea Nora
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
| | - Calvin Yeang
- Vascular Medicine Program, Sulpizio Cardiovascular Center, Division of Cardiovascular Diseases, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
| | - Joseph L Witztum
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
| |
Collapse
|
23
|
Dijk W, Cariou B. Efficacy and safety of proprotein convertase subtilisin/kexin 9 inhibitors in people with diabetes and dyslipidaemia. Diabetes Obes Metab 2019; 21 Suppl 1:39-51. [PMID: 31002456 DOI: 10.1111/dom.13636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/05/2019] [Accepted: 01/07/2019] [Indexed: 12/11/2022]
Abstract
Diabetic dyslipidaemia, characterized by quantitative, qualitative and kinetic changes in all major circulating lipids, contributes to the increased cardiovascular risk in patients with type 2 diabetes mellitus (T2DM). A promising therapeutic avenue is the inhibition of the proprotein convertase subtilisin kexin 9 (PCSK9) with human monoclonal antibodies (mAbs) that potently reduce plasma low-density lipoprotein cholesterol (LDL-C) levels on top of statin treatment. The aim of this review is to evaluate the efficacy of PCSK9 inhibitors to lower the residual cardiovascular risk of T2DM patients and to discuss the safety of PCSK9 inhibition in these patients. PCSK9 inhibitors potently lower plasma LDL-C levels in T2DM patients and reduce risk for the development of cardiovascular disease. Anti-PCSK9 mAbs are generally not more or less effective in T2DM patients compared to a general high-risk population. Nevertheless, due to their higher cardiovascular risk, the absolute risk reduction of major cardiovascular events is more significant in T2DM patients. This suggests that treatment of T2DM patients with anti-PCSK9 mAbs could be attractive from a cost-effectiveness perspective. Treatment with anti-PCSK9 mAbs did not result in significant treatment-emergent adverse effects. While genetic studies suggest a potential link between PCSK9 inhibition and glucose homeostasis, anti-PCSK9 mAbs did not worsen glycaemic control in T2DM patients, but their safety should be verified after a longer-term follow-up.
Collapse
Affiliation(s)
- Wieneke Dijk
- L'institut du thorax, INSERM, CNRS, University of Nantes, CHU Nantes, Nantes, France
| | - Bertrand Cariou
- L'institut du thorax, INSERM, CNRS, University of Nantes, CHU Nantes, Nantes, France
- CIC INSERM 1413, CHU Nantes, Department of Endocrinology, L'institut du thorax, Nantes, France
| |
Collapse
|
24
|
A Meta-Analysis of the Effect of PCSK9-Monoclonal Antibodies on Circulating Lipoprotein (a) Levels. Am J Cardiovasc Drugs 2019; 19:87-97. [PMID: 30229525 DOI: 10.1007/s40256-018-0303-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is an atherogenic lipoprotein. While no effective therapy for Lp(a) is currently available, recently, several pooled analyses with small sample sizes have suggested that proprotein convertase subtilisin/kexin type 9 monoclonal antibodies (PCSK9-mAbs) could reduce circulating Lp(a) levels. This meta-analysis was performed to comprehensively investigate the efficacy of PCSK9-mAbs with respect to serum Lp(a) concentrations. METHODS PubMed, MEDLINE, Embase, ClinicalTrials.gov, Cochrane CENTRAL, Web of Science and recent conferences up to July 2018 were searched. Randomized clinical trials evaluating the effect of PCSK9-mAbs and control treatment on plasma Lp(a) concentrations were included. Mean differences and odds ratios with 95% confidence intervals (CIs) were used. RESULTS Twenty-seven randomized clinical trials with a total of 11,864 participants were included. PCSK9-mAbs showed a significant efficacy in reducing Lp(a) (- 21.9%, 95% CI - 24.3 to - 19.5), irrespective of PCSK9-mAb types, treatment duration, participant characteristics, treatment methods, differences of control treatment, baseline Lp(a) levels, and test methods. The greatest reduction was achieved with 150 mg alirocumab biweekly (- 24.6%, 95% CI - 28.0 to - 21.2) and 140 mg evolocumab monthly (- 26.8%, 95% CI - 31.6 to - 21.9). Meta-regression analyses found that the more intense low-density lipoprotein cholesterol levels declined during PCSK9-mAb treatment, the greater the reduction in Lp(a) levels. Safety was in accordance with previous reports. CONCLUSIONS The results of this analysis suggested that PCSK9-mAbs could significantly reduce circulating Lp(a) levels. Long-term studies may be needed to confirm the effect of PCSK9-mAbs on Lp(a) in the future.
Collapse
|
25
|
Zawacki AW, Dodge A, Woo KM, Ralphe JC, Peterson AL. In pediatric familial hypercholesterolemia, lipoprotein(a) is more predictive than LDL-C for early onset of cardiovascular disease in family members. J Clin Lipidol 2018; 12:1445-1451. [DOI: 10.1016/j.jacl.2018.07.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/11/2018] [Accepted: 07/25/2018] [Indexed: 10/28/2022]
|
26
|
Enkhmaa B, Anuurad E, Zhang W, Li CS, Kaplan R, Lazar J, Merenstein D, Karim R, Aouizerat B, Cohen M, Butler K, Pahwa S, Ofotokun I, Adimora AA, Golub E, Berglund L. Effect of antiretroviral therapy on allele-associated Lp(a) level in women with HIV in the Women's Interagency HIV Study. J Lipid Res 2018; 59:1967-1976. [PMID: 30012717 DOI: 10.1194/jlr.p084517] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 06/08/2018] [Indexed: 11/20/2022] Open
Abstract
We previously demonstrated an association between lipoprotein (a) [Lp(a)] levels and atherosclerosis in human immunodeficiency virus (HIV)-seropositive women. The effects of antiretroviral therapy (ART) on Lp(a) levels in relation to apo(a) size polymorphism remain unclear. ART effects on allele-specific apo(a) level (ASL), an Lp(a) level associated with individual apo(a) alleles within each allele-pair, were determined in 126 HIV-seropositive women. ART effects were tested by a mixed-effects model across pre-ART and post-ART first and third visits. Data from 120 HIV-seronegative women were used. The mean age was 38 years; most were African-American (∼70%). Pre-ART ASLs associated with the larger (4.6 mg/dl vs. 8.0 mg/dl, P = 0.024) or smaller (13 mg/dl vs. 19 mg/dl, P = 0.041) apo(a) sizes were lower in the HIV-seropositive versus HIV-seronegative group, as was the prevalence of a high Lp(a) level (P = 0.013). Post-ART ASL and prevalence of high Lp(a) or apo(a) sizes and frequency of small size apo(a) (22 kringles) did not differ between the two groups. ART increased Lp(a) level (from 18 to 24 mg/dl, P < 0.0001) and both ASLs (P < 0.001). In conclusion, regardless of genetic control, Lp(a) can be modulated by HIV and its treatment. ART initiation abrogates HIV-induced suppression of Lp(a) levels and ASLs, contributing to promote CVD risk in HIV-seropositive individuals.
Collapse
Affiliation(s)
- Byambaa Enkhmaa
- Departments of Internal Medicine University of California, Davis, Davis, CA 95616
| | - Erdembileg Anuurad
- Departments of Internal Medicine University of California, Davis, Davis, CA 95616
| | - Wei Zhang
- Departments of Internal Medicine University of California, Davis, Davis, CA 95616
| | - Chin-Shang Li
- Public Health Sciences, University of California, Davis, Davis, CA 95616
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Jason Lazar
- Department of Cardiovascular Disease, State University of New York Downstate Medical Center, Brooklyn, NY 11203
| | - Dan Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC 20007
| | - Roksana Karim
- Department Preventive Medicine, University of Southern California, Los Angeles, Los Angeles, CA 90007
| | - Brad Aouizerat
- Department of Oral and Maxillofacial Surgery, New York University, New York, NY 10003
| | - Mardge Cohen
- Department of Medicine, Rush University and Stroger Hospital, Cook County Bureau of Health Services, Chicago, IL 60612
| | - Kenneth Butler
- Division of Geriatric Medicine/Gerontology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Savita Pahwa
- Miami Center for AIDS Research, University of Miami, Miami, FL 33136
| | - Igho Ofotokun
- Department of Medicine, Infectious Diseases, Emory School of Medicine, Atlanta, GA 30307
| | - Adaora A Adimora
- Division of Infectious Diseases, University of North Carolina, Chapel Hill, NC 27599
| | - Elizabeth Golub
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Lars Berglund
- Departments of Internal Medicine University of California, Davis, Davis, CA 95616
| |
Collapse
|
27
|
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: 53] [Impact Index Per Article: 8.8] [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
|
28
|
Hulin A, Hego A, Lancellotti P, Oury C. Advances in Pathophysiology of Calcific Aortic Valve Disease Propose Novel Molecular Therapeutic Targets. Front Cardiovasc Med 2018; 5:21. [PMID: 29594151 PMCID: PMC5862098 DOI: 10.3389/fcvm.2018.00021] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 02/26/2018] [Indexed: 01/17/2023] Open
Abstract
Calcific Aortic Valve Disease (CAVD) is the most common heart valve disease and its incidence is expected to rise with aging population. No medical treatment so far has shown slowing progression of CAVD progression. Surgery remains to this day the only way to treat it. Effective drug therapy can only be achieved through a better insight into the pathogenic mechanisms underlying CAVD. The cellular and molecular events leading to leaflets calcification are complex. Upon endothelium cell damage, oxidized LDLs trigger a proinflammatory response disrupting healthy cross-talk between valve endothelial and interstitial cells. Therefore, valve interstitial cells transform into osteoblasts and mineralize the leaflets. Studies have investigated signaling pathways driving and connecting lipid metabolism, inflammation and osteogenesis. This review draws a summary of the recent advances and discusses their exploitation as promising therapeutic targets to treat CAVD and reduce valve replacement.
Collapse
Affiliation(s)
- Alexia Hulin
- GIGA Cardiovascular Sciences, Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, University of Liège, CHU Sart Tilman, Liège, Belgium
| | - Alexandre Hego
- GIGA Cardiovascular Sciences, Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, University of Liège, CHU Sart Tilman, Liège, Belgium
| | - Patrizio Lancellotti
- GIGA Cardiovascular Sciences, Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, University of Liège, CHU Sart Tilman, Liège, Belgium.,GIGA Cardiovascular Sciences, Department of Cardiology, University of Liège Hospital, Heart Valve Clinic, CHU Sart Tilman, Liège, Belgium.,Gruppo Villa Maria Care and Research, Anthea Hospital, Bari, Italy
| | - Cécile Oury
- GIGA Cardiovascular Sciences, Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, University of Liège, CHU Sart Tilman, Liège, Belgium
| |
Collapse
|
29
|
Viney NJ, Yeang C, Yang X, Xia S, Witztum JL, Tsimikas S. Relationship between "LDL-C", estimated true LDL-C, apolipoprotein B-100, and PCSK9 levels following lipoprotein(a) lowering with an antisense oligonucleotide. J Clin Lipidol 2018; 12:702-710. [PMID: 29574075 DOI: 10.1016/j.jacl.2018.02.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/21/2018] [Accepted: 02/24/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND The laboratory measurement of "low-density lipoprotein cholesterol (LDL-C)" includes the cholesterol content of lipoprotein(a) (Lp(a)-C). OBJECTIVE To estimate the "true" LDL-C in relation to changes in apolipoprotein B-100 (apoB-100) and assess changes in proprotein convertase subtilisin/kexin 9 (PCSK9) levels in patients with elevated Lp(a) treated with IONIS-APO(a)Rx. METHODS: A pooled placebo group (n = 29), and cohort A (n = 24, baseline Lp(a) 50-175 mg/dL) and cohort B (n = 8, baseline Lp(a) > 175 mg/dL) treated with IONIS-APO(a)Rx were studied. Lp(a) particle number, ultracentrifugation-measured "LDL-C", apoB-100, total PCSK9, and lipoprotein-associated PCSK9 (PCSK9-Lp(a), PCSK9-apoB, PCSK9-apoAI) were measured. Lp(a)-cholesterol (Lp(a)-C) and LDL-C corrected for Lp(a)-C (LDL-Ccorr) were calculated. RESULTS Baseline mean (standard deviation) "LDL-C" was 120 (42), 128 (45), and 112 (39) mg/dL in placebo, cohorts A and B, respectively, whereas LDL-Ccorr was 86 (48), 96 (43), and 57 (37) mg/dL (P < .001 compared with placebo), representing 28%, 25%, and 50% lower levels than "LDL-C". Following IONIS-APO(a)Rx treatment at day 85/99, Lp(a) particle number and Lp(a)-C decreased -66.8% and -71.6%, apoB-100 -10.3% and -17.5%, "LDL-C" -11.8% and -22.7%, (P < .001 for all vs placebo), whereas LDL-Ccorr increased +10.4% (P = .66) and +49.9% (P < .001) in cohorts A and B, respectively. Total PCSK9 did not change but PCSK9-Lp(a) decreased with IONIS-APO(a)Rx vs placebo (-39.0% vs +8.4%, P < .001). CONCLUSION LDL-Ccorr is lower than laboratory "LDL-C" in patients with elevated Lp(a). Following apolipoprotein(a) inhibition and decline in Lp(a) and Lp(a)-C, the decline in apoB-100 is consistent with the notion that LDL devoid of apo(a) is cleared faster than Lp(a). These types of analyses may provide insights into the mechanisms of drugs affecting Lp(a) levels in clinical trials.
Collapse
Affiliation(s)
| | - Calvin Yeang
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Xiaohong Yang
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Joseph L Witztum
- Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| | - Sotirios Tsimikas
- Ionis Pharmaceuticals, Carlsbad, CA, USA; Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
30
|
Giardina S, Hernández-Alonso P, Díaz-López A, Salas-Huetos A, Salas-Salvadó J, Bulló M. Changes in circulating miRNAs in healthy overweight and obese subjects: Effect of diet composition and weight loss. Clin Nutr 2017; 38:438-443. [PMID: 29233588 DOI: 10.1016/j.clnu.2017.11.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/20/2017] [Accepted: 11/19/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) are small non-coding RNA molecules that can play an important role in several chronic metabolic conditions, including obesity. However, to date little is known about how they are regulated. Weight loss induced by surgical procedures has been effective at modulating specific circulating miRNAs, but the effect of energy-restricted diets with different macronutrient compositions on circulating miRNAs is not well understood. The objective of the present analysis was to explore the effect of three energy-restricted diets of different macronutrient composition and carbohydrate quality on plasma miRNA levels. METHODS The GLYNDIET study is a 6-month, parallel, randomized clinical trial conducted on overweight and obese subjects who were randomized to one of three different dietary intervention groups: i) a moderate-carbohydrate and low glycemic index diet (LGI), ii) a moderate-carbohydrate and high glycemic index diet (HGI), and iii) a low-fat and high glycemic index diet (LF). We assessed the genome-wide circulating miRNA profile in a subsample of eight randomly selected participants. A total of 8 miRNAs (miR-411, miR-432, miR-99b, miR-340, miR-423, miR-361, let-7c) were differently quantified according to diet intervention, and were therefore longitudinally validated in 103 participants before and after the energy-restricted diets. RESULTS Circulating miR-361 levels were lower in the LGI group than in the HGI group, even after adjusting for differences in weight loss. The intra-group analyses demonstrated a significant down-regulation of all miRNAs screened in our study subjects after the LGI intervention. Similarly, miR-139 and miR-340 were down-regulated after the HGI intervention, while miR-139, miR-432 and miR-423 were down-regulated after the low-fat diet. Changes in circulating miR-139 and let-7c were significantly associated with changes in lipid profile and insulin resistance. CONCLUSION An energy-restricted low-glycemic index diet down-regulates circulating miRNA-361 more than an energy-restricted high-glycemic index, regardless of the magnitude of the weight loss.
Collapse
Affiliation(s)
- S Giardina
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - P Hernández-Alonso
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - A Díaz-López
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - A Salas-Huetos
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - J Salas-Salvadó
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - M Bulló
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|
31
|
Vuorio A, Watts GF, Kovanen PT. Depicting new pharmacological strategies for familial hypercholesterolaemia involving lipoprotein (a). Eur Heart J 2017; 38:3555-3559. [DOI: 10.1093/eurheartj/ehx546] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 09/04/2017] [Indexed: 01/17/2023] Open
|
32
|
Enkhmaa B, Anuurad E, Zhang W, Yue K, Li CS, Berglund L. The roles of apo(a) size, phenotype, and dominance pattern in PCSK9-inhibition-induced reduction in Lp(a) with alirocumab. J Lipid Res 2017; 58:2008-2016. [PMID: 28798072 DOI: 10.1194/jlr.m078212] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/09/2017] [Indexed: 11/20/2022] Open
Abstract
An elevated level of lipoprotein (a) [Lp(a)] is a risk factor for CVD. Alirocumab, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9, is reported to reduce Lp(a) levels. The relationship of Lp(a) reduction with apo(a) size polymorphism, phenotype, and dominance pattern and LDL cholesterol (LDL-C) reduction was evaluated in a pooled analysis of 155 hypercholesterolemic patients (75 with heterozygous familial hypercholesterolemia) from two clinical trials. Alirocumab significantly reduced total Lp(a) (pooled median: -21%, P = 0.0001) and allele-specific apo(a), an Lp(a) level carried by the smaller (median: -18%, P = 0.002) or the larger (median: -37%, P = 0.0005) apo(a) isoform, at week 8 versus baseline. The percent reduction in Lp(a) level with alirocumab was similar across apo(a) phenotypes (single vs. double bands) and carriers and noncarriers of a small size apo(a) (≤22 kringles). The percent reduction in LDL-C correlated significantly with the percent reduction in Lp(a) level (r = 0.407, P < 0.0001) and allele-specific apo(a) level associated with the smaller (r = 0.390, P < 0.0001) or larger (r = 0.270, P = 0.0183) apo(a) sizes. In conclusion, alirocumab-induced Lp(a) reduction was independent of apo(a) phenotypes and the presence or absence of a small size apo(a).
Collapse
Affiliation(s)
- Byambaa Enkhmaa
- Departments of Internal Medicine University of California, Davis, CA
| | | | - Wei Zhang
- Departments of Internal Medicine University of California, Davis, CA
| | - Kun Yue
- Public Health Sciences, University of California, Davis, CA.,Department of Statistics and Actuarial Science, University of Hong Kong, Pokfulam, Hong Kong
| | - Ching-Shang Li
- Public Health Sciences, University of California, Davis, CA
| | - Lars Berglund
- Departments of Internal Medicine University of California, Davis, CA
| |
Collapse
|
33
|
Xu MX, Liu C, He YM, Yang XJ, Zhao X. Long-term statin therapy could be efficacious in reducing the lipoprotein (a) levels in patients with coronary artery disease modified by some traditional risk factors. J Thorac Dis 2017; 9:1322-1332. [PMID: 28616285 DOI: 10.21037/jtd.2017.04.32] [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] [Indexed: 01/14/2023]
Abstract
BACKGROUND Lipoprotein (a) [Lp (a)] is a well-established risk factor for coronary artery disease (CAD). However, up till now, treatment of patients with higher Lp (a) levels is challenging. This current study aimed to investigate the therapeutic effects of short-, medium and long-term statin use on the Lp (a) reduction and its modifying factors. METHODS The therapeutic duration was categorized into short-term (median, 39 days), medium term (median, 219 days) and long-term (median, 677 days). The lipid profiles before therapy served as baselines. Patients at short-, medium or long-term exactly matched with those at baseline. Every patient's lipid profiles during the follow-ups were compared to his own ones at baselines. RESULTS The current study demonstrated that long-term statin therapy significantly decreased the Lp (a) levels in CAD patients while short-term or medium term statin therapy didn't. When grouped by statin use, only long-term simvastatin use significantly decreased the Lp (a) levels while long-term atorvastatin use insignificantly decreased the Lp (a) levels. Primary hypertension (PH), DM, low density lipoprotein cholesterol (LDL-C) and high density lipoprotein cholesterol (HDL-C) could modify the therapeutic effects of statin use on the Lp (a) levels in CAD patients. CONCLUSIONS The long-term statin therapy could be efficacious in reducing the Lp (a) levels in CAD patients, which has been modified by some traditional risk factors. In the era of commercial unavailability of more reliable Lp (a) lowering drugs, our findings will bolster confidence in fighting higher Lp (a) abnormalities both for patients and for doctors.
Collapse
Affiliation(s)
- Ming-Xing Xu
- Division of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Chang Liu
- Division of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yong-Ming He
- Division of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiang-Jun Yang
- Division of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xin Zhao
- Division of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
34
|
Feng Z, Li HL, Bei WJ, Guo XS, Wang K, Yi SX, Luo DM, Li XD, Chen SQ, Ran P, Chen PY, Islam SMS, Chen JY, Liu Y, Zhou YL. Association of lipoprotein(a) with long-term mortality following coronary angiography or percutaneous coronary intervention. Clin Cardiol 2017; 40:674-678. [PMID: 28444976 DOI: 10.1002/clc.22712] [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: 12/16/2016] [Revised: 02/23/2017] [Accepted: 02/28/2017] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND There is no consistent evidence to suggest the association of plasma lipoprotein(a) (Lp[a]) with long-term mortality in patients undergoing coronary angiography (CAG) or percutaneous coronary intervention (PCI). HYPOTHESIS Level of Lp(a) is associated with long-term mortality following CAG or PCI. METHODS We enrolled 1684 patients with plasma Lp(a) data undergoing CAG or PCI between April 2009 and December 2013. The patients were divided into 2 groups: a low-Lp(a) group (Lp[a] <16.0 mg/dL; n = 842) and a high-Lp(a) group (Lp[a] ≥16.0 mg/dL; n = 842). RESULTS In-hospital mortality was not significantly different between the high and low Lp(a) groups (0.8% vs 0.5%, respectively; P = 0.364). During the median follow-up period of 1.95 years, the high-Lp(a) group had a higher long-term mortality than did the low-Lp(a) group (5.8% vs 2.5%, respectively; P = 0.003). After adjustment of confounders, multivariate Cox regression analysis revealed that a higher Lp(a) level was an independent predictor of long-term mortality (hazard ratio: 1.96, 95% confidence interval: 1.07-3.59, P = 0.029). CONCLUSIONS Our data suggested that an elevated Lp(a) level was significantly associated with long-term mortality following CAG or PCI. However, additional larger multicenter studies will be required to investigate the predictive value of Lp(a) levels and evaluate the benefit of controlling Lp(a) levels for patients undergoing CAG or PCI.
Collapse
Affiliation(s)
- Zhe Feng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Hua-Long Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Wei-Jie Bei
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiao-Sheng Guo
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Kun Wang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.,Department of Graduate School (Wang), Southern Medical University, Guangzhou, Guangdong, China
| | - Shi-Xin Yi
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - De-Mou Luo
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xi-da Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Shi-Qun Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Peng Ran
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Peng-Yuan Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | | | - Ji-Yan Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yong Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Ying-Ling Zhou
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Key Laboratory of Coronary Disease, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
35
|
Sjouke B, Yahya R, Tanck MWT, Defesche JC, de Graaf J, Wiegman A, Kastelein JJP, Mulder MT, Hovingh GK, Roeters van Lennep JE. Plasma lipoprotein(a) levels in patients with homozygous autosomal dominant hypercholesterolemia. J Clin Lipidol 2017; 11:507-514. [PMID: 28502508 DOI: 10.1016/j.jacl.2017.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 01/25/2023]
Abstract
BACKGROUND Patients with autosomal dominant hypercholesterolemia (ADH), caused by mutations in either low-density lipoprotein receptor (LDLR), apolipoprotein B (APOB), or proprotein convertase subtilisin-kexin type 9 (PCSK9) are characterized by high low-density lipoprotein cholesterol levels and in some studies also high lipoprotein(a) (Lp(a)) levels were observed. The question remains whether this effect on Lp(a) levels is gene-dose-dependent in individuals with either 0, 1, or 2 LDLR or APOB mutations. OBJECTIVE We set out to study whether Lp(a) levels differ among bi-allelic ADH mutation carriers, and their relatives, in the Netherlands. METHODS Bi-allelic ADH mutation carriers were identified in the database of the national referral laboratory for DNA diagnostics of inherited dyslipidemias. Family members were invited by the index cases to participate. Clinical parameters and Lp(a) levels were measured in bi-allelic ADH mutation carriers and their heterozygous and unaffected relatives. RESULTS We included a total of 119 individuals; 34 bi-allelic ADH mutation carriers (20 homozygous/compound heterozygous LDLR mutation carriers (HoFH), 2 homozygous APOB mutation carriers (HoFDB), and 12 double heterozygotes for an LDLR and APOB mutation), 63 mono-allelic ADH mutation carriers (50 heterozygous LDLR [HeFH], 13 heterozygous APOB [HeFDB] mutation carriers), and 22 unaffected family members. Median Lp(a) levels in unaffected relatives, HeFH, and HoFH patients were 19.9 (11.1-41.5), 24.4 (5.9-70.6), and 47.3 (14.9-111.7) mg/dL, respectively (P = .150 for gene-dose dependency). Median Lp(a) levels in HeFDB and HoFDB patients were 50.3 (18.7-120.9) and 205.5 (no interquartile range calculated), respectively (P = .012 for gene-dose-dependency). Double heterozygous carriers of LDLR and APOB mutations had median Lp(a) levels of 27.0 (23.5-45.0), which did not significantly differ from HoFH and HoFDB patients (P = .730 and .340, respectively). CONCLUSION A (trend toward) increased plasma Lp(a) levels in homozygous ADH patients compared with both heterozygous ADH and unaffected relatives was observed. Whether increased Lp(a) levels in homozygous ADH patients add to the increased cardiovascular disease risk and whether this risk can be reduced by therapies that lower both low-density lipoprotein cholesterol and Lp(a) levels remains to be elucidated.
Collapse
Affiliation(s)
- Barbara Sjouke
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands.
| | - Reyhana Yahya
- Department of Internal Medicine, Erasmus Medical Centre, Erasmus University of Rotterdam, Rotterdam, The Netherlands
| | - Michael W T Tanck
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Amsterdam, The Netherlands
| | - Joep C Defesche
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Jacqueline de Graaf
- Division of Vascular Medicine, Department of Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Albert Wiegman
- Department of Pediatrics, Academic Medical Center, Amsterdam, The Netherlands
| | - John J P Kastelein
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Erasmus Medical Centre, Erasmus University of Rotterdam, Rotterdam, The Netherlands
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Jeanine E Roeters van Lennep
- Department of Internal Medicine, Erasmus Medical Centre, Erasmus University of Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
36
|
Reyes-Soffer G, Pavlyha M, Ngai C, Thomas T, Holleran S, Ramakrishnan R, Karmally W, Nandakumar R, Fontanez N, Obunike J, Marcovina SM, Lichtenstein AH, Matthan NR, Matta J, Maroccia M, Becue F, Poitiers F, Swanson B, Cowan L, Sasiela WJ, Surks HK, Ginsberg HN. Effects of PCSK9 Inhibition With Alirocumab on Lipoprotein Metabolism in Healthy Humans. Circulation 2016; 135:352-362. [PMID: 27986651 PMCID: PMC5262523 DOI: 10.1161/circulationaha.116.025253] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/07/2016] [Indexed: 12/02/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Alirocumab, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 (PCSK9), lowers plasma low-density lipoprotein (LDL) cholesterol and apolipoprotein B100 (apoB). Although studies in mice and cells have identified increased hepatic LDL receptors as the basis for LDL lowering by PCSK9 inhibitors, there have been no human studies characterizing the effects of PCSK9 inhibitors on lipoprotein metabolism. In particular, it is not known whether inhibition of PCSK9 has any effects on very low-density lipoprotein or intermediate-density lipoprotein (IDL) metabolism. Inhibition of PCSK9 also results in reductions of plasma lipoprotein (a) levels. The regulation of plasma Lp(a) levels, including the role of LDL receptors in the clearance of Lp(a), is poorly defined, and no mechanistic studies of the Lp(a) lowering by alirocumab in humans have been published to date. Methods: Eighteen (10 F, 8 mol/L) participants completed a placebo-controlled, 2-period study. They received 2 doses of placebo, 2 weeks apart, followed by 5 doses of 150 mg of alirocumab, 2 weeks apart. At the end of each period, fractional clearance rates (FCRs) and production rates (PRs) of apoB and apo(a) were determined. In 10 participants, postprandial triglycerides and apoB48 levels were measured. Results: Alirocumab reduced ultracentrifugally isolated LDL-C by 55.1%, LDL-apoB by 56.3%, and plasma Lp(a) by 18.7%. The fall in LDL-apoB was caused by an 80.4% increase in LDL-apoB FCR and a 23.9% reduction in LDL-apoB PR. The latter was due to a 46.1% increase in IDL-apoB FCR coupled with a 27.2% decrease in conversion of IDL to LDL. The FCR of apo(a) tended to increase (24.6%) without any change in apo(a) PR. Alirocumab had no effects on FCRs or PRs of very low-density lipoproteins-apoB and very low-density lipoproteins triglycerides or on postprandial plasma triglycerides or apoB48 concentrations. Conclusions: Alirocumab decreased LDL-C and LDL-apoB by increasing IDL- and LDL-apoB FCRs and decreasing LDL-apoB PR. These results are consistent with increases in LDL receptors available to clear IDL and LDL from blood during PCSK9 inhibition. The increase in apo(a) FCR during alirocumab treatment suggests that increased LDL receptors may also play a role in the reduction of plasma Lp(a). Clinical Trial Registration: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01959971.
Collapse
Affiliation(s)
- Gissette Reyes-Soffer
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.).
| | - Marianna Pavlyha
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Colleen Ngai
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Tiffany Thomas
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Stephen Holleran
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Rajasekhar Ramakrishnan
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Wahida Karmally
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Renu Nandakumar
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Nelson Fontanez
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Joseph Obunike
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Santica M Marcovina
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Alice H Lichtenstein
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Nirupa R Matthan
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - James Matta
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Magali Maroccia
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Frederic Becue
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Franck Poitiers
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Brian Swanson
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Lisa Cowan
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - William J Sasiela
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Howard K Surks
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.)
| | - Henry N Ginsberg
- From Columbia University College of Physicians and Surgeons, New York (G.R.-S., M.P., C.N., T.T., S.H., R.R., W.K., R.N., N.F., H.N.G.); The City University of New York (J.O.); Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.M.); Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L., N.R.M.); Sanofi, Bridgewater, NJ (J.M., B.S., L.C., H.K.S.); Umanis, Levallois-Perret, France (M.M.); Sanofi, Montpellier, France (F.B.); Sanofi, Paris, France (F.P.); and Regeneron Pharmaceuticals, Inc., Tarrytown, NY (W.J.S.).
| |
Collapse
|
37
|
Abstract
Lipoprotein(a) [Lp(a)] is a highly atherogenic lipoprotein that is under strong genetic control by the LPA gene locus. Genetic variants including a highly polymorphic copy number variation of the so called kringle IV repeats at this locus have a pronounced influence on Lp(a) concentrations. High concentrations of Lp(a) as well as genetic variants which are associated with high Lp(a) concentrations are both associated with cardiovascular disease which very strongly supports causality between Lp(a) concetrations and cardiovascular disease. This method of using a genetic variant that has a pronounced influence on a biomarker to support causality with an outcome is called Mendelian randomization approach and was applied for the first time two decades ago with data from Lp(a) and cardiovascular disease. This approach was also used to demonstrate a causal association between high Lp(a) concentrations and aortic valve stenosis, between low concentrations and type-2 diabetes mellitus and to exclude a causal association between Lp(a) concentrations and venous thrombosis. Considering the high frequency of these genetic variants in the population makes Lp(a) the strongest genetic risk factor for cardiovascular disease identified so far. Promising drugs that lower Lp(a) are on the horizon but their efficacy in terms of reducing clinical outcomes still has to be shown.
Collapse
|
38
|
Kostner KM, Kostner GM. Lipoprotein (a): a historical appraisal. J Lipid Res 2016; 58:1-14. [PMID: 27821413 DOI: 10.1194/jlr.r071571] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/01/2016] [Indexed: 11/20/2022] Open
Abstract
Initially, lipoprotein (a) [Lp(a)] was believed to be a genetic variant of lipoprotein (Lp)-B. Because its lipid moiety is almost identical to LDL, Lp(a) has been deliberately considered to be highly atherogenic. Lp(a) was detected in 1963 by Kare Berg, and individuals who were positive for this factor were called Lpa+ Lpa+ individuals were found more frequently in patients with coronary heart disease than in controls. After the introduction of quantitative methods for monitoring of Lp(a), it became apparent that Lp(a), in fact, is present in all individuals, yet to a greatly variable extent. The genetics of Lp(a) had been a mystery for a long time until Gerd Utermann discovered that apo(a) is expressed by a variety of alleles, giving rise to a unique size heterogeneity. This size heterogeneity, as well as countless mutations, is responsible for the great variability in plasma Lp(a) concentrations. Initially, we proposed to evaluate the risk of myocardial infarction at a cut-off for Lp(a) of 30-50 mg/dl, a value that still is adopted in numerous epidemiological studies. Due to new therapies that lower Lp(a) levels, there is renewed interest and still rising research activity in Lp(a). Despite all these activities, numerous gaps exist in our knowledge, especially as far as the function and metabolism of this fascinating Lp are concerned.
Collapse
Affiliation(s)
- Karam M Kostner
- Department of Cardiology, Mater Hospital and University of Queensland, Brisbane, 4101 Queensland, Australia
| | - Gert M Kostner
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, A-8010 Graz, Austria
| |
Collapse
|
39
|
Feinberg MW. No small task: therapeutic targeting of Lp(a) for cardiovascular disease. Lancet 2016; 388:2211-2212. [PMID: 27665227 DOI: 10.1016/s0140-6736(16)31329-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 07/29/2016] [Indexed: 11/26/2022]
Affiliation(s)
- Mark W Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
40
|
Pichardo-Almarza C, Diaz-Zuccarini V. From PK/PD to QSP: Understanding the Dynamic Effect of Cholesterol-Lowering Drugs on Atherosclerosis Progression and Stratified Medicine. Curr Pharm Des 2016; 22:6903-6910. [PMID: 27592718 PMCID: PMC5403958 DOI: 10.2174/1381612822666160905095402] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 08/29/2016] [Indexed: 01/18/2023]
Abstract
Current computational and mathematical tools are demonstrating the high value of using systems modeling approaches (e.g. Quantitative Systems Pharmacology) to understand the effect of a given compound on the biological and physiological mechanisms related to a specific disease. This review provides a short survey of the evolution of the mathematical approaches used to understand the effect of particular cholesterol-lowering drugs, from pharmaco-kinetic (PK) / pharmaco-dynamic (PD) models, through physiologically based pharmacokinetic models (PBPK) to QSP. These mathematical models introduce more mechanistic information related to the effect of these drugs on atherosclerosis progression and demonstrate how QSP could open new ways for stratified medicine in this field.
Collapse
Affiliation(s)
- Cesar Pichardo-Almarza
- UCL Mechanical Engineering, University College London, Roberts Building, Torrington Place, WC1E 7JE, London, United Kingdom
| | | |
Collapse
|
41
|
Tsimikas S. The re-emergence of lipoprotein(a) in a broader clinical arena. Prog Cardiovasc Dis 2016; 59:135-144. [PMID: 27497506 DOI: 10.1016/j.pcad.2016.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 07/24/2016] [Indexed: 01/13/2023]
Abstract
Lipoprotein(a) [Lp(a)] is a genetic, independent and likely causal risk factor for cardiovascular disease (CVD) and calcific aortic valve stenosis (CAVS). Lp(a) levels are primarily genetically determined and tend to fluctuate only mildly around a pre-determined level. In primary care settings, one Lp(a) measurement can reclassify up to 40% of patients in intermediate risk score categories. In secondary care settings, recent data from the JUPITER and AIM-HIGH trials demonstrate that elevated Lp(a) remains part of the "residual risk" despite achievement of low-density lipoprotein cholesterol levels <70 mg/dL. Recent reports suggest that statins can increase Lp(a) levels, potentially further contributing to this residual risk. Current therapies to lower Lp(a) are limited to niacin, mipomersen and proprotein convertase subtilisin kexin-type 9 inhibitors, but these drugs are limited by weak efficacy and not specifically approved for Lp(a) lowering. Emerging therapies to lower Lp(a) may shed new light into the potential clinical benefit of lowering Lp(a) in CVD and CAVS.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA.
| |
Collapse
|
42
|
Tavori H, Christian D, Minnier J, Plubell D, Shapiro MD, Yeang C, Giunzioni I, Croyal M, Duell PB, Lambert G, Tsimikas S, Fazio S. PCSK9 Association With Lipoprotein(a). Circ Res 2016; 119:29-35. [PMID: 27121620 DOI: 10.1161/circresaha.116.308811] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 04/26/2016] [Indexed: 12/12/2022]
Abstract
RATIONALE Lipoprotein(a) [Lp(a)] is a highly atherogenic low-density lipoprotein-like particle characterized by the presence of apoprotein(a) [apo(a)] bound to apolipoprotein B. Proprotein convertase subtilisin/kexin type 9 (PCSK9) selectively binds low-density lipoprotein; we hypothesized that it can also be associated with Lp(a) in plasma. OBJECTIVE Characterize the association of PCSK9 and Lp(a) in 39 subjects with high Lp(a) levels (range 39-320 mg/dL) and in transgenic mice expressing either human apo(a) only or human Lp(a) (via coexpression of human apo(a) and human apolipoprotein B). METHODS AND RESULTS We show that PCSK9 is physically associated with Lp(a) in vivo using 3 different approaches: (1) analysis of Lp(a) fractions isolated by ultracentrifugation; (2) immunoprecipitation of plasma using antibodies to PCSK9 and immunodetection of apo(a); (3) ELISA quantification of Lp(a)-associated PCSK9. Plasma PCSK9 levels correlated with Lp(a) levels, but not with the number of kringle IV-2 repeats. PCSK9 did not bind to apo(a) only, and the association of PCSK9 with Lp(a) was not affected by the loss of the apo(a) region responsible for binding oxidized phospholipids. Preferential association of PCSK9 with Lp(a) versus low-density lipoprotein (1.7-fold increase) was seen in subjects with high Lp(a) and normal low-density lipoprotein. Finally, Lp(a)-associated PCSK9 levels directly correlated with plasma Lp(a) levels but not with total plasma PCSK9 levels. CONCLUSIONS Our results show, for the first time, that plasma PCSK9 is found in association with Lp(a) particles in humans with high Lp(a) levels and in mice carrying human Lp(a). Lp(a)-bound PCSK9 may be pursued as a biomarker for cardiovascular risk.
Collapse
Affiliation(s)
- Hagai Tavori
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.).
| | - Devon Christian
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Jessica Minnier
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Deanna Plubell
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Michael D Shapiro
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Calvin Yeang
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Ilaria Giunzioni
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Mikael Croyal
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - P Barton Duell
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Gilles Lambert
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Sotirios Tsimikas
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.)
| | - Sergio Fazio
- From the Department of Medicine, Center for Preventive Cardiology, Knight Cardiovascular Institute, Portland, OR (H.T., D.C., D.P., M.D.S., I.G., P.B.D., S.F.); School of Public Health, Oregon Health and Science University, Portland (J.M.); Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California at San Diego, La Jolla (C.Y., S.T.); Inra UMR1280, Université de Nantes, CHU Hôtel-Dieu, Nantes, France (M.C., G.L.); Inserm UMR 1188, Sainte-Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint-Denis, France (G.L.); and CHU de la Réunion, Saint-Denis, France (G.L.).
| |
Collapse
|
43
|
Raal FJ, Giugliano RP, Sabatine MS, Koren MJ, Blom D, Seidah NG, Honarpour N, Lira A, Xue A, Chiruvolu P, Jackson S, Di M, Peach M, Somaratne R, Wasserman SM, Scott R, Stein EA. PCSK9 inhibition-mediated reduction in Lp(a) with evolocumab: an analysis of 10 clinical trials and the LDL receptor's role. J Lipid Res 2016; 57:1086-96. [PMID: 27102113 DOI: 10.1194/jlr.p065334] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Indexed: 12/12/2022] Open
Abstract
Lipoprotein (a) [Lp(a)] is independently associated with CVD risk. Evolocumab, a monoclonal antibody (mAb) to proprotein convertase subtilisin/kexin type 9 (PCSK9), decreases Lp(a). The potential mechanisms were assessed. A pooled analysis of Lp(a) and LDL cholesterol (LDL-C) in 3,278 patients from 10 clinical trials (eight phase 2/3; two extensions) was conducted. Within each parent study, biweekly and monthly doses of evolocumab statistically significantly reduced Lp(a) at week 12 versus control (P < 0.001 within each study); pooled median (quartile 1, quartile 3) percent reductions were 24.7% (40.0, 3.6) and 21.7% (39.9, 4.2), respectively. Reductions were maintained through week 52 of the open-label extension, and correlated with LDL-C reductions [with and without correction for Lp(a)-cholesterol] at both time points (P < 0.0001). The effect of LDL and LDL receptor (LDLR) availability on Lp(a) cell-association was measured in HepG2 cells: cell-associated LDL fluorescence was reversed by unlabeled LDL and Lp(a). Lp(a) cell-association was reduced by coincubation with LDL and PCSK9 and reversed by adding PCSK9 mAb. These studies support that reductions in Lp(a) with PCSK9 inhibition are partly due to increased LDLR-mediated uptake. In most situations, Lp(a) appears to compete poorly with LDL for LDLR binding and internalization, but when LDLR expression is increased with evolocumab, particularly in the setting of low circulating LDL, Lp(a) is reduced.
Collapse
Affiliation(s)
- Frederick J Raal
- Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Robert P Giugliano
- TIMI Study Group, Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA
| | - Marc S Sabatine
- TIMI Study Group, Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA
| | | | - Dirk Blom
- Division of Lipidology, University of Cape Town, Cape Town, South Africa
| | | | | | | | | | | | | | - Mei Di
- Amgen Inc., San Francisco, CA
| | | | | | | | | | - Evan A Stein
- Metabolic and Atherosclerosis Research Center, Cincinnati, OH
| |
Collapse
|
44
|
Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of lipoprotein (a). J Lipid Res 2016; 57:1339-59. [PMID: 27074913 DOI: 10.1194/jlr.r067314] [Citation(s) in RCA: 333] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Indexed: 12/29/2022] Open
Abstract
Lipoprotein (a) [Lp(a)] has attracted the interest of researchers and physicians due to its intriguing properties, including an intragenic multiallelic copy number variation in the LPA gene and the strong association with coronary heart disease (CHD). This review summarizes present knowledge of the structure, function, and genetics of Lp(a) with emphasis on the molecular and population genetics of the Lp(a)/LPA trait, as well as aspects of genetic epidemiology. It highlights the role of genetics in establishing Lp(a) as a risk factor for CHD, but also discusses uncertainties, controversies, and lack of knowledge on several aspects of the genetic Lp(a) trait, not least its function.
Collapse
Affiliation(s)
- Konrad Schmidt
- Divisions of Human Genetics Medical University of Innsbruck, Innsbruck, Austria Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Asma Noureen
- Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gerd Utermann
- Divisions of Human Genetics Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
45
|
Park SH, Rha SW, Choi BG, Park JY, Jeon U, Seo HS, Kim EJ, Na JO, Choi CU, Kim JW, Lim HE, Park CG, Oh DJ. Impact of high lipoprotein(a) levels on in-stent restenosis and long-term clinical outcomes of angina pectoris patients undergoing percutaneous coronary intervention with drug-eluting stents in Asian population. Clin Exp Pharmacol Physiol 2016; 42:588-95. [PMID: 25865336 DOI: 10.1111/1440-1681.12396] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 02/22/2015] [Accepted: 03/28/2015] [Indexed: 11/29/2022]
Abstract
Lipoprotein(a) (Lp(a)) is known to be associated with cardiovascular complications and atherothrombotic properties in general populations. However, it has not been examined whether Lp(a) levels are able to predict adverse cardiovascular outcomes in patients undergoing percutaneous coronary intervention (PCI) with drug-eluting stents (DES). A total of 595 consecutive patients with angina pectoris who underwent elective PCI with DES were enrolled from 2004 to 2010. The patients were divided into two groups according to the levels of Lp(a): Lp(a) < 50 mg/dL (n = 485 patients), and Lp(a) ≥ 50 mg/dL (n = 111 patients). The 6-9-month angiographic outcomes and 3-year cumulative major clinical outcomes were compared between the two groups. Binary restenosis occurred in 26 of 133 lesions (19.8%) in the high Lp(a) group and 43 of 550 lesions (7.9%) in the low Lp(a) group (P = 0.001). In multivariate analysis, the reference vessel diameter, low density lipoprotein cholesterol, total lesion length, and Lp(a) ≥ 50 mg/dL were predictors of binary restenosis. In the Cox proportional hazards regression analysis, Lp(a) > 50 mg/dL was significantly associated with the 3-year adverse clinical outcomes including any myocardial infarction, revascularization (target lesion revascularization (TLR) and target vessel revascularization (TVR)), TLR-major adverse cardiac events (MACEs), TVR-MACE, and All-MACEs. In our study, high Lp(a) level ≥ 50 mg/dL in angina pectoris patients undergoing elective PCI with DES was significantly associated with binary restenosis and 3-year adverse clinical outcomes in an Asian population.
Collapse
Affiliation(s)
- Sang-Ho Park
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan, Korea
| | - Seung-Woon Rha
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Korea
| | - Byoung-Geol Choi
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Korea
| | - Ji-Young Park
- Division of Cardiology, Department of Internal Medicine, Eulji Medical University, Seoul, Korea
| | - Ung Jeon
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan, Korea
| | - Hong-Seog Seo
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Korea
| | - Eung-Ju Kim
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Korea
| | - Jin-Oh Na
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Korea
| | - Cheol-Ung Choi
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Korea
| | - Jin-Won Kim
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Korea
| | - Hong-Euy Lim
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Korea
| | - Chang-Gyu Park
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Korea
| | - Dong-Joo Oh
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Korea
| |
Collapse
|
46
|
Kollerits B, Drechsler C, Krane V, Lamina C, März W, Dieplinger H, Ritz E, Wanner C, Kronenberg F. Lipoprotein(a) concentrations, apolipoprotein(a) isoforms and clinical endpoints in haemodialysis patients with type 2 diabetes mellitus: results from the 4D Study. Nephrol Dial Transplant 2016; 31:1901-1908. [DOI: 10.1093/ndt/gfv428] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/23/2015] [Indexed: 11/13/2022] Open
|
47
|
Thakkar S, Wang X, Khaidakov M, Dai Y, Gokulan K, Mehta JL, Varughese KI. Structure-based Design Targeted at LOX-1, a Receptor for Oxidized Low-Density Lipoprotein. Sci Rep 2015; 5:16740. [PMID: 26578342 PMCID: PMC4649741 DOI: 10.1038/srep16740] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 10/19/2015] [Indexed: 01/16/2023] Open
Abstract
Atherosclerosis related cardiovascular diseases continue to be the primary cause of mortality in developed countries. The elevated level of low density lipoprotein (LDL) is generally considered to be the driver of atherosclerosis, but recent years have seen a shift in this perception in that the vascular plaque buildup is mainly caused by oxidized LDL (ox-LDL) rather than native-LDL. The scavenger receptor LOX-1 found in endothelial cells binds and internalizes ox-LDL which leads to the initiation of plaque formation in arteries. Using virtual screening techniques, we identified a few potential small molecule inhibitors of LOX-1 and tested their inhibitory potential using differential scanning fluorimetry and various cellular assays. Two of these molecules significantly reduced the uptake of ox-LDL by human endothelial cells, LOX-1 transcription and the activation of ERK1/2 and p38 MAPKs in human endothelial cells. In addition, these molecules suppressed ox-LDL-induced VCAM-1 expression and monocyte adhesion onto human endothelial cells demonstrating their therapeutic potential.
Collapse
Affiliation(s)
- Shraddha Thakkar
- Department of Physiology and Biophysics College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Xianwei Wang
- Department of Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Magomed Khaidakov
- Department of Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Yao Dai
- Department of Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kuppan Gokulan
- Department of Physiology and Biophysics College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jawahar L Mehta
- Department of Physiology and Biophysics College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kottayil I Varughese
- Department of Physiology and Biophysics College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
48
|
Affiliation(s)
- Maximillian A Rogers
- From Center for Interdisciplinary Cardiovascular Sciences (M.A.R., E.A.) and Center for Excellence in Vascular Biology (E.A.), Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- From Center for Interdisciplinary Cardiovascular Sciences (M.A.R., E.A.) and Center for Excellence in Vascular Biology (E.A.), Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
49
|
Yeang C, Witztum JL, Tsimikas S. 'LDL-C' = LDL-C + Lp(a)-C: implications of achieved ultra-low LDL-C levels in the proprotein convertase subtilisin/kexin type 9 era of potent LDL-C lowering. Curr Opin Lipidol 2015; 26:169-78. [PMID: 25943842 DOI: 10.1097/mol.0000000000000171] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The measurement that is termed 'LDL-cholesterol' (LDL-C) includes the cholesterol content of lipoprotein(a) [Lp(a)-C], which can contribute approximately 30-45% to measured LDL-C levels as a percentage of its mass. We review the implications of achieved very low LDL-C levels in patients treated with potent LDL-C-lowering agents in the context of varying Lp(a) levels. RECENT FINDINGS Combination therapy with statins, ezetimibe, and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors can lower LDL-C to unprecedentedly low levels. Recent PCSK9 trials have shown that routine achievement of mean LDL-C less than 50 mg/dl is feasible, along with the modest reductions in Lp(a). Many patients will achieve LDL-C less than 25 mg/dl with concomitantly elevated Lp(a) levels that contribute substantially to the measured 'LDL-C'. Therefore, it is possible that some of these patients may have little to no circulating LDL-C. SUMMARY As the new era of ultralow LDL-C levels ensues, it is imperative to understand the contribution of Lp(a)-C to measured LDL-C and the consequences of achieving ultralow or potentially absent LDL-C in the setting of elevated Lp(a) levels and possibly free apo(a). We review this concept and suggest avenues of research, including analyses of existing datasets in current clinical trials and new research studies, to understand its pathophysiological and clinical significance.
Collapse
Affiliation(s)
- Calvin Yeang
- University of California San Diego, La Jolla, California, USA
| | | | | |
Collapse
|
50
|
Palazhy S, Kamath P, Vasudevan DM. Elevated oxidative stress among coronary artery disease patients on statin therapy: A cross sectional study. Indian Heart J 2015; 67:227-32. [PMID: 26138179 PMCID: PMC4495669 DOI: 10.1016/j.ihj.2015.03.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 02/21/2015] [Accepted: 03/29/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Statins are a major group of drugs that reduces LDL-C levels, which are proven to have other beneficial effects such as preventing coronary events. The objective of this study was to evaluate oxidative stress and select novel coronary artery disease risk factors among coronary artery disease patients on statins. METHODS In this observational, cross-sectional study, we compared total cholesterol, triglycerides, HDL-cholesterol, LDL-cholesterol, apolipoprotein B, lipoprotein (a), homocysteine, reduced glutathione, glutathione peroxidase, superoxide dismutase, ascorbic acid, malondialdehyde and oxidized LDL among male coronary artery disease patients on statin therapy (group 2, n = 151) with sex-matched, diabetic patients (group 3, n = 80) as well as healthy controls (group 1, n = 84). RESULTS Total cholesterol, triglycerides, HDL-cholesterol and LDL-cholesterol were significantly lower among subjects of group 2 compared to other two groups. The novel risk factors studied did not differ significantly between groups, except for a higher homocysteine level among group 2 subjects compared to the other two groups. Elevated oxidative stress, indicated by lower reduced glutathione, glutathione peroxidase, and ascorbic acid as well as higher malondialdehyde and oxidized LDL was observed among group 2 subjects. Triglycerides, HDL-cholesterol, ascorbic acid and malondialdehyde were found to be independent predictors for coronary artery disease among this study population. CONCLUSIONS Though coronary artery disease subjects had healthy lipid profile, oxidative stress, a recognized risk factor for coronary events, was still elevated among this patient group. Novel risk factors were not found to be major predictors for coronary artery disease among the study subjects.
Collapse
Affiliation(s)
- Sabitha Palazhy
- Department of Biochemistry, Amrita School of Medicine, Kochi 682041, India.
| | - Prakash Kamath
- Department of Cardiology, Amrita Institute of Medical Sciences, Kochi 682041, India
| | | |
Collapse
|