151
|
Albuquerque J, Medeiros AM, Alves AC, Bourbon M, Antunes M. Performance comparison of different classification algorithms applied to the diagnosis of familial hypercholesterolemia in paediatric subjects. Sci Rep 2022; 12:1164. [PMID: 35064162 PMCID: PMC8782861 DOI: 10.1038/s41598-022-05063-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/28/2021] [Indexed: 12/03/2022] Open
Abstract
Familial Hypercholesterolemia (FH) is an inherited disorder of lipid metabolism, characterized by increased low density lipoprotein cholesterol (LDLc) levels. The main purpose of the current work was to explore alternative classification methods to traditional clinical criteria for FH diagnosis, based on several biochemical and biological indicators. Logistic regression (LR), decision tree (DT), random forest (RF) and naive Bayes (NB) algorithms were developed for this purpose, and thresholds were optimized by maximization of Youden index (YI). All models presented similar accuracy (Acc), specificity (Spec) and positive predictive values (PPV). Sensitivity (Sens) and G-mean values were significantly higher in LR and RF models, compared to the DT. When compared to Simon Broome (SB) biochemical criteria for FH diagnosis, all models presented significantly higher Acc, Spec and G-mean values (p < 0.01), and lower negative predictive value (NPV, p < 0.05). Moreover, LR and RF models presented comparable Sens values. Adjustment of the cut-off point by maximizing YI significantly increased Sens values, with no significant loss in Acc. The obtained results suggest such classification algorithms can be a viable alternative to be used as a widespread screening method. An online application has been developed to assess the performance of the LR model in a wider population.
Collapse
Affiliation(s)
- João Albuquerque
- Departamento de Biomedicina, Unidade de Bioquímica, Faculdade de Medicina, Universidade do Porto, 4200-319, Porto, Portugal.
- Centro de Estatística e Aplicações, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal.
| | - Ana Margarida Medeiros
- Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, 1649-016, Lisboa, Portugal
- Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal
| | - Ana Catarina Alves
- Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, 1649-016, Lisboa, Portugal
- Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal
| | - Mafalda Bourbon
- Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, 1649-016, Lisboa, Portugal
- Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal
| | - Marília Antunes
- Centro de Estatística e Aplicações, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal
- Departamento de Estatística e Investigação Operacional, Faculdade de Ciências, Universidade de Lisboa, 1749-016, Lisboa, Portugal
| |
Collapse
|
152
|
Strandkjær N, Hansen MK, Nielsen ST, Frikke-Schmidt R, Tybjærg-Hansen A, Nordestgaard BG, Tabor A, Bundgaard H, Iversen K, Kamstrup PR. Lipoprotein(a) Levels at Birth and in Early Childhood: The COMPARE Study. J Clin Endocrinol Metab 2022; 107:324-335. [PMID: 34618900 DOI: 10.1210/clinem/dgab734] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND OBJECTIVE High lipoprotein(a) is a genetically determined causal risk factor for cardiovascular disease, and 20% of the adult population has high levels (ie, >42 mg/dL, >88 nmol/L). We investigated whether early life lipoprotein(a) levels measured in cord blood may serve as a proxy for neonatal venous blood levels, whether lipoprotein(a) birth levels (ie, cord or venous) predict levels later in life, and whether early life and parental levels correlate. METHODS The Compare study is a prospective cohort study of newborns (N = 450) from Copenhagen, Denmark, including blood sampling of parents. Plasma lipoprotein(a) was measured in cord blood (N = 402), neonatal venous blood (N = 356), and at 2 (N = 320) and 15 months follow-up (N = 148) of infants, and in parents (N = 705). RESULTS Mean lipoprotein(a) levels were 2.2 (95% CI, 1.9-2.5), 2.4 (2.0-2.7), 4.1 (3.4-4.9), and 14.6 (11.4-17.9) mg/dL in cord, neonatal venous, and 2- and 15-month venous samples, respectively. Lipoprotein(a) levels in cord blood correlated strongly with neonatal venous blood levels (R2 = 0.95, P < 0.001) and neonatal levels correlated moderately with 2- and 15-month levels (R2 = 0.68 and 0.67, both P < 0.001). Birth levels ≥ 90th percentile predicted lipoprotein(a) > 42 mg/dL at 15 months with positive predictive values of 89% and 85% for neonatal venous and cord blood. Neonatal and infant levels correlated weakly with parental levels, most pronounced at 15 months (R2 = 0.22, P < 0.001). CONCLUSIONS Lipoprotein(a) levels are low in early life, cord blood may serve as a proxy for neonatal venous blood, and birth levels ≥ 90th percentile can identify newborns at risk of developing high levels.
Collapse
Affiliation(s)
- Nina Strandkjær
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Malene Kongsgaard Hansen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Sofie Taageby Nielsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen Ø, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen Ø, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen Ø, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen Ø, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen Ø, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen Ø, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Ann Tabor
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen Ø, Denmark
- Department of Obstetrics, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen Ø, Denmark
| | - Henning Bundgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen Ø, Denmark
- Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen Ø, Denmark
| | - Kasper Iversen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen Ø, Denmark
| | - Pia R Kamstrup
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| |
Collapse
|
153
|
Metzner T, Leitner DR, Mellitzer K, Beck A, Sourij H, Stojakovic T, Reishofer G, März W, Landmesser U, Scharnagl H, Toplak H, Silbernagel G. Effects of Alirocumab on Triglyceride Metabolism: A Fat-Tolerance Test and Nuclear Magnetic Resonance Spectroscopy Study. Biomedicines 2022; 10:biomedicines10010193. [PMID: 35052871 PMCID: PMC8774139 DOI: 10.3390/biomedicines10010193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 01/06/2023] Open
Abstract
Background: PCSK9 antibodies strongly reduce LDL cholesterol. The effects of PCSK9 antibodies on triglyceride metabolism are less pronounced. The present study aimed to investigate in detail the effects of alirocumab on triglycerides, triglyceride-rich lipoproteins, and lipase regulators. Methods: A total of 24 patients with an indication for treatment with PCSK9 antibodies were recruited. There were two visits at the study site: the first before initiation of treatment with alirocumab and the second after 10 weeks of treatment. Fat-tolerance tests, nuclear magnetic resonance spectroscopy, and enzyme-linked immunosorbent assays were performed to analyze lipid metabolism. Results: A total of 21 participants underwent the first and second investigation. Among these, two participants only received alirocumab twice and 19 patients completed the trial per protocol. All of them had atherosclerotic vascular disease. There was no significant effect of alirocumab treatment on fasting triglycerides, post-prandial triglycerides, or lipoprotein-lipase regulating proteins. Total, large, and small LDL particle concentrations decreased, while the HDL particle concentration increased (all p < 0.001). Mean total circulating PCSK9 markedly increased in response to alirocumab treatment (p < 0.001). Whereas PCSK9 increased more than three-fold in all 19 compliant patients, it remained unchanged in those two patients with two injections only. Conclusion: Significant effects of alirocumab on triglyceride metabolism were not detectable in the ALIROCKS trial. The total circulating PCSK9 concentration might be a useful biomarker to differentiate non-adherence from non-response to PCSK9 antibodies.
Collapse
Affiliation(s)
- Thomas Metzner
- Department of Internal Medicine, Division of Angiology, Medical University of Graz, 8036 Graz, Austria
- Department of Medical Affairs, Sanofi-Aventis GmbH, 1100 Vienna, Austria
| | - Deborah R Leitner
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria
| | - Karin Mellitzer
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria
| | - Andrea Beck
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria
| | - Harald Sourij
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, 8036 Graz, Austria
| | - Gernot Reishofer
- Department of Radiology, Clinical Division of Neuroradiology, Vascular and Interventional Radiology, Medical University of Graz, 8036 Graz, Austria
| | - Winfried März
- Department of Internal Medicine 5 (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Mannheim Medical Faculty, University of Heidelberg, 68167 Mannheim, Germany
- Synlab Academy, Synlab Holding Germany GmbH, 86156 Augsburg, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria
| | - Ulf Landmesser
- German Center for Cardiovascular Research (DZHK)-Partner Site Berlin, Department of Cardiology, Berlin Institute of Health, Charité University Medicine Berlin, 12200 Berlin, Germany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria
| | - Hermann Toplak
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria
| | - Günther Silbernagel
- Department of Internal Medicine, Division of Angiology, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
154
|
Yao Y, Liu J, Wang B, Zhou Z, Lu X, Huang Z, Deng J, Yang Y, Tan N, Chen S, Chen J, Liu Y. Baseline Low-Density-Lipoprotein Cholesterol Modifies the Risk of All-Cause Death Associated With Elevated Lipoprotein(a) in Coronary Artery Disease Patients. Front Cardiovasc Med 2022; 8:817442. [PMID: 35097030 PMCID: PMC8792964 DOI: 10.3389/fcvm.2021.817442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022] Open
Abstract
Background: The prognostic value of elevated lipoprotein(a) [Lp(a)] in coronary artery disease (CAD) patients is inconsistent in previous studies, and whether such value changes at different low-density-lipoprotein cholesterol (LDL-C) levels is unclear. Methods and Findings: CAD patients treated with statin therapy from January 2007 to December 2018 in the Guangdong Provincial People's Hospital (NCT04407936) were consecutively enrolled. Individuals were categorized according to the baseline LDL-C at cut-off of 70 and 100 mg/dL. The primary outcome was 5-year all-cause death. Multivariate Cox proportional models and penalized spline analyses were used to evaluate the association between Lp(a) and all-cause mortality. Among 30,908 patients, the mean age was 63.1 ± 10.7 years, and 76.7% were men. A total of 2,383 (7.7%) patients died at 5-year follow-up. Compared with Lp(a) <50 mg/dL, Lp(a) ≥ 50 mg/dL predicted higher all-cause mortality (multivariable adjusted HR = 1.19, 95% CI 1.07-1.31) in the total cohort. However, when analyzed within each LDL-C category, there was no significant association between Lp(a) ≥ 50 mg/dL and higher all-cause mortality unless the baseline LDL-C was ≥ 100 mg/dL (HR = 1.19, 95% CI 1.04-1.36). The results from penalized spline analyses were robust. Conclusions: In statin-treated CAD patients, elevated Lp(a) was associated with increased risks of all-cause death, and such an association was modified by the baseline LDL-C levels. Patients with Lp(a) ≥ 50 mg/dL had higher long-term risks of all-cause death compared with those with Lp(a) <50 mg/dL only when their baseline LDL-C was ≥ 100 mg/dL.
Collapse
Affiliation(s)
- Younan Yao
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jin Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Bo Wang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ziyou Zhou
- School of Medicine, Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou, China
| | - Xiaozhao Lu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhidong Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jingru Deng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yongquan Yang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ning Tan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shiqun Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiyan Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yong Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
155
|
Ye CY, Xin JR, Li Z, Yin XY, Guo SL, Li JM, Zhao TY, Wang L, Yang L. ALDH2, ADCY3 and BCMO1 polymorphisms and lifestyle-induced traits are jointly associated with CAD risk in Chinese Han people. Gene 2022; 807:145948. [PMID: 34481002 DOI: 10.1016/j.gene.2021.145948] [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: 05/06/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUNDS To investigate associations of genetic and environmental factors with coronary artery disease (CAD), we collected medical reports, lifestyle details, and blood samples of 2113 individuals, and then used the polymerase chain reaction (PCR)-ligase detection reaction (LDR) to genotype the targeted 102 SNPs. METHODS We adopted elastic net algorithm to build an association model that considered simultaneously genetic and lifestyle/clinical factors associated with CAD in Chinese Han population. RESULTS In this study, we developed an all covariates-based model to explain the risk of CAD, which incorporated 8 lifestyle/clinical factors and a gene-score variable calculated from 3 significant SNPs (rs671, rs6751537 and rs11641677), attaining an area under the curve (AUC) value of 0.71. It was found that, in terms of genetic variants, the AA genotype of rs671 in the additive (adjusted odds ratio (OR) = 2.51, p = 0.008) and recessive (adjusted OR = 2.12, p = 0.021) models, the GG genotype of rs6751537 in the additive (adjusted OR = 3.36, p = 0.001) and recessive (adjusted OR = 3.47, p = 0.001) models were associated with increased risk of CAD, while GG genotype of rs11641677 in additive model (adjusted OR = 0.39, p = 0.044) was associated with decreased risk of CAD. In terms of lifestyle/clinical factors, the history of hypertension (unadjusted OR = 2.37, p < 0.001) and dyslipidemia (unadjusted OR = 1.82, p = 0.007), age (unadjusted OR = 1.07, p < 0.001) and waist circumference (unadjusted OR = 1.02, p = 0.05) would significantly increase the risk of CAD, while height (unadjusted OR = 0.97, p = 0.006) and regular intake of chicken (unadjusted OR = 0.78, p = 0.008) reduced the risk of CAD. A significantinteraction was foundbetween rs671 and dyslipidemia (the relative excess risk due to interaction (RERI) = 3.36, p = 0.05). CONCLUSION In this study, we constructed an association model and identified a set of SNPs and lifestyle/clinical risk factors of CAD in Chinese Han population. By considering both genetic and non-genetic risk factors, the built model may provide implications for CAD pathogenesis and clues for screening tool development in Chinese Han population.
Collapse
Affiliation(s)
- Cheng-Yin Ye
- School of Public Health, Hangzhou Normal University, Hangzhou 310000, China.
| | - Jia-Rui Xin
- School of Public Health, Hangzhou Normal University, Hangzhou 310000, China.
| | - Zheng Li
- Wu Yun Shan Hospital, Hangzhou 31000, China.
| | - Xiao-Yu Yin
- School of Public Health, Hangzhou Normal University, Hangzhou 310000, China.
| | - Shu-Li Guo
- School of Public Health, Hangzhou Normal University, Hangzhou 310000, China.
| | - Jin-Mei Li
- School of Public Health, Hangzhou Normal University, Hangzhou 310000, China.
| | - Tian-Yu Zhao
- School of Public Health, Hangzhou Normal University, Hangzhou 310000, China
| | - Li Wang
- School of Public Health, Hangzhou Normal University, Hangzhou 310000, China.
| | - Lei Yang
- School of Public Health, Hangzhou Normal University, Hangzhou 310000, China.
| |
Collapse
|
156
|
de Carvalho JF, Rodrigues CEM. Primary Antiphospholipid Syndrome with and Without Acute Myocardial Infarction/Angina: A Cross-Sectional Study. Rheumatol Ther 2022; 9:581-588. [PMID: 35000119 PMCID: PMC8964883 DOI: 10.1007/s40744-021-00419-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/14/2021] [Indexed: 11/25/2022] Open
Abstract
Introduction Acute myocardial infarct/angina (AMI-A) is a possible complication in primary antiphospholipid syndrome (pAPS) patients. This study compares data obtained from pAPS patients with and without AMI-A. Methods This cross-sectional study of 66 (85.2% female) pAPS patients (Sidney criteria). Demographics, clinical data, medication use, and antiphospholipid antibodies were evaluated. Patients were divided into two groups: pAPS with AMI-A and pAPS without AMI-A. Results Sixty-six patients with primary APS (six with AMI-A and 60 without AMI-A) were selected. They were similar for demographics, disease duration, and anthropometrics (p > 0.05). Patients with AMI-A compared to those patients without AMI-A had more frequently dyslipidemia (66 vs. 28%, p = 0.05), systemic hypertension (83 vs. 37%, p = 0.02), and increased levels of lipoprotein (a) (116 ± 67 vs. 36 ± 35 mg/dl, p = 0.0002). Interesting, current physical activity (66.7 vs. 23%, p = 0.04) was more seen in the first group when compared to the second one. Patients with AMI-A used more statins (66 vs. 22%, p = 0.017) and acetylsalicylic (100 vs. 28%, p = 0.05). Higher median levels of IgM anticardiolipin antibodies [70 (0–120) vs. 9 (0–120), p = 0.03] were observed in the first group. Conclusions pAPS patients and AMI-A have distinct clinical and laboratory spectra from those without AMI-A. It is characterized by dyslipidemia and hypertension, hyper lipoprotein(a), and a lower IgM anticardiolipin frequency.
Collapse
Affiliation(s)
- Jozélio Freire de Carvalho
- Institute for Health Sciences From Federal University of Bahia, Rua das Violetas, 42, ap. 502, Pituba, Salvador, BA, Brazil.
| | - Carlos Ewerton Maia Rodrigues
- Medical Sciences, Medical School, University of Fortaleza (Unifor), Fortaleza, Brazil
- Department of Internal Medicine, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
157
|
Di Maio S, Lamina C, Coassin S, Forer L, Würzner R, Schönherr S, Kronenberg F. Lipoprotein(a) and SARS-CoV-2 infections: Susceptibility to infections, ischemic heart disease and thromboembolic events. J Intern Med 2022; 291:101-107. [PMID: 34096654 PMCID: PMC8242884 DOI: 10.1111/joim.13338] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Comorbidities including ischemic heart disease (IHD) worsen outcomes after SARS-CoV-2 infections. High lipoprotein(a) [Lp(a)] concentrations are a strong risk factor for IHD and possibly for thromboembolic events. We therefore evaluated whether SARS-CoV-2 infections modify the risk of high Lp(a) concentrations for IHD or thromboembolic events during the first 8.5 months follow-up of the pandemic. METHOD Cohort study using data from the UK Biobank during the SARS-CoV-2 pandemic. Baseline Lp(a) was compared between SARS-CoV-2 positive patients and the population controls. RESULTS SARS-CoV-2 positive patients had Lp(a) concentrations similar to the population controls. The risk for IHD increased with higher Lp(a) concentrations in both, the population controls (n = 435,104) and SARS-CoV-2 positive patients (n = 6937). The causality of the findings was supported by a genetic risk score for Lp(a). A SARS-CoV-2 infection modified the association with a steeper increase in risk for infected patients (interaction p-value = 0.03). Although SARS-CoV-2 positive patients had a five-times higher frequency of thromboembolic events compared to the population controls (1.53% vs. 0.31%), the risk was not influenced by Lp(a). CONCLUSIONS SARS-CoV-2 infections enforce the association between high Lp(a) and IHD but the risk for thromboembolic events is not influenced by Lp(a).
Collapse
Affiliation(s)
- Silvia Di Maio
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Coassin
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Forer
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Reinhard Würzner
- Department of Hygiene, Microbiology and Public Health, Institute of Hygiene & Medical Microbiology, Medical University Innsbruck, Innsbruck, Austria
| | - Sebastian Schönherr
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
158
|
Yoshida H, Hirowatari Y, Ogura M, Harada-Shiba M. Current concept and residual issues of lipoprotein(a) determination for a cardiovascular risk factor. Eur J Clin Invest 2022; 52:e13700. [PMID: 34747007 DOI: 10.1111/eci.13700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Hiroshi Yoshida
- Department of Laboratory Medicine, The Jikei University Kashiwa Hospital, Chiba, Japan.,Internal Medicine of Metabolism and Nutrition, The Jikei University Graduate School of Medicine, Minato city, Japan
| | - Yuji Hirowatari
- Department of Health Science, Saitama Prefectural University, Saitama, Japan
| | - Masatsune Ogura
- Department of General Medical Science, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| |
Collapse
|
159
|
Jiang X, Xu J, Hao X, Xue J, Li K, Jin A, Lin J, Meng X, Zheng L, Wang Y. Elevated lipoprotein(a) and lipoprotein-associated phospholipase A 2 are associated with unfavorable functional outcomes in patients with ischemic stroke. J Neuroinflammation 2021; 18:307. [PMID: 34963487 PMCID: PMC8715597 DOI: 10.1186/s12974-021-02359-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
Background The association of lipoprotein(a) [Lp(a)] and stroke functional outcomes was conflicting. The aim of the study was to clarify whether high Lp(a) is associated with unfavorable functional outcomes in patients with ischemic stroke. Methods A total of 9709 individuals from the third China National Stroke Registry cohort were recruited. Plasma level of Lp(a) at admission was measured with enzyme-linked immunosorbent assay. The cut-off was set at the median for Lp(a). Functional outcome was assessed using the modified Rankin scale (mRS) at 3 months and 1 year after ischemic stroke. The association between Lp(a) and functional outcomes was evaluated using a logistic regression model. Results The median age was 63.0 years, and 31.1% participants were women. Patients in higher Lp(a) group had higher incidences of unfavorable functional outcomes at 3 months. In logistic regression model, elevated Lp(a) levels were associated with unfavorable functional outcomes at 3 months (Q4 vs. Q1: odds ratio 1.33, 95% confidence interval 1.11–1.61). Subgroup analysis showed that in the lower Lp-PLA2 group, Lp(a) level was not associated with functional outcomes, but in the higher Lp-PLA2 group, Lp(a) level was significantly associated with functional outcomes. After grouped by different levels of Lp(a) and Lp-PLA2, the Lp(a) high/ Lp-PLA2 high group showed the highest incidence of unfavorable functional outcomes at 3 months and 1 year. Conclusions Elevated Lp(a) level is associated with unfavorable functional outcomes in patients with ischemic stroke. The increment in both Lp(a) and Lp-PLA2 are associated with unfavorable functional outcomes at 3 months and 1 year after ischemic stroke. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02359-w.
Collapse
Affiliation(s)
- Xue Jiang
- Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Jie Xu
- Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Xiwa Hao
- Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.,Department of Neurology, Baotou Center Hospital, Inner Mongolia, China
| | - Jing Xue
- Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Ke Li
- Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Aoming Jin
- Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Jinxi Lin
- Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Xia Meng
- Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Lemin Zheng
- Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China. .,China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China. .,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China. .,The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, No. 38 Xueyuan Road, Haidian District, Beijing, 100871, China.
| | - Yongjun Wang
- Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China. .,China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing, China. .,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
160
|
Lipoprotein(a) is associated with coronary atheroma progression: analysis from a serial coronary computed tomography angiography study. J Geriatr Cardiol 2021; 18:996-1007. [PMID: 35136395 PMCID: PMC8782765 DOI: 10.11909/j.issn.1671-5411.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] has been closely related to coronary atherosclerosis and might affect perivascular inflammation due to its proinflammatory properties. However, there are limited data about Lp(a) and related perivascular inflammation on coronary atheroma progression. Therefore, this study aimed to investigate the associations between Lp(a) and the perivascular fat attenuation index (FAI) with coronary atheroma progression detected by coronary computed tomography angiography (CCTA). METHODS Patients who underwent serial CCTA examinations without a history of revascularization and with available data for Lp(a) within one month before or after baseline and follow-up CCTA imaging scans were considered to be included. CCTA quantitative analyses were performed to obtain the total plaque volume (TPV) and the perivascular FAI. Coronary plaque progression (PP) was defined as a ≥ 10% increase in the change of the TPV at the patient level or the presence of new-onset coronary atheroma lesions. The associations between Lp(a) or the perivascular FAI with PP were examined by multivariate logistic regression. RESULTS A total of 116 patients were ultimately enrolled in the present study with a mean CCTA interscan interval of 30.80 ± 13.50 months. Among the 116 patients (mean age: 53.49 ± 10.21 years, males: 83.6%), 32 patients presented PP during the follow-up interval. Lp(a) levels were significantly higher among PP patients than those among non-PP patients at both baseline [15.80 (9.09-33.60) mg/dLvs. 10.50 (4.75-19.71) mg/dL,P = 0.029] and follow-up [20.60 (10.45-34.55) mg/dLvs. 8.77 (5.00-18.78) mg/dL,P = 0.004]. However, there were no differences in the perivascular FAI between PP group and non-PP group at either baseline or follow-up. Multivariate logistic regression analysis showed that elevated baseline Lp(a) level (OR = 1.031, 95% CI: 1.005-1.058,P = 0.019) was an independent risk factor for PP after adjustment for other conventional variables. CONCLUSIONS Lp(a) was independently associated with coronary atheroma progression beyond low-density lipoprotein cholesterol and other conventional risk factors. Further studies are warranted to identify the inflammation effect exhibited as the perivascular FAI on coronary atheroma progression.
Collapse
|
161
|
Abstract
PURPOSE OF REVIEW To provide an overview of the associations between elevated blood pressure and lipoprotein (a) and possible causal links, as well as data on the prevalence of elevated lipoprotein (a) in a cohort of hypertensive patients. RECENT FINDINGS Elevated lipoprotein (a) is now considered to be an independent and causal risk factor for atherosclerotic cardiovascular disease and calcific aortic valve disease. Despite this, there are limited data demonstrating an association between elevated lipoprotein (a) and hypertension. Further, there is limited mechanistic data linking lipoprotein (a) and hypertension through either renal impairment or direct effects on the vasculature. Despite the links between lipoprotein (a) and atherosclerosis, there are limited data demonstrating an association with hypertension. Evidence from our clinic suggests that ~ 30% of the patients in this at-risk, hypertensive cohort had elevated lipoprotein (a) levels and that measurement of lipoprotein (a) maybe useful in risk stratification.
Collapse
|
162
|
Kaltoft M, Sigvardsen PE, Afzal S, Langsted A, Fuchs A, Kühl JT, Køber L, Kamstrup PR, Kofoed KF, Nordestgaard BG. Elevated lipoprotein(a) in mitral and aortic valve calcification and disease: The Copenhagen General Population Study. Atherosclerosis 2021; 349:166-174. [PMID: 34903381 DOI: 10.1016/j.atherosclerosis.2021.11.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/25/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS We tested the hypotheses (i) that elevated lipoprotein(a) is causally associated with both mitral and aortic valve calcification and disease, and (ii) that aortic valve calcification mediates the effect of elevated lipoprotein(a) on aortic valve stenosis. METHODS From the Copenhagen General Population study, we included 12,006 individuals who underwent cardiac computed tomography to measure mitral and aortic valve calcification and 85,884 to examine risk of heart valve disease. Participants had information on plasma lipoprotein(a) and genetic instruments associated with plasma lipoprotein(a) to investigate potential causality. RESULTS At age 70-79 years, 29% and 54% had mitral and aortic valve calcification, respectively. For 10-fold higher lipoprotein(a) levels, multifactorially adjusted odds ratios for mitral and aortic valve calcification were 1.26 (95% confidence interval: 1.13-1.41) and 1.62 (1.48-1.77). For mitral and aortic valve stenosis, corresponding hazard ratios were 0.93 (95%CI:0.40-2.15, 19 events) and 1.54 (1.38-1.71, 1158 events), respectively. For ≤23 versus ≥36 kringle IV type 2 number of repeats, the age and sex adjusted odds ratios for mitral and aortic valve calcification were 1.53 (1.18-1.99) and 2.23 (1.81-2.76). For carriers versus non-carriers of LPA rs10455872, odds ratios for mitral and aortic valve calcification were 1.33 (1.13-1.57) and 1.86 (1.64-2.13). For aortic valve stenosis, 31% (95%CI:16%-76%) of the effect of lipoprotein(a) was mediated through calcification. CONCLUSIONS Elevated lipoprotein(a) was genetically and observationally associated with mitral and aortic valve calcification and aortic valve stenosis. Aortic valve calcification mediated 31% of the effect of elevated lipoprotein(a) on aortic valve stenosis.
Collapse
Affiliation(s)
- Morten Kaltoft
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Per E Sigvardsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Shoaib Afzal
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Anne Langsted
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Andreas Fuchs
- Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Jørgen Tobias Kühl
- Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Lars Køber
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Pia R Kamstrup
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark
| | - Klaus F Kofoed
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Cardiology, The Heart Center, Rigshospitalet, Copenhagen University Hospital, Denmark; Department of Radiology, The Diagnostic Center, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
163
|
Durlach V, Bonnefont-Rousselot D, Boccara F, Varret M, Di-Filippo Charcosset M, Cariou B, Valero R, Charriere S, Farnier M, Morange PE, Meilhac O, Lambert G, Moulin P, Gillery P, Beliard-Lasserre S, Bruckert E, Carrié A, Ferrières J, Collet X, Chapman MJ, Anglés-Cano E. Lipoprotein(a): Pathophysiology, measurement, indication and treatment in cardiovascular disease. A consensus statement from the Nouvelle Société Francophone d'Athérosclérose (NSFA). Arch Cardiovasc Dis 2021; 114:828-847. [PMID: 34840125 DOI: 10.1016/j.acvd.2021.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 10/19/2022]
Abstract
Lipoprotein(a) is an apolipoprotein B100-containing low-density lipoprotein-like particle that is rich in cholesterol, and is associated with a second major protein, apolipoprotein(a). Apolipoprotein(a) possesses structural similarity to plasminogen but lacks fibrinolytic activity. As a consequence of its composite structure, lipoprotein(a) may: (1) elicit a prothrombotic/antifibrinolytic action favouring clot stability; and (2) enhance atherosclerosis progression via its propensity for retention in the arterial intima, with deposition of its cholesterol load at sites of plaque formation. Equally, lipoprotein(a) may induce inflammation and calcification in the aortic leaflet valve interstitium, leading to calcific aortic valve stenosis. Experimental, epidemiological and genetic evidence support the contention that elevated concentrations of lipoprotein(a) are causally related to atherothrombotic risk and equally to calcific aortic valve stenosis. The plasma concentration of lipoprotein(a) is principally determined by genetic factors, is not influenced by dietary habits, remains essentially constant over the lifetime of a given individual and is the most powerful variable for prediction of lipoprotein(a)-associated cardiovascular risk. However, major interindividual variations (up to 1000-fold) are characteristic of lipoprotein(a) concentrations. In this context, lipoprotein(a) assays, although currently insufficiently standardized, are of considerable interest, not only in stratifying cardiovascular risk, but equally in the clinical follow-up of patients treated with novel lipid-lowering therapies targeted at lipoprotein(a) (e.g. antiapolipoprotein(a) antisense oligonucleotides and small interfering ribonucleic acids) that markedly reduce circulating lipoprotein(a) concentrations. We recommend that lipoprotein(a) be measured once in subjects at high cardiovascular risk with premature coronary heart disease, in familial hypercholesterolaemia, in those with a family history of coronary heart disease and in those with recurrent coronary heart disease despite lipid-lowering treatment. Because of its clinical relevance, the cost of lipoprotein(a) testing should be covered by social security and health authorities.
Collapse
Affiliation(s)
- Vincent Durlach
- Champagne-Ardenne University, UMR CNRS 7369 MEDyC & Cardio-Thoracic Department, Reims University Hospital, 51092 Reims, France
| | - Dominique Bonnefont-Rousselot
- Metabolic Biochemistry Department, Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France; Université de Paris, CNRS, INSERM, UTCBS, 75006 Paris, France
| | - Franck Boccara
- Sorbonne University, GRC n(o) 22, C(2)MV, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, IHU ICAN, 75012 Paris, France; Service de Cardiologie, Hôpital Saint-Antoine, AP-HP, 75012 Paris, France
| | - Mathilde Varret
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalier Universitaire Xavier Bichat, 75018 Paris, France; Université de Paris, 75018 Paris, France
| | - Mathilde Di-Filippo Charcosset
- Hospices Civils de Lyon, UF Dyslipidémies, 69677 Bron, France; Laboratoire CarMen, INSERM, INRA, INSA, Université Claude-Bernard Lyon 1, 69495 Pierre-Bénite, France
| | - Bertrand Cariou
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'Institut du Thorax, 44000 Nantes, France
| | - René Valero
- Endocrinology Department, La Conception Hospital, AP-HM, Aix-Marseille University, INSERM, INRAE, C2VN, 13005 Marseille, France
| | - Sybil Charriere
- Hospices Civils de Lyon, INSERM U1060, Laboratoire CarMeN, Université Lyon 1, 69310 Pierre-Bénite, France
| | - Michel Farnier
- PEC2, EA 7460, University of Bourgogne Franche-Comté, 21079 Dijon, France; Department of Cardiology, CHU Dijon Bourgogne, 21000 Dijon, France
| | - Pierre E Morange
- Aix-Marseille University, INSERM, INRAE, C2VN, 13385 Marseille, France
| | - Olivier Meilhac
- INSERM, UMR 1188 DéTROI, Université de La Réunion, 97744 Saint-Denis de La Réunion, Reunion; CHU de La Réunion, CIC-EC 1410, 97448 Saint-Pierre, Reunion
| | - Gilles Lambert
- INSERM, UMR 1188 DéTROI, Université de La Réunion, 97744 Saint-Denis de La Réunion, Reunion; CHU de La Réunion, CIC-EC 1410, 97448 Saint-Pierre, Reunion
| | - Philippe Moulin
- Hospices Civils de Lyon, INSERM U1060, Laboratoire CarMeN, Université Lyon 1, 69310 Pierre-Bénite, France
| | - Philippe Gillery
- Laboratory of Biochemistry-Pharmacology-Toxicology, Reims University Hospital, University of Reims Champagne-Ardenne, UMR CNRS/URCA n(o) 7369, 51092 Reims, France
| | - Sophie Beliard-Lasserre
- Endocrinology Department, La Conception Hospital, AP-HM, Aix-Marseille University, INSERM, INRAE, C2VN, 13005 Marseille, France
| | - Eric Bruckert
- Service d'Endocrinologie-Métabolisme, Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France; IHU ICAN, Sorbonne University, 75013 Paris, France
| | - Alain Carrié
- Sorbonne University, UMR INSERM 1166, IHU ICAN, Laboratory of Endocrine and Oncological Biochemistry, Obesity and Dyslipidaemia Genetic Unit, Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France
| | - Jean Ferrières
- Department of Cardiology and INSERM UMR 1295, Rangueil University Hospital, TSA 50032, 31059 Toulouse, France
| | - Xavier Collet
- INSERM U1048, Institute of Metabolic and Cardiovascular Diseases, Rangueil University Hospital, BP 84225, 31432 Toulouse, France
| | - M John Chapman
- Sorbonne University, Hôpital Pitié-Salpêtrière and National Institute for Health and Medical Research (INSERM), 75013 Paris, France
| | - Eduardo Anglés-Cano
- Université de Paris, INSERM, Innovative Therapies in Haemostasis, 75006 Paris, France.
| |
Collapse
|
164
|
de Boer LM, Oorthuys AOJ, Wiegman A, Langendam MW, Kroon J, Spijker R, Zwinderman AH, Hutten BA. Statin therapy and lipoprotein(a) levels: a systematic review and meta-analysis. Eur J Prev Cardiol 2021; 29:779-792. [PMID: 34849724 DOI: 10.1093/eurjpc/zwab171] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/02/2021] [Indexed: 12/12/2022]
Abstract
AIMS Lipoprotein(a) [Lp(a)] is a causal and independent risk factor for cardiovascular disease (CVD). People with elevated Lp(a) are often prescribed statins as they also often show elevated low-density lipoprotein cholesterol (LDL-C) levels. While statins are well-established in lowering LDL-C, their effect on Lp(a) remains unclear. We evaluated the effect of statins compared to placebo on Lp(a) and the effects of different types and intensities of statin therapy on Lp(a). METHODS AND RESULTS We conducted a systematic review and meta-analysis of randomized trials with a statin and placebo arm. Medline and EMBASE were searched until August 2019. Quality assessment of studies was done using Cochrane risk-of-bias tool (RoB 2). Mean difference of absolute and percentage changes of Lp(a) in the statin vs. the placebo arms were pooled using a random-effects meta-analysis. We compared effects of different types and intensities of statin therapy using subgroup- and network meta-analyses. Certainty of the evidence was determined using GRADE (Grading of Recommendations, Assessment, Development, and Evaluation). Overall, 39 studies (24 448 participants) were included. Mean differences (95% confidence interval) of absolute and percentage changes in the statin vs. the placebo arms were 1.1 mg/dL (0.5-1.6, P < 0.0001) and 0.1% (-3.6% to 4.0%, P = 0.95), respectively (moderate-certainty evidence). None of the types of statins changed Lp(a) significantly compared to placebo (very low- to high-certainty evidence), as well as intensities of statin therapy (low- to moderate-certainty evidence). CONCLUSION Statin therapy does not lead to clinically important differences in Lp(a) compared to placebo in patients at risk for CVD. Our findings suggest that in these patients, statin therapy will not change Lp(a)-associated CVD risk.
Collapse
Affiliation(s)
- Lotte M de Boer
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Anna O J Oorthuys
- Department of Pediatrics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Albert Wiegman
- Department of Pediatrics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Miranda W Langendam
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - René Spijker
- Department of Medical Library, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Aeilko H Zwinderman
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health, Amsterdam, The Netherlands
| | - Barbara A Hutten
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
165
|
Ying Q, Chan DC, Barrett PHR, Watts GF. Unravelling lipoprotein metabolism with stable isotopes: tracing the flow. Metabolism 2021; 124:154887. [PMID: 34508741 DOI: 10.1016/j.metabol.2021.154887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/16/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022]
Abstract
Dysregulated lipoprotein metabolism is a major cause of atherosclerotic cardiovascular disease (ASCVD). Use of stable isotope tracers and compartmental modelling have provided deeper understanding of the mechanisms underlying lipid disorders in patients at high risk of ASCVD, including familial hypercholesterolemia (FH), elevated lipoprotein(a) [Lp(a)] and metabolic syndrome (MetS). In patients with FH, deficiency in low-density lipoprotein (LDL) receptor activity not only impairs the catabolism of LDL, but also induces hepatic overproduction and decreases catabolism of triglyceride-rich lipoproteins (TRLs). Patients with elevated Lp(a) are characterized by increased hepatic secretion of Lp(a) particles. Atherogenic dyslipidemia in MetS patients relates to a combination of overproduction of very-low density lipoprotein-apolipoprotein (apo) B-100, decreased catabolism of apoB-100-containing particles, and increased catabolism of high-density lipoprotein-apoA-I particles, as well as to impaired clearance of TRLs in the postprandial state. Kinetic studies show that weight loss, fish oils, statins and fibrates have complementary modes of action that correct atherogenic dyslipidemia. Defining the kinetic mechanisms of action of proprotein convertase subtilisin/kexin type 9 and angiopoietin-like 3 inhibitors on lipid and lipoprotein mechanism in dyslipidemic subjects will further our understanding of these therapies in decreasing the development of ASCVD. "Everything changes but change itself. Everything flows and nothing remains the same... You cannot step twice into the same river, for other waters and yet others go flowing ever on." Heraclitus (c.535- c. 475 BCE).
Collapse
Affiliation(s)
- Qidi Ying
- Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
| | - Dick C Chan
- Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
| | - P Hugh R Barrett
- Faculty of Medicine and Health, University of New England, Armidale, Australia
| | - Gerald F Watts
- Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia.
| |
Collapse
|
166
|
Lipoprotein (a) and Cardiovascular Disease: A Missing Link for Premature Atherosclerotic Heart Disease and/or Residual Risk. J Cardiovasc Pharmacol 2021; 79:e18-e35. [PMID: 34694242 DOI: 10.1097/fjc.0000000000001160] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/30/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Lipoprotein(a) or lipoprotein "little a" is an under-recognized causal risk factor for cardiovascular (CV) disease (CVD), including coronary atherosclerosis, aortic valvular stenosis, ischemic stroke, heart failure and peripheral arterial disease. Elevated plasma Lp(a) (≥50 mg/dL or ≥100 nmol/L) is commonly encountered in almost 1 in 5 individuals and confers a higher CV risk compared to those with normal Lp(a) levels, although such normal levels have not been generally agreed upon. Elevated Lp(a) is considered a cause of premature and accelerated atherosclerotic CVD. Thus, in patients with a positive family or personal history of premature coronary artery disease (CAD), Lp(a) should be measured. However, elevated Lp(a) may confer increased risk for incident CAD even in the absence of a family history of CAD, and even in those who have guideline-lowered LDL-cholesterol (<70 mg/dl) and continue to have a persisting CV residual risk. Thus, measurement of Lp(a) will have a significant clinical impact on the assessment of atherosclerotic CVD risk, and will assume a more important role in managing patients with CVD with the advent and clinical application of specific Lp(a)-lowering therapies. Conventional therapeutic approaches like lifestyle modification and statin therapy remain ineffective at lowering Lp(a). Newer treatment modalities, such as gene silencing via RNA interference with use of antisense oligonucleotide(s) or small interfering RNA molecules targeting Lp(a) seem very promising. These issues are herein reviewed, accumulated data are scrutinized, meta-analyses and current guidelines are tabulated and Lp(a)-related CVDs and newer therapeutic modalities are pictorially illustrated.
Collapse
|
167
|
Lee H, Park KS, Jeon YJ, Park EJ, Park S, Ann SH, Kim YG, Lee Y, Choi SH, Park GM. Lipoprotein(a) and subclinical coronary atherosclerosis in asymptomatic individuals. Atherosclerosis 2021; 349:190-195. [PMID: 34607706 DOI: 10.1016/j.atherosclerosis.2021.09.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS There are limited data regarding the association between lipoprotein(a) (Lp[a]) and subclinical coronary atherosclerosis. This study investigated the association between Lp(a) and subclinical coronary atherosclerosis detected by coronary computed tomographic angiography (CCTA) in an asymptomatic population. METHODS We retrospectively analyzed 7201 asymptomatic individuals (mean age 54.4 ± 7.9 years; 65.3% men with no prior history of coronary artery disease who voluntarily underwent CCTA as part of a general health examination). The degree and extent of subclinical coronary atherosclerosis were evaluated by CCTA. Study participants were stratified into quartiles according to their Lp(a) levels (<4.3, 4.3-8.9, 9.0-20.1, and ≥20.2 mg/dL). RESULTS Of the study participants, any coronary plaque on CCTA was observed in 2557 (35.5%). Specifically, calcified, non-calcified, and mixed plaques were observed in 2411 (33.5%), 363 (5.0%) and 249 (3.5%) participants, respectively. After adjustment for the presence of cardiovascular risk factors, the fourth Lp(a) quartile was significantly associated with any coronary (odds ratio [OR] 1.212; 95% confidence interval [CI] 1.038-1.416), calcified (1.205, 95% CI 1.030-1.410), non-calcified (1.588, 95% CI 1.152-2.189), or mixed (1.674, 95% CI 1.172-2.391) plaque compared with the first Lp(a) quartile. In addition, 442 (6.1%) had significant coronary artery stenosis (≥50% diameter stenosis). The odds ratio for significant stenosis (1.537, 95% CI 1.153-2.048) was higher in the fourth quartile compared with the first quartile. CONCLUSIONS In this large cross-sectional study with asymptomatic individuals undergoing CCTA, higher Lp(a) level was associated with subclinical coronary atherosclerosis.
Collapse
Affiliation(s)
- Hyeji Lee
- Department Emergency Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Kyung Sun Park
- Department of Nephrology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Young-Jee Jeon
- Department of Family Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Eun Ji Park
- Big Data Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Sangwoo Park
- Department of Cardiology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Soe Hee Ann
- Department of Cardiology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Yong-Giun Kim
- Department of Cardiology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Yongjik Lee
- Department of Thoracic and Cardiovascular Surgery, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Seong Hoon Choi
- Department of Radiology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Gyung-Min Park
- Department of Cardiology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea.
| |
Collapse
|
168
|
Handhle A, Viljoen A, Wierzbicki AS. Elevated Lipoprotein(a): Background, Current Insights and Future Potential Therapies. Vasc Health Risk Manag 2021; 17:527-542. [PMID: 34526771 PMCID: PMC8436116 DOI: 10.2147/vhrm.s266244] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
Lipoprotein(a) forms a subfraction of the lipid profile and is characterized by the addition of apolipprotein(a) (apo(a)) to apoB100 derived particles. Its levels are mostly genetically determined inversely related to the number of protein domain (kringle) repeats in apo(a). In epidemiological studies, it shows consistent association with cardiovascular disease (CVD) and most recently with extent of aortic stenosis. Issues with standardizing the measurement of Lp(a) are being resolved and consensus statements favor its measurement in patients at high risk of, or with family histories of CVD events. Major lipid-lowering therapies such as statin, fibrates, and ezetimibe have little effect on Lp(a) levels. Therapies such as niacin or cholesterol ester transfer protein (CETP) inhibitors lower Lp(a) as well as reducing other lipid-related risk factors but have failed to clearly reduce CVD events. Proprotein convertase subtilisin kexin-9 (PCSK9) inhibitors reduce cholesterol and Lp(a) as well as reducing CVD events. New antisense therapies specifically targeting apo(a) and hence Lp(a) have greater and more specific effects and will help clarify the extent to which intervention in Lp(a) levels will reduce CVD events.
Collapse
Affiliation(s)
- Ahmed Handhle
- Department of Metabolic Medicine/Chemical Pathology, Addenbrookes Hospital, Cambridge, UK
| | - Adie Viljoen
- Department of Metabolic Medicine/Chemical Pathology, North & East Hertfordshire Hospitals Trust, Lister Hospital, Hertfordshire, UK
| | - Anthony S Wierzbicki
- Department of Metabolic Medicine/Chemical Pathology, Guy's & St Thomas', Hospitals, London, SE1 7EH, UK
| |
Collapse
|
169
|
Wohlfahrt P, Jenča D, Melenovský V, Franeková J, Jabor A, Šramko M, Staněk V, Želízko M, Poledne R, Piťha J, Adámková V, Kautzner J. Very low lipoprotein(a) and increased mortality risk after myocardial infarction. Eur J Intern Med 2021; 91:33-39. [PMID: 33972150 DOI: 10.1016/j.ejim.2021.04.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/12/2021] [Accepted: 04/18/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Inconclusive data exist on risk associated with Lp(a) in patients after myocardial infarction (MI). Aims of the present study were to evaluate the association of Lp(a) level with total mortality and recurrent cardiovascular events. DESIGN AND METHODS Single center prospective registry of consecutive patients hospitalized for acute myocardial infarction between June 2017 and June 2020 at a large tertiary cardiac center with available blood samples drawn <24h of admission. RESULTS Data from 851 consecutive patients hospitalized for MI were evaluated. During the median follow-up of 19 months (interquartile range 10-27), 58 (6.8%) patients died. Nonlinear modelling revealed a U-shaped association between Lp(a) and total mortality risk. Compared to patients with Lp(a) ranging between 10-30 nmol/L and after multivariate adjustment, total mortality risk was increased both in patients with Lp(a)<7 nmol/L (hazard ratio (HR) 4.08, 95% confidence interval (CI) 1.72-9.68) and Lp(a) ≥125 nmol/L (HR 2.92, 95% CI 1.16-7.37), respectively. Similarly, the risk of combined endpoint of acute coronary syndrome recurrence or cardiovascular mortality was increased both in patients with low (sub-HR 2.60, 95% CI 1.33-5.08) and high (sub-HR 2.10, 95% CI 1.00-4.39) Lp(a). Adjustment for heart failure signs at the time of hospitalization weakened the association with total mortality and recurrent cardiovascular events. CONCLUSIONS In the present analysis, both high and low concentrations of Lp(a) were associated with an increased risk of total mortality and recurrent cardiovascular events after MI. The excess of mortality associated with Lp(a) was partially attributable to more prevalent heart failure.
Collapse
Affiliation(s)
- Peter Wohlfahrt
- Department of Preventive Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Centre for Cardiovascular Prevention, Charles University Medical School I and Thomayer Hospital, Prague; Charles University Medical School III, Prague, Czech Republic.
| | - Dominik Jenča
- Charles University Medical School III, Prague, Czech Republic; Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Janka Franeková
- Charles University Medical School III, Prague, Czech Republic; Department of Laboratory Methods, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Antonín Jabor
- Department of Laboratory Methods, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Marek Šramko
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vladimír Staněk
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Michael Želízko
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Rudolf Poledne
- Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Piťha
- Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Věra Adámková
- Department of Preventive Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Josef Kautzner
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Palacky University Medical School, Olomouc, Czech Republic
| |
Collapse
|
170
|
Ruscica M, Sirtori CR, Corsini A, Watts GF, Sahebkar A. Lipoprotein(a): Knowns, unknowns and uncertainties. Pharmacol Res 2021; 173:105812. [PMID: 34450317 DOI: 10.1016/j.phrs.2021.105812] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
Over the last 10 years, there have been advances on several aspects of lipoprotein(a) which are reviewed in the present article. Since the standard immunoassays for measuring lipoprotein(a) are not fully apo(a) isoform-insensitive, the application of an LC-MS/MS method for assaying molar concentrations of lipoprotein(a) has been advocated. Genome wide association, epidemiological, and clinical studies have established high lipoprotein(a) as a causal risk factor for atherosclerotic cardiovascular diseases (ASCVD). However, the relative importance of molar concentration, apo(a) isoform size or variants within the LPA gene is still controversial. Lipoprotein(a)-raising single nucleotide polymorphisms has not been shown to add on value in predicting ASCVD beyond lipoprotein(a) concentrations. Although hyperlipoproteinemia(a) represents an important confounder in the diagnosis of familial hypercholesterolemia (FH), it enhances the risk of ASCVD in these patients. Thus, identification of new cases of hyperlipoproteinemia(a) during cascade testing can increase the identification of high-risk individuals. However, it remains unclear whether FH itself increases lipoprotein(a). The ASCVD risk associated with lipoprotein(a) seems to follow a linear gradient across the distribution, regardless of racial subgroups and other risk factors. The inverse association with the risk of developing type 2 diabetes needs consideration as effective lipoprotein(a) lowering therapies are progressing towards the market. Considering that Mendelian randomization analyses have identified the degree of lipoprotein(a)-lowering that is required to achieve ASCVD benefit, the findings of the ongoing outcome trial with pelacarsen will clarify whether dramatically lowering lipoprotein(a) levels can reduce the risk of ASCVD.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy.
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Cardiometabolic Services, Department of Cardiology, Royal Perth Hospital, Australia
| | - Amirhossein Sahebkar
- School of Medicine, University of Western Australia, Perth, Australia; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
171
|
Parthymos I, Kostapanos MS, Mikhailidis DP, Florentin M. Lipoprotein (a) as a treatment target for cardiovascular disease prevention and related therapeutic strategies: a critical overview. Eur J Prev Cardiol 2021; 29:739-755. [PMID: 34389859 DOI: 10.1093/eurjpc/zwab052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/30/2020] [Accepted: 03/15/2021] [Indexed: 12/21/2022]
Abstract
Advances in several fields of cardiovascular (CV) medicine have produced new treatments (e.g. to treat dyslipidaemia) that have proven efficacy in terms of reducing deaths and providing a better quality of life. However, the burden of CV disease (CVD) remains high. Thus, there is a need to search for new treatment targets. Lipoprotein (a) [Lp(a)] has emerged as a potential novel target since there is evidence that it contributes to CVD events. In this narrative review, we present the current evidence of the potential causal relationship between Lp(a) and CVD and discuss the likely magnitude of Lp(a) lowering required to produce a clinical benefit. We also consider current and investigational treatments targeting Lp(a), along with the potential cost of these interventions.
Collapse
Affiliation(s)
- Ioannis Parthymos
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina 45110, Greece
| | - Michael S Kostapanos
- Department of General Medicine, Lipid Clinic, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London NW3 2QG, UK
| | - Matilda Florentin
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina 45110, Greece
| |
Collapse
|
172
|
Lee CK, Liao CW, Meng SW, Wu WK, Chiang JY, Wu MS. Lipids and Lipoproteins in Health and Disease: Focus on Targeting Atherosclerosis. Biomedicines 2021; 9:biomedicines9080985. [PMID: 34440189 PMCID: PMC8393881 DOI: 10.3390/biomedicines9080985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/01/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Despite advances in pharmacotherapy, intervention devices and techniques, residual cardiovascular risks still cause a large burden on public health. Whilst most guidelines encourage achieving target levels of specific lipids and lipoproteins to reduce these risks, increasing evidence has shown that molecular modification of these lipoproteins also has a critical impact on their atherogenicity. Modification of low-density lipoprotein (LDL) by oxidation, glycation, peroxidation, apolipoprotein C-III adhesion, and the small dense subtype largely augment its atherogenicity. Post-translational modification by oxidation, carbamylation, glycation, and imbalance of molecular components can reduce the capacity of high-density lipoprotein (HDL) for reverse cholesterol transport. Elevated levels of triglycerides (TGs), apolipoprotein C-III and lipoprotein(a), and a decreased level of apolipoprotein A-I are closely associated with atherosclerotic cardiovascular disease. Pharmacotherapies aimed at reducing TGs, lipoprotein(a), and apolipoprotein C-III, and enhancing apolipoprotein A-1 are undergoing trials, and promising preliminary results have been reported. In this review, we aim to update the evidence on modifications of major lipid and lipoprotein components, including LDL, HDL, TG, apolipoprotein, and lipoprotein(a). We also discuss examples of translating findings from basic research to potential therapeutic targets for drug development.
Collapse
Affiliation(s)
- Chih-Kuo Lee
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu 300, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Che-Wei Liao
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Department of Internal Medicine, National Taiwan University Cancer Center, Taipei 106, Taiwan
| | - Shih-Wei Meng
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu 300, Taiwan
| | - Wei-Kai Wu
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Jiun-Yang Chiang
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei 100, Taiwan
- Correspondence: (J.-Y.C.); (M.-S.W.)
| | - Ming-Shiang Wu
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Correspondence: (J.-Y.C.); (M.-S.W.)
| |
Collapse
|
173
|
Littmann K, Hagström E, Häbel H, Bottai M, Eriksson M, Parini P, Brinck J. Plasma lipoprotein(a) measured in the routine clinical care is associated to atherosclerotic cardiovascular disease during a 14-year follow-up. Eur J Prev Cardiol 2021; 28:2038-2047. [PMID: 34343284 DOI: 10.1093/eurjpc/zwab016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/13/2020] [Accepted: 01/20/2021] [Indexed: 01/22/2023]
Abstract
AIMS To investigate plasma lipoprotein(a) [Lp(a)] levels measured in routine clinical care and their association with mortality and cardiovascular disease. METHODS AND RESULTS This retrospective registry-based observational cohort study includes all individuals with plasma Lp(a) results measured at the Karolinska University Laboratory 2003-17. Outcome data were captured in national outcome registries. Levels of Lp(a) expressed in mass or molar units were examined separately. In adjusted Cox regression models, association between deciles of plasma Lp(a) concentrations, mortality, and cardiovascular outcomes were assessed. A total of 23 398 individuals [52% females, mean (standard deviation) age 55.5 (17.2) years, median Lp(a) levels 17 mg/dL or 19.5 nmol/L] were included. Individuals with an Lp(a) level >90th decile (>90 mg/dL or >180 nmol/L) had hazard ratios (95% confidence interval) of 1.25 (1.05-1.50) for major adverse cardiovascular events (P = 0.013), 1.37 (1.14-1.64) for atherosclerotic cardiovascular disease (P = 0.001), and 1.62 (1.28-2.05) for coronary artery disease (P ≤ 0.001), compared to individuals with Lp(a) ≤50th decile. No association between Lp(a) and mortality, peripheral artery disease, or ischaemic stroke was observed. CONCLUSION High Lp(a) levels are associated with adverse cardiovascular disease outcomes also in individuals with Lp(a) measured in routine clinical care. This supports the 2019 ESC/EAS recommendation to measure Lp(a) at least once during lifetime to assess cardiovascular risk and implies the need for intensive preventive therapy in patients with elevated Lp(a).
Collapse
Affiliation(s)
- Karin Littmann
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, H7 Medicine Huddinge, 171 77 Stockholm, Sweden.,Medical Unit of Endocrinology, Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| | - Emil Hagström
- Department of Medical Sciences, Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Henrike Häbel
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matteo Bottai
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mats Eriksson
- Department of Medicine Huddinge, Karolinska Institutet, H7 Medicine Huddinge, 171 77 Stockholm, Sweden.,Medical Unit of Endocrinology, Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| | - Paolo Parini
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, H7 Medicine Huddinge, 171 77 Stockholm, Sweden.,Medical Unit of Endocrinology, Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Brinck
- Department of Medicine Huddinge, Karolinska Institutet, H7 Medicine Huddinge, 171 77 Stockholm, Sweden.,Medical Unit of Endocrinology, Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
174
|
Iannuzzo G, Tripaldella M, Mallardo V, Morgillo M, Vitelli N, Iannuzzi A, Aliberti E, Giallauria F, Tramontano A, Carluccio R, Calcaterra I, Di Minno MND, Gentile M. Lipoprotein(a) Where Do We Stand? From the Physiopathology to Innovative Terapy. Biomedicines 2021; 9:838. [PMID: 34356902 PMCID: PMC8301358 DOI: 10.3390/biomedicines9070838] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/02/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022] Open
Abstract
A number of epidemiologic studies have demonstrated a strong association between increasing lipoprotein a [Lp(a)] and cardiovascular disease. This correlation was demonstrated independent of other known cardiovascular (CV) risk factors. Screening for Lp(a) in the general population is not recommended, although Lp(a) levels are predominantly genetically determined so a single assessment is needed to identify patients at risk. In 2019 ESC/EAS guidelines recommend Lp(a) measurement at least once a lifetime, fo subjects at very high and high CV risk and those with a family history of premature cardiovascular disease, to reclassify patients with borderline risk. As concerning medications, statins play a key role in lipid lowering therapy, but present poor efficacy on Lp(a) levels. Actually, treatment options for elevated serum levels of Lp(a) are very limited. Apheresis is the most effective and well tolerated treatment in patients with high levels of Lp(a). However, promising new therapies, in particular antisense oligonucleotides have showed to be able to significantly reduce Lp(a) in phase II RCT. This review provides an overview of the biology and epidemiology of Lp(a), with a view to future therapies.
Collapse
Affiliation(s)
- Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.T.); (V.M.); (M.M.); (N.V.); (I.C.); (M.N.D.D.M.); (M.G.)
| | - Maria Tripaldella
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.T.); (V.M.); (M.M.); (N.V.); (I.C.); (M.N.D.D.M.); (M.G.)
| | - Vania Mallardo
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.T.); (V.M.); (M.M.); (N.V.); (I.C.); (M.N.D.D.M.); (M.G.)
| | - Mena Morgillo
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.T.); (V.M.); (M.M.); (N.V.); (I.C.); (M.N.D.D.M.); (M.G.)
| | - Nicoletta Vitelli
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.T.); (V.M.); (M.M.); (N.V.); (I.C.); (M.N.D.D.M.); (M.G.)
| | - Arcangelo Iannuzzi
- Department of Medicine and Medical Specialties, A. Cardarelli Hospital, 80131 Naples, Italy;
| | - Emilio Aliberti
- North Tees University Hospital Stockton-on-Tees, Stockton TS19 8PE, UK;
| | - Francesco Giallauria
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (F.G.); (A.T.); (R.C.)
| | - Anna Tramontano
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (F.G.); (A.T.); (R.C.)
| | - Raffaele Carluccio
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (F.G.); (A.T.); (R.C.)
| | - Ilenia Calcaterra
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.T.); (V.M.); (M.M.); (N.V.); (I.C.); (M.N.D.D.M.); (M.G.)
| | - Matteo Nicola Dario Di Minno
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.T.); (V.M.); (M.M.); (N.V.); (I.C.); (M.N.D.D.M.); (M.G.)
| | - Marco Gentile
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.T.); (V.M.); (M.M.); (N.V.); (I.C.); (M.N.D.D.M.); (M.G.)
| |
Collapse
|
175
|
Paciullo F, Giannandrea D, Virgili G, Cagini C, Gresele P. Role of Increased Lipoprotein (a) in Retinal Vein Occlusion: A Systematic Review and Meta-analysis. TH OPEN 2021; 5:e295-e302. [PMID: 34240002 PMCID: PMC8260280 DOI: 10.1055/s-0041-1732803] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/26/2021] [Indexed: 12/24/2022] Open
Abstract
Background
Increased lipoprotein (a) [Lp(a)] has been associated with enhanced risk of cardiovascular events and more recently with venous thromboembolism. However, there is inconclusive data on the association between enhanced Lp(a) and retinal vein occlusion (RVO). We aimed to assess the role of Lp(a) in RVO.
Methods
We performed a systematic review and meta-analysis of the studies addressing the role of Lp(a) in RVO. A systematic literature search was performed to identify all published papers reporting Lp(a) levels. Main outcome measures consisted of Lp(a) levels in patients with (cases) or without (controls) RVO.
Results
We included 13 studies for a total of 1,040 cases and 16,648 controls. Lp(a) levels above normal limits were associated with RVO (OR 2.38, 95% CI 1.7–3.34) and patients with RVO had higher Lp(a) levels than controls (weighted mean difference: 13.4 mg/dL, 95% CI 8.2–18.6).
Conclusion
Increased Lp(a) levels associate with RVO and should be included among diagnostic and prognostic indexes for this unusual-site vein thrombosis. Therapeutic interventions aimed to lower Lp(a) should be tested in RVO patients.
Collapse
Affiliation(s)
- Francesco Paciullo
- Division of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - David Giannandrea
- Division of Neurology and Stroke Unit, Department of Neurology, Gubbio and Città di Castello Hospital, Perugia, Italy
| | - Gianni Virgili
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Ophthalmology Clinic, University of Firenze and AOU Careggi, Florence, Italy
| | - Carlo Cagini
- Section of Ophthalmology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Gresele
- Division of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
176
|
Kamstrup PR. Lipoprotein(a) and Cardiovascular Disease. Clin Chem 2021; 67:154-166. [PMID: 33236085 DOI: 10.1093/clinchem/hvaa247] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/25/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND High lipoprotein(a) concentrations present in 10%-20% of the population have long been linked to increased risk of ischemic cardiovascular disease. It is unclear whether high concentrations represent an unmet medical need. Lipoprotein(a) is currently not a target for treatment to prevent cardiovascular disease. CONTENT The present review summarizes evidence of causality for high lipoprotein(a) concentrations gained from large genetic epidemiologic studies and discusses measurements of lipoprotein(a) and future treatment options for high values found in an estimated >1 billion individuals worldwide. SUMMARY Evidence from mechanistic, observational, and genetic studies support a causal role of lipoprotein(a) in the development of cardiovascular disease, including coronary heart disease and peripheral arterial disease, as well as aortic valve stenosis, and likely also ischemic stroke. Effect sizes are most pronounced for myocardial infarction, peripheral arterial disease, and aortic valve stenosis where high lipoprotein(a) concentrations predict 2- to 3-fold increases in risk. Lipoprotein(a) measurements should be performed using well-validated assays with traceability to a recognized calibrator to ensure common cut-offs for high concentrations and risk assessment. Randomized cardiovascular outcome trials are needed to provide final evidence of causality and to assess the potential clinical benefit of novel, potent lipoprotein(a) lowering therapies.
Collapse
Affiliation(s)
- Pia R Kamstrup
- Department of Clinical Biochemistry.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark
| |
Collapse
|
177
|
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
|
178
|
Kille A, Nührenberg T, Franke K, Valina CM, Leibundgut G, Tsimikas S, Neumann FJ, Hochholzer W. Association of lipoprotein(a) with intrinsic and on-clopidogrel platelet reactivity. J Thromb Thrombolysis 2021; 53:1-9. [PMID: 34213715 PMCID: PMC8791920 DOI: 10.1007/s11239-021-02515-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2021] [Indexed: 12/01/2022]
Abstract
Lipoprotein(a) [Lp(a)] is an independent, genetically determined, and causal risk factor for cardiovascular disease. Laboratory data have suggested an interaction of Lp(a) with platelet function, potentially caused by its interaction with platelet receptors. So far, the potential association of Lp(a) with platelet activation and reactivity has not been proven in larger clinical cohorts. This study analyzed intrinsic platelet reactivity before loading with clopidogrel 600 mg and on-treatment platelet reactivity tested 24 h following loading in patients undergoing elective coronary angiography. Platelet reactivity was tested by optical aggregometry following stimulation with collagen or adenosine diphosphate as well as by flow cytometry. Lp(a) levels were directly measured in all patients from fresh samples. The present analysis included 1912 patients. Lp(a) levels ranged between 0 and 332 mg/dl. There was a significant association of rising levels of Lp(a) with a higher prevalence of a history of ischemic heart disease (p < 0.001) and more extensive coronary artery disease (p = 0.001). Results for intrinsic (p = 0.80) and on-clopidogrel platelet reactivity (p = 0.81) did not differ between quartiles of Lp(a) levels. Flow cytometry analyses of expression of different platelet surface proteins (CD41, CD62P or PAC-1) confirmed these findings. Correlation analyses of levels of Lp(a) with any of the tested platelet activation markers did not show any correlation. The present data do not support the hypothesis of an interaction of Lp(a) with platelet reactivity.
Collapse
Affiliation(s)
- Alexander Kille
- Department of Cardiology and Angiology II, Medical Center, University of Freiburg, University Heart Center Freiburg-Bad Krozingen, Suedring 15, 79189, Bad Krozingen, Germany.
| | - Thomas Nührenberg
- Department of Cardiology and Angiology II, Medical Center, University of Freiburg, University Heart Center Freiburg-Bad Krozingen, Suedring 15, 79189, Bad Krozingen, Germany
| | - Kilian Franke
- Department of Cardiology and Angiology II, Medical Center, University of Freiburg, University Heart Center Freiburg-Bad Krozingen, Suedring 15, 79189, Bad Krozingen, Germany
| | - Christian M Valina
- Department of Cardiology and Angiology II, Medical Center, University of Freiburg, University Heart Center Freiburg-Bad Krozingen, Suedring 15, 79189, Bad Krozingen, Germany
| | | | - Sotirios Tsimikas
- Sulpizio Cardiovascular Center, Division of Cardiovascular Medicine, University of California San Diego, San Diego, USA
| | - Franz-Josef Neumann
- Department of Cardiology and Angiology II, Medical Center, University of Freiburg, University Heart Center Freiburg-Bad Krozingen, Suedring 15, 79189, Bad Krozingen, Germany
| | - Willibald Hochholzer
- Department of Cardiology and Angiology II, Medical Center, University of Freiburg, University Heart Center Freiburg-Bad Krozingen, Suedring 15, 79189, Bad Krozingen, Germany
| |
Collapse
|
179
|
The Role of Antisense Therapies Targeting Lipoprotein(a). J Cardiovasc Pharmacol 2021; 78:e5-e11. [PMID: 34232223 DOI: 10.1097/fjc.0000000000001045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 04/03/2021] [Indexed: 01/09/2023]
Abstract
ABSTRACT Atherosclerotic cardiovascular disease (ASCVD) continues to be the leading cause of preventable death in the United States. Elevated low-density lipoprotein cholesterol (LDL-C) is well known to result in cardiovascular disease. Mainstay therapy for reducing LDL-C and ASCVD risk is statin therapy. Despite achieving desired LDL-C levels with lipid-lowering therapy, cardiovascular residual risk often persists. Elevated lipoprotein(a) [Lp(a)] levels have been highlighted as an inherent independent predictor of ASCVD, and decreasing Lp(a) levels may result in a significant reduction in the residual risk in high-risk patients. To date, there are no approved medications to lower Lp(a) levels. Nicotinic acid, proprotein convertase subtilisin/kexin 9 inhibitors, and antisense oligonucleotide have demonstrated modest to potent Lp(a) reduction. Spotlight has been placed on antisense oligonucleotides and their role in Lp(a) lowering. APO(a)LRx is in the frontline for selectively decreasing Lp(a) concentrations and ongoing research may prove that this medication may lower Lp(a)-mediated residual risk, translating into cardiovascular benefit.
Collapse
|
180
|
Afshar M, Yazdan-Ashoori S, Engert JC, Thanassoulis G. Drugs for Prevention and Treatment of Aortic Stenosis: How Close Are We? Can J Cardiol 2021; 37:1016-1026. [DOI: 10.1016/j.cjca.2021.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/25/2022] Open
|
181
|
Arnold M, Schweizer J, Nakas CT, Schütz V, Westphal LP, Inauen C, Pokorny T, Luft A, Leichtle A, Arnold M, Bicvic A, Fischer U, De Marchis GM, Bonati LH, Müller MD, Kahles T, Nedeltchev K, Cereda CW, Kägi G, Bustamante A, Montaner J, Ntaios G, Foerch C, Spanaus K, von Eckardstein A, Katan M. Lipoprotein(a) is associated with large artery atherosclerosis stroke aetiology and stroke recurrence among patients below the age of 60 years: results from the BIOSIGNAL study. Eur Heart J 2021; 42:2186-2196. [PMID: 33709115 DOI: 10.1093/eurheartj/ehab081] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/02/2020] [Accepted: 01/28/2021] [Indexed: 12/24/2022] Open
Abstract
AIMS Lipoprotein(a) [Lp(a)] is a recognized causal risk factor for atherosclerotic cardiovascular disease but its role for acute ischaemic stroke (AIS) is controversial. In this study, we evaluated the association of Lp(a) with large artery atherosclerosis (LAA) stroke and risk of recurrent cerebrovascular events in AIS patients. METHODS AND RESULTS For this analysis of the prospective, observational, multicentre BIOSIGNAL cohort study we measured Lp(a) levels in plasma samples of 1733 primarily Caucasian (98.6%) AIS patients, collected within 24 h after symptom onset. Primary outcomes were LAA stroke aetiology and recurrent cerebrovascular events (ischaemic stroke or transient ischaemic attack) within 1 year. We showed that Lp(a) levels are independently associated with LAA stroke aetiology [adjusted odds ratio 1.48, 95% confidence interval (CI) 1.14-1.90, per unit log10Lp(a) increase] and identified age as a potent effect modifier (Pinteraction =0.031) of this association. The adjusted odds ratio for LAA stroke in patients aged <60 years was 3.64 (95% CI 1.76-7.52) per unit log10Lp(a) increase and 4.04 (95% CI 1.73-9.43) using the established cut-off ≥100 nmol/l. For 152 recurrent cerebrovascular events, we did not find a significant association in the whole cohort. However, Lp(a) levels ≥100 nmol/l were associated with an increased risk for recurrent events among patients who were either <60 years [adjusted hazard ratio (HR) 2.40, 95% CI 1.05-5.47], had evident LAA stroke aetiology (adjusted HR 2.18, 95% CI 1.08-4.40), or had no known atrial fibrillation (adjusted HR 1.60, 95% CI 1.03-2.48). CONCLUSION Elevated Lp(a) was independently associated with LAA stroke aetiology and risk of recurrent cerebrovascular events among primarily Caucasian individuals aged <60 years or with evident arteriosclerotic disease.
Collapse
Affiliation(s)
- Markus Arnold
- Department for Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Juliane Schweizer
- Department for Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Christos T Nakas
- Department of Clinical Chemistry, Inselspital, University Hospital and University of Bern, Bern, Switzerland.,Laboratory of Biometry, University of Thessaly, Volos, Greece
| | - Valerie Schütz
- Department for Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Laura P Westphal
- Department for Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Corinne Inauen
- Department for Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Pokorny
- Department for Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Andreas Luft
- Department for Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Alexander Leichtle
- Department of Clinical Chemistry, Inselspital, University Hospital and University of Bern, Bern, Switzerland
| | - Marcel Arnold
- Department for Neurology, Inselspital, University Hospital and University of Bern, Bern, Switzerland
| | - Antonela Bicvic
- Department for Neurology, University Hospital Zurich, Zurich, Switzerland.,Department for Neurology, Inselspital, University Hospital and University of Bern, Bern, Switzerland
| | - Urs Fischer
- Department for Neurology, Inselspital, University Hospital and University of Bern, Bern, Switzerland
| | - Gian Marco De Marchis
- Department for Neurology & Stroke Center, University Hospital of Basel & University of Basel, Basel, Switzerland
| | - Leo H Bonati
- Department for Neurology & Stroke Center, University Hospital of Basel & University of Basel, Basel, Switzerland
| | - Mandy D Müller
- Department for Neurology & Stroke Center, University Hospital of Basel & University of Basel, Basel, Switzerland
| | - Timo Kahles
- Department of Neurology, Cantonal Hospital Aarau, Switzerland
| | | | - Carlo W Cereda
- Neurocentro della Svizzera Italiana, Stroke Center EOC, Lugano, Switzerland
| | - Georg Kägi
- Department of Neurology, Cantonal Hospital St, Gallen, Switzerland
| | - Alejandro Bustamante
- Department for Neurology, Vall d'Hebron Institute of Research (VHIR), Universitat Autónoma de Barcelona, Spain
| | - Joan Montaner
- Department for Neurology, Vall d'Hebron Institute of Research (VHIR), Universitat Autónoma de Barcelona, Spain
| | - George Ntaios
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Christian Foerch
- Department of Neurology, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | - Katharina Spanaus
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | | | - Mira Katan
- Department for Neurology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
182
|
Speer T, Ridker PM, von Eckardstein A, Schunk SJ, Fliser D. Lipoproteins in chronic kidney disease: from bench to bedside. Eur Heart J 2021; 42:2170-2185. [PMID: 33393990 DOI: 10.1093/eurheartj/ehaa1050] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/16/2020] [Accepted: 12/08/2020] [Indexed: 12/24/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with high cardiovascular risk. CKD patients exhibit a specific lipoprotein pattern termed 'uraemic dyslipidaemia', which is characterized by rather normal low-density lipoprotein cholesterol, low high-density lipoprotein cholesterol, and high triglyceride plasma levels. All three lipoprotein classes are involved in the pathogenesis of CKD-associated cardiovascular diseases (CVDs). Uraemia leads to several modifications of the structure of lipoproteins such as changes of the proteome and the lipidome, post-translational protein modifications (e.g. carbamylation) and accumulation of small-molecular substances within the lipoprotein moieties, which affect their functionality. Lipoproteins from CKD patients interfere with lipid transport and promote inflammation, oxidative stress, endothelial dysfunction as well as other features of atherogenesis, thus contributing to the development of CKD-associated CVD. While, lipid-modifying therapies play an important role in the management of CKD patients, their efficacy is modulated by kidney function. Novel therapeutic agents to prevent the adverse remodelling of lipoproteins in CKD and to improve their functional properties are highly desirable and partially under development.
Collapse
Affiliation(s)
- Thimoteus Speer
- Translational Cardio-Renal Medicine, Saarland University, Kirrberger Strasse, Building 41, D-66421 Homburg/Saar, Germany.,Department of Internal Medicine IV, Saarland University Hospital, Nephrology and Hypertension, Kirrberger Strasse, Building 41, D-66421 Homburg/Saar, Germany
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02215, USA
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland
| | - Stefan J Schunk
- Translational Cardio-Renal Medicine, Saarland University, Kirrberger Strasse, Building 41, D-66421 Homburg/Saar, Germany
| | - Danilo Fliser
- Translational Cardio-Renal Medicine, Saarland University, Kirrberger Strasse, Building 41, D-66421 Homburg/Saar, Germany
| |
Collapse
|
183
|
Langsted A, Nordestgaard BG. Genetics of Lipoprotein(a): Cardiovascular Disease and Future Therapy. Curr Atheroscler Rep 2021; 23:46. [PMID: 34148150 DOI: 10.1007/s11883-021-00937-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Lipoprotein(a) levels are determined 80-90% by genetics and differ by up to 1000-fold between individuals. This review discusses the most recent literature on lipoprotein(a) as a risk factor for cardiovascular disease, as well as future lipoprotein(a)lowering therapies. RECENT FINDINGS Over the past few decades, numerous studies have observed that high lipoprotein(a) levels are associated observationally and causally through human genetics with increased risk of cardiovascular disease. Also, the development of safe and effective therapies to lower lipoprotein(a) is ongoing, most importantly using antisense oligonucleotides to prevent production of lipoprotein(a). Finally, both observational and genetic studies have estimated the extent to which lowering of lipoprotein(a) is needed to obtain a clinically meaningful reduction in the risk of cardiovascular disease. Lipoprotein(a) is a causal risk factor for cardiovascular disease; however, currently no approved safe and effective therapy is available to lower lipoprotein(a) levels. That said, promising randomized studies using antisense oligonucleotides show up to 80% reductions in lipoprotein(a), reductions that hopefully will result in lowering the risk of cardiovascular disease as presently tested in the ongoing HORIZON phase 3 trial.
Collapse
Affiliation(s)
- Anne Langsted
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark. .,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark. .,Department of Clinical Medicine Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark.,Department of Clinical Medicine Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
184
|
Abstract
PURPOSE OF REVIEW Genetic, epidemiological, and translational data indicate that Lipoprotein (a) [Lp(a)] is likely in the causal pathway for atherosclerotic cardiovascular diseases as well as calcification of the aortic valves. RECENT FINDINGS Lp(a) is structurally similar to low-density lipoprotein, but in addition to apolipoprotein B-100, it has a glycoprotein apolipoprotein(a) [apo(a)], which is attached to the apolipoprotein B-100. Several distinctive properties of Lp(a) can be attributed to the presence of apo(a). This review discusses the current state of literature on pathophysiological and clinical aspects of Lp(a). After five decades of research, the understanding of Lp(a) structure, biochemistry, and pathophysiology of its cardiovascular manifestations still remains less than fully understood. Universally, Lp(a) elevation may be the most predominant monogenetic lipid disorder with approximate prevalence of Lp(a)>50 mg/dL among estimated >1.4 billion people. This makes a compelling rationale for diagnosing and managing Lp(a)-mediated risk. In addition to discussing various cardiovascular phenotypes of Lp(a) and associated morbidity, we also outline current and emerging therapies aimed at identifying a definitive treatment for elevated Lp(a) levels.
Collapse
Affiliation(s)
- Anum Saeed
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA USA
| | - Sina Kinoush
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX USA
| | - Salim S. Virani
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX USA
- Health Policy, Quality & Informatics Program, Michael E. DeBakey Veterans Affairs Medical Center Health Services Research and Development Center for Innovations, Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, 2002 Holcombe Blvd, Houston, TX 77030 USA
- Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston, TX USA
| |
Collapse
|
185
|
Lipoprotein(a) levels and atherosclerotic plaque characteristics in the carotid artery: The Plaque at RISK (PARISK) study. Atherosclerosis 2021; 329:22-29. [PMID: 34216874 DOI: 10.1016/j.atherosclerosis.2021.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/01/2021] [Accepted: 06/03/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Lipoprotein(a) is an independent risk factor for cardiovascular disease and recurrent ischemic stroke. Lipoprotein(a) levels are known to be associated with carotid artery stenosis, but the relation of lipoprotein(a) levels to carotid atherosclerotic plaque composition and morphology is less known. We hypothesize that higher lipoprotein(a) levels and lipoprotein(a)-related SNPs are associated with a more vulnerable carotid plaque and that this effect is sex-specific. METHODS In 182 patients of the Plaque At RISK study we determined lipoprotein(a) concentrations, apo(a) KIV-2 repeats and LPA SNPs. Imaging characteristics of carotid atherosclerosis were determined by MDCTA (n = 161) and/or MRI (n = 171). Regressions analyses were used to investigate sex-stratified associations between lipoprotein(a) levels, apo(a) KIV-2 repeats, and LPA SNPs and imaging characteristics. RESULTS Lipoprotein(a) was associated with presence of lipid-rich necrotic core (LRNC) (aOR = 1.07, 95% CI: 1.00; 1.15), thin-or-ruptured fibrous cap (TRFC) (aOR = 1.07, 95% CI: 1.01; 1.14), and degree of stenosis (β = 0.44, 95% CI: 0.00; 0.88). In women, lipoprotein(a) was associated with presence of intraplaque hemorrhage (IPH) (aOR = 1.25, 95% CI: 1.06; 1.61). In men, lipoprotein(a) was associated with degree of stenosis (β = 0.58, 95% CI: 0.04; 1.12). Rs10455872 was significantly associated with increased calcification volume (β = 1.07, 95% CI: 0.25; 1.89) and absence of plaque ulceration (aOR = 0.25, 95% CI: 0.04; 0.93). T3888P was associated with absence of LRNC (aOR = 0.36, 95% CI: 0.16; 0.78) and smaller maximum vessel wall area (β = -10.24, 95%CI: -19.03; -1.44). CONCLUSIONS In patients with symptomatic carotid artery stenosis, increased lipoprotein(a) levels were associated with degree of stenosis, and IPH, LRNC, and TRFC, known as vulnerable plaque characteristics, in the carotid artery. T3888P was associated with lower LRNC prevalence and smaller maximum vessel wall area. Further research in larger study populations is needed to confirm these results.
Collapse
|
186
|
Morris G, Berk M, Walder K, O'Neil A, Maes M, Puri BK. The lipid paradox in neuroprogressive disorders: Causes and consequences. Neurosci Biobehav Rev 2021; 128:35-57. [PMID: 34118292 DOI: 10.1016/j.neubiorev.2021.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 04/27/2021] [Accepted: 06/06/2021] [Indexed: 02/07/2023]
Abstract
Chronic systemic inflammation is associated with an increased risk of cardiovascular disease in an environment of low low-density lipoprotein (LDL) and low total cholesterol and with the pathophysiology of neuroprogressive disorders. The causes and consequences of this lipid paradox are explored. Circulating activated neutrophils can release inflammatory molecules such as myeloperoxidase and the pro-inflammatory cytokines interleukin-1 beta, interleukin-6 and tumour necrosis factor-alpha. Since activated neutrophils are associated with atherosclerosis and cardiovascular disease and with major depressive disorder, bipolar disorder and schizophrenia, it seems reasonable to hypothesise that the inflammatory molecules released by them may act as mediators of the link between systemic inflammation and the development of atherosclerosis in neuroprogressive disorders. This hypothesis is tested by considering the association at a molecular level of systemic inflammation with increased LDL oxidation; increased small dense LDL levels; increased lipoprotein (a) concentration; secretory phospholipase A2 activation; cytosolic phospholipase A2 activation; increased platelet activation; decreased apolipoprotein A1 levels and function; decreased paroxonase-1 activity; hyperhomocysteinaemia; and metabolic endotoxaemia. These molecular mechanisms suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand
| | | |
Collapse
|
187
|
Abstract
PURPOSE OF REVIEW Lipoprotein(a) [Lp(a)] is a plasma circulating apoB100 (apoB) containing lipoprotein. It has a unique glycoprotein bound to the apoB100, apolipoprotein(a) [apo(a)]. The majority of the population expresses two apo(a) isoforms, when bound to apoB100 they create two circulating Lp(a) particles. Lp(a) levels are genetically determined and epidemiological studies have established elevated levels of Lp(a) to be a causal risk factor of cardiovascular disease (CVD). Lp(a) levels differ across racial groups and Blacks of Sub-Saharan decent have higher levels when compared to white. In comparison to white populations, studies in minorities are less represented in the published literature. Additionally, there is a lack of standardization in the commercial assays used to measured Lp(a) levels, and hence it is difficult to assess risk based on individual Lp(a) levels, but risk seems to occur in the upper percentiles of the population. RECENT FINDINGS A recent study using data from the UK biobank highlights the racial differences in Lp(a) levels and the increase risk in CVD amongst all races. SUMMARY This review will highlight Lp(a) biology and physiology with a focus on available data from racially diverse cohorts. There is a need to perform studies in diverse populations to understand if they are at higher risk than whites are.
Collapse
|
188
|
Chakraborty A, Pang J, Chan DC, Barnett W, Woodward AM, Vorster M, Watts GF. Effectiveness of proprotein convertase subtilisin/kexin-9 monoclonal antibody treatment on plasma lipoprotein(a) concentrations in patients with elevated lipoprotein(a) attending a clinic. Clin Cardiol 2021; 44:805-813. [PMID: 33955565 PMCID: PMC8207967 DOI: 10.1002/clc.23607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Lipoprotein(a) (Lp[a]) is a causal risk factor for atherosclerotic cardiovascular disease (ASCVD). Proprotein convertase subtilisin/kexin-9 monoclonal antibodies (PCSK9mAbs) can lower Lp(a) levels in clinical trials, but their effects in patients with elevated Lp(a) in clinical practice remain unclear. AIMS To investigate the effectiveness and safety of PCSK9mAbs in lowering plasma Lp(a) in patients with elevated Lp(a) concentrations in a lipid clinic. METHODS This was an open-label study of 53 adult patients with elevated Lp(a) concentration (≥0.5 g/L). Clinical, biochemical, and safety data were collected before and on treatment with evolocumab or alirocumab over a mean period of 11 months. RESULTS Treatment with a PCSK9mAb resulted in a significant reduction of 0.29 g/L (-22%) in plasma Lp(a) concentration (p<.001). There were also significant reductions in low-density lipoprotein-cholesterol (LDL-C) (-53%), remnant-cholesterol (-12%) and apolipoprotein B (-43%) concentrations. The change in Lp(a) concentration was significantly different from a comparable group of 35 patients with elevated Lp(a) who were not treated with a PCSK9mAb (-22% vs. -2%, p<.001). The reduction in Lp(a) concentration was not associated with the corresponding changes in LDL-C, remnant-cholesterol, and apolipoprotein B (p>.05 in all). 7.5% and 47% of the patients attained a target concentration of Lp(a) <0.5 g/L and LDL-C <1.8 mmol/L, respectively. PCSK9mAbs were well tolerated, the common adverse effects being pharyngitis (9.4%), nasal congestion (7.6%), myalgia (9.4%), diarrhoea (7.6%), arthralgia (9.4%) and injection site reactions (11%). CONCLUSION PCSK9mAbs can effectively and safely lower plasma Lp(a) concentrations in patients with elevated Lp(a) in clinical practice; the impact of the fall in Lp(a) on ASCVD outcomes requires further investigation.
Collapse
Affiliation(s)
- Anindita Chakraborty
- School of Medicine, Faculty of Health and Medical SciencesUniversity of Western AustraliaPerthAustralia
| | - Jing Pang
- School of Medicine, Faculty of Health and Medical SciencesUniversity of Western AustraliaPerthAustralia
| | - Dick C. Chan
- School of Medicine, Faculty of Health and Medical SciencesUniversity of Western AustraliaPerthAustralia
| | - Wendy Barnett
- Lipid Disorders Clinic, Cardiometabolic Services, Department of CardiologyRoyal Perth HospitalPerthAustralia
| | - Ann Marie Woodward
- Lipid Disorders Clinic, Cardiometabolic Services, Department of CardiologyRoyal Perth HospitalPerthAustralia
| | - Mary Vorster
- Lipid Disorders Clinic, Cardiometabolic Services, Department of CardiologyRoyal Perth HospitalPerthAustralia
| | - Gerald F. Watts
- School of Medicine, Faculty of Health and Medical SciencesUniversity of Western AustraliaPerthAustralia
- Lipid Disorders Clinic, Cardiometabolic Services, Department of CardiologyRoyal Perth HospitalPerthAustralia
| |
Collapse
|
189
|
Langsted A, Nordestgaard BG, Kamstrup PR. Low lipoprotein(a) levels and risk of disease in a large, contemporary, general population study. Eur Heart J 2021; 42:1147-1156. [PMID: 33724357 DOI: 10.1093/eurheartj/ehaa1085] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/02/2020] [Accepted: 12/18/2020] [Indexed: 12/13/2022] Open
Abstract
AIMS With the current focus on lipoprotein(a) as a likely causal risk factor for cardiovascular disease and new drugs potentially on the market to lower lipoprotein(a) levels, the safety of lowering lipoprotein(a) to low levels becomes increasingly important. We tested whether low levels of lipoprotein(a) and corresponding LPA genotypes associate with major disease groups including cancers and infectious disease. METHODS AND RESULTS We included 109 440 individuals from the Copenhagen General Population Study. For main World Health Organization International Classification of Diseases 10th edition chapter diseases, the only concordant association of low levels of lipoprotein(a) plasma levels and corresponding LPA genotypes with risk of disease was with low risk of diseases of the circulatory system. Furthermore, no concordant association of low levels of lipoprotein(a) plasma levels and corresponding LPA genotypes with the risk of any cancer (i.e. cancer subtypes combined) or infectious disease was seen. The hazard ratio for the risk of any cancer was 1.06 [95% confidence interval (CI): 0.97-1.15] for the first vs. the fourth quartile of lipoprotein(a), 1.02 (0.97-1.07) for the fourth vs. the first quartile of KIV-2 number of repeats, and 1.01 (0.96-1.07) for rs10455872 non-carriers vs. carriers. The corresponding hazard ratios for the risk of hospitalization for infection were 1.05 (95% CI: 0.99-1.10), 1.02 (0.98-1.07), and 0.97 (0.93-1.03), respectively. CONCLUSION In a large, contemporary, general population cohort, apart from the well-established association with cardiovascular disease, low levels of lipoprotein(a) and corresponding LPA genotypes did not concordantly associate with any major disease groups including cancers and infections. There is no safety signal from our results to indicate that low levels of lipoprotein(a) are harmful.
Collapse
Affiliation(s)
- Anne Langsted
- Department of Clinical Biochemistry, Borgmester Ib Juuls Vej 73, opgang 7, Herlev 2730, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, opgang 7, Herlev 2730, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Borgmester Ib Juuls Vej 73, opgang 7, Herlev 2730, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, opgang 7, Herlev 2730, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Pia R Kamstrup
- Department of Clinical Biochemistry, Borgmester Ib Juuls Vej 73, opgang 7, Herlev 2730, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, opgang 7, Herlev 2730, Denmark
| |
Collapse
|
190
|
Affiliation(s)
- Zareen M Farukhi
- Center for Lipid Metabolomics, Divisions of Preventive and Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | - Samia Mora
- Center for Lipid Metabolomics, Divisions of Preventive and Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
191
|
Langlois MR, Nordestgaard BG, Langsted A, Chapman MJ, Aakre KM, Baum H, Borén J, Bruckert E, Catapano A, Cobbaert C, Collinson P, Descamps OS, Duff CJ, von Eckardstein A, Hammerer-Lercher A, Kamstrup PR, Kolovou G, Kronenberg F, Mora S, Pulkki K, Remaley AT, Rifai N, Ros E, Stankovic S, Stavljenic-Rukavina A, Sypniewska G, Watts GF, Wiklund O, Laitinen P. Quantifying atherogenic lipoproteins for lipid-lowering strategies: consensus-based recommendations from EAS and EFLM. Clin Chem Lab Med 2021; 58:496-517. [PMID: 31855562 DOI: 10.1515/cclm-2019-1253] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Indexed: 12/15/2022]
Abstract
The joint consensus panel of the European Atherosclerosis Society (EAS) and the European Federation of Clinical Chemistry and Laboratory Medicine (EFLM) recently addressed present and future challenges in the laboratory diagnostics of atherogenic lipoproteins. Total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDLC), LDL cholesterol (LDLC), and calculated non-HDLC (=total - HDLC) constitute the primary lipid panel for estimating risk of atherosclerotic cardiovascular disease (ASCVD) and can be measured in the nonfasting state. LDLC is the primary target of lipid-lowering therapies. For on-treatment follow-up, LDLC shall be measured or calculated by the same method to attenuate errors in treatment decisions due to marked between-method variations. Lipoprotein(a) [Lp(a)]-cholesterol is part of measured or calculated LDLC and should be estimated at least once in all patients at risk of ASCVD, especially in those whose LDLC declines poorly upon statin treatment. Residual risk of ASCVD even under optimal LDL-lowering treatment should be also assessed by non-HDLC or apolipoprotein B (apoB), especially in patients with mild-to-moderate hypertriglyceridemia (2-10 mmol/L). Non-HDLC includes the assessment of remnant lipoprotein cholesterol and shall be reported in all standard lipid panels. Additional apoB measurement can detect elevated LDL particle (LDLP) numbers often unidentified on the basis of LDLC alone. Reference intervals of lipids, lipoproteins, and apolipoproteins are reported for European men and women aged 20-100 years. However, laboratories shall flag abnormal lipid values with reference to therapeutic decision thresholds.
Collapse
Affiliation(s)
- Michel R Langlois
- Department of Laboratory Medicine, AZ St-Jan, Ruddershove 10, 8000 Brugge, Belgium.,University of Ghent, Ghent, Belgium
| | - Børge G Nordestgaard
- Herlev and Gentofte Hospital, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anne Langsted
- Herlev and Gentofte Hospital, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - M John Chapman
- National Institute for Health and Medical Research (INSERM), Paris, France.,Endocrinology-Metabolism Service, Pitié-Salpetriere University Hospital, Paris, France
| | - Kristin M Aakre
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Hannsjörg Baum
- Institute for Laboratory Medicine, Mikrobiologie und Blutdepot, Regionale Kliniken Holding RKH GmbH, Ludwigsburg, Germany
| | - Jan Borén
- Institute of Medicine, Sahlgrenska Academy at Göteborg University, Gothenburg, Sweden.,Wallenberg Laboratory for Cardiovascular and Metabolic Research, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Eric Bruckert
- Department of Endocrinology and Prevention of Cardiovascular Disease, Pitié-Salpetriere University Hospital, Paris, France
| | - Alberico Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.,IRCCS Multimedica, Milan, Italy
| | - Christa Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul Collinson
- Department of Clinical Blood Sciences, St George's University Hospitals NHS Foundation Trust and St George's University of London, London, UK.,Department of Cardiology, St George's University Hospitals NHS Foundation Trust and St George's University of London, London, UK
| | - Olivier S Descamps
- Department of Internal Medicine, Centres Hospitaliers Jolimont, Haine-Saint-Paul, Belgium.,Department of Cardiology, UCL Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christopher J Duff
- Department of Clinical Biochemistry, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | | | | | - Pia R Kamstrup
- Herlev and Gentofte Hospital, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Genovefa Kolovou
- Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece
| | - Florian Kronenberg
- Department of Medical Genetics, Molecular and Clinical Pharmacology, Division of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Samia Mora
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kari Pulkki
- Department of Clinical Chemistry, University of Turku and Turku University Hospital, Turku, Finland
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nader Rifai
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emilio Ros
- Lipid Clinic, Department of Endocrinology and Nutrition, Institut d'Investigacions Biomèdiques August Pi Sunyer, Hospital Clínic, Barcelona, Spain.,Ciber Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Sanja Stankovic
- Center for Medical Biochemistry, Clinical Center of Serbia, Belgrade, Serbia
| | | | - Grazyna Sypniewska
- Department of Laboratory Medicine, Collegium Medicum, NC University, Bydgoszcz, Poland
| | - Gerald F Watts
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, University of Western Australia, Perth, Australia
| | - Olov Wiklund
- Institute of Medicine, Sahlgrenska Academy at Göteborg University, Gothenburg, Sweden.,Wallenberg Laboratory for Cardiovascular and Metabolic Research, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Päivi Laitinen
- Department of Clinical Chemistry, HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | | |
Collapse
|
192
|
Szarek M, Bittner VA, Aylward P, Baccara-Dinet M, Bhatt DL, Diaz R, Fras Z, Goodman SG, Halvorsen S, Harrington RA, Jukema JW, Moriarty PM, Pordy R, Ray KK, Sinnaeve P, Tsimikas S, Vogel R, White HD, Zahger D, Zeiher AM, Steg PG, Schwartz GG. Lipoprotein(a) lowering by alirocumab reduces the total burden of cardiovascular events independent of low-density lipoprotein cholesterol lowering: ODYSSEY OUTCOMES trial. Eur Heart J 2021; 41:4245-4255. [PMID: 33051646 PMCID: PMC7724642 DOI: 10.1093/eurheartj/ehaa649] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/24/2020] [Accepted: 07/24/2020] [Indexed: 12/11/2022] Open
Abstract
Aims Lipoprotein(a) concentration is associated with first cardiovascular events in clinical trials. It is unknown if this relationship holds for total (first and subsequent) events. In the ODYSSEY OUTCOMES trial in patients with recent acute coronary syndrome (ACS), the proprotein convertase subtilisin/kexin type 9 inhibitor alirocumab reduced lipoprotein(a), low-density lipoprotein cholesterol (LDL-C), and cardiovascular events compared with placebo. This post hoc analysis determined whether baseline levels and alirocumab-induced changes in lipoprotein(a) and LDL-C [corrected for lipoprotein(a) cholesterol] independently predicted total cardiovascular events. Methods and results Cardiovascular events included cardiovascular death, non-fatal myocardial infarction, stroke, hospitalization for unstable angina or heart failure, ischaemia-driven coronary revascularization, peripheral artery disease events, and venous thromboembolism. Proportional hazards models estimated relationships between baseline lipoprotein(a) and total cardiovascular events in the placebo group, effects of alirocumab treatment on total cardiovascular events by baseline lipoprotein(a), and relationships between lipoprotein(a) reduction with alirocumab and subsequent risk of total cardiovascular events. Baseline lipoprotein(a) predicted total cardiovascular events with placebo, while higher baseline lipoprotein(a) levels were associated with greater reduction in total cardiovascular events with alirocumab (hazard ratio P
trend = 0.045). Alirocumab-induced reductions in lipoprotein(a) (median −5.0 [−13.6, 0] mg/dL) and corrected LDL-C (median −51.3 [−67.1, −34.0] mg/dL) independently predicted lower risk of total cardiovascular events. Each 5-mg/dL reduction in lipoprotein(a) predicted a 2.5% relative reduction in cardiovascular events. Conclusion Baseline lipoprotein(a) predicted the risk of total cardiovascular events and risk reduction by alirocumab. Lipoprotein(a) lowering contributed independently to cardiovascular event reduction, supporting the concept of lipoprotein(a) as a treatment target after ACS. ![]()
Collapse
Affiliation(s)
- Michael Szarek
- Department of Epidemiology and Biostatistics, State University of New York, Downstate School of Public Health, 450 Clarkson Avenue, MS 43, Brooklyn, NY 11203, USA
| | - Vera A Bittner
- Division of Cardiovascular Disease, University of Alabama at Birmingham, 701 19th Street South - LHRB 310, Birmingham, AL 35294, USA
| | - Philip Aylward
- Department of Cardiology, South Australian Health and Medical Research Institute, Flinders University and Medical Centre, South Australia 5042, Australia
| | - Marie Baccara-Dinet
- Sanofi R&D, Global Development, 371 Rue du Professeur Blayac, 34080 Montpellier, France
| | - Deepak L Bhatt
- Department of Medicine, Brigham and Women's Hospital Heart and Vascular Center, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Rafael Diaz
- Estudios Clínicos Latinoamérica, Instituto Cardiovascular de Rosario, Paraguay 160, Rosario, Santa Fe, Rosario 2000, Argentina
| | - Zlatko Fras
- Preventive Cardiology Unit, Department of Vascular Medicine, Division of Medicine, University Medical Centre Ljubljana, Zaloška cesta 7, SI-1525 Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, 87 Ave NW, Edmonton, Alberta T6G 2E1, Canada.,Division of Cardiology, St. Michael's Hospital, Room 6-034 Donnelly Wing, Toronto, Ontario M5B 1W8, Canada
| | - Sigrun Halvorsen
- Department of Cardiology, Oslo Universitetssykehus HF Ulleval, and University of Oslo, Problemveien 7, 0315 Oslo, Norway
| | - Robert A Harrington
- Stanford Center for Clinical Research, Department of Medicine, Stanford University, 291 Campus Drive, Li Ka Shing Building, Stanford, CA 94305-5101, USA
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands.,Netherlands Heart Institute, Utrecht, the Netherlands
| | - Patrick M Moriarty
- Clinical Pharmacology-University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Robert Pordy
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Rd., Tarrytown, NY 10591, USA
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College London, Reynolds Building, St Dunstans Road, London W6 8RP, UK
| | - Peter Sinnaeve
- Department of Cardiovascular Medicine, University Hospitals Leuven, Herestraat 49, Leuven 3000, Belgium
| | - Sotirios Tsimikas
- Sulpizio Cardiovascular Center, Division of Cardiovascular Medicine, University of California San Diego, 9434 Medical Center Dr, La Jolla, CA 92037, USA
| | - Robert Vogel
- Department of Medicine, University of Colorado Denver, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Harvey D White
- Green Lane Cardiovascular Services, Auckland City Hospital, 5 Park Road, Grafton, Auckland 1142, New Zealand
| | - Doron Zahger
- Department of Cardiology, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University of the Negev, PO Box 151 Beer Sheva 8410, Israel
| | - Andreas M Zeiher
- Department of Medicine III, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany
| | - Ph Gabriel Steg
- Université de Paris, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Université de Paris, FACT (French Alliance for Cardiovascular Trials), INSERM U1148, 46 Rue Henri Huchard, Paris 75018, France.,National Heart and Lung Institute, Imperial College, Royal Brompton Hospital, Sydney St, Chelsea, London SW3 6NP, UK
| | - Gregory G Schwartz
- Division of Cardiology, University of Colorado School of Medicine, Box B130, Aurora, CO 80045, USA
| | | |
Collapse
|
193
|
Borén J, Chapman MJ, Krauss RM, Packard CJ, Bentzon JF, Binder CJ, Daemen MJ, Demer LL, Hegele RA, Nicholls SJ, Nordestgaard BG, Watts GF, Bruckert E, Fazio S, Ference BA, Graham I, Horton JD, Landmesser U, Laufs U, Masana L, Pasterkamp G, Raal FJ, Ray KK, Schunkert H, Taskinen MR, van de Sluis B, Wiklund O, Tokgozoglu L, Catapano AL, Ginsberg HN. Low-density lipoproteins cause atherosclerotic cardiovascular disease: pathophysiological, genetic, and therapeutic insights: a consensus statement from the European Atherosclerosis Society Consensus Panel. Eur Heart J 2021; 41:2313-2330. [PMID: 32052833 PMCID: PMC7308544 DOI: 10.1093/eurheartj/ehz962] [Citation(s) in RCA: 780] [Impact Index Per Article: 195.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/10/2019] [Accepted: 01/08/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - M John Chapman
- Endocrinology-Metabolism Division, Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, France.,National Institute for Health and Medical Research (INSERM), Paris, France
| | - Ronald M Krauss
- Department of Atherosclerosis Research, Children's Hospital Oakland Research Institute and UCSF, Oakland, CA 94609, USA
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Jacob F Bentzon
- Department of Clinical Medicine, Heart Diseases, Aarhus University, Aarhus, Denmark.,Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Mat J Daemen
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Linda L Demer
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Physiology, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert A Hegele
- Department of Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Australia
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, University of Copenhagen, Denmark
| | - Gerald F Watts
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia.,Department of Cardiology, Lipid Disorders Clinic, Royal Perth Hospital, Perth, Australia
| | - Eric Bruckert
- INSERM UMRS1166, Department of Endocrinology-Metabolism, ICAN - Institute of CardioMetabolism and Nutrition, AP-HP, Hopital de la Pitie, Paris, France
| | - Sergio Fazio
- Departments of Medicine, Physiology and Pharmacology, Knight Cardiovascular Institute, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Brian A Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, UK.,Institute for Advanced Studies, University of Bristol, Bristol, UK.,MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - Jay D Horton
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ulf Landmesser
- Department of Cardiology, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Liebigstraße 20, Leipzig, Germany
| | - Luis Masana
- Research Unit of Lipids and Atherosclerosis, IISPV, CIBERDEM, University Rovira i Virgili, C. Sant Llorenç 21, Reus 43201, Spain
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frederick J Raal
- Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Kausik K Ray
- Department of Primary Care and Public Health, Imperial Centre for Cardiovascular Disease Prevention, Imperial College London, London, UK
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Faculty of Medicine, Technische Universität München, Lazarettstr, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Marja-Riitta Taskinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Olov Wiklund
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lale Tokgozoglu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, and IRCCS MultiMedica, Milan, Italy
| | - Henry N Ginsberg
- Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA
| |
Collapse
|
194
|
Dong H, Cong H, Wang J, Jiang Y, Liu C, Zhang Y, Zhu Y, Wang Q. Correlations between lipoprotein(a) gene polymorphisms and calcific aortic valve disease and coronary heart disease in Han Chinese. J Int Med Res 2021; 48:300060520965353. [PMID: 33100089 PMCID: PMC7645393 DOI: 10.1177/0300060520965353] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Objective To investigate the relationship between lipoprotein(a) gene (LPA) polymorphisms and calcific aortic valve disease (CAVD) and coronary heart disease (CHD) in Han Chinese. Methods A total of 148 patients were recruited (n = 71 with CAVD and n = 77 with CHD) based on a diagnosis achieved using color Doppler echocardiography, coronary angiography, or computed tomography angiography. Seventy-one control individuals without CAVD or CHD were also recruited. Biomarkers including levels of lipoprotein(a) [Lp(a)], low-density lipoprotein and high-density lipoprotein cholesterol, apolipoprotein A1, and apolipoprotein B were tested. LPA polymorphisms rs10455872, rs6415084, rs3798221, and rs7770628 were analyzed using SNaPshot SNP. Results Lp(a) levels were significantly higher in CAVD and CHD groups compared with controls. There was no significant difference in the allelic frequency distribution of rs3798221, rs7770628, or rs6415084 between CHD, CAVD, and control groups. Linear regression showed that rs3798221, rs7770628, and rs6415084 were associated with increased Lp(a) concentrations. Two CAVD patients among the 219 participants carried AG minor alleles at rs10455872, while the remainder carried AA minor alleles. Conclusion rs3798221, rs6415084, and rs7770628 polymorphisms within LPA are associated with higher Lp(a) plasma levels, which correlate with increased CAVD and CHD risks.
Collapse
Affiliation(s)
- Hongzhi Dong
- Graduate School, Tianjin Medical University, Tianjin, China.,Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Hongliang Cong
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Jing Wang
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Yiyao Jiang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Anhui, China.,Department of Cardiovascular Surgery, The First Central Hospital Affiliated to Nankai University, Tianjin, China
| | - Chao Liu
- Institution of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, China
| | - Yingyi Zhang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Yanbo Zhu
- Department of Ultrasound, Tianjin Chest Hospital, Tianjin, China
| | - Qingtong Wang
- Department of Clinical Laboratory, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
195
|
Barbagallo CM, Cefalù AB, Giammanco A, Noto D, Caldarella R, Ciaccio M, Averna MR, Nardi E. Lipoprotein Abnormalities in Chronic Kidney Disease and Renal Transplantation. Life (Basel) 2021; 11:life11040315. [PMID: 33916487 PMCID: PMC8067409 DOI: 10.3390/life11040315] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) is one of the most important risk factors for cardiovascular disease (CVD). Despite the kidney having no direct implications for lipoproteins metabolism, advanced CKD dyslipidemia is usually present in patients with CKD, and the frequent lipid and lipoprotein alterations occurring in these patients play a role of primary importance in the development of CVD. Although hypertriglyceridemia is the main disorder, a number of lipoprotein abnormalities occur in these patients. Different enzymes pathways and proteins involved in lipoprotein metabolism are impaired in CKD. In addition, treatment of uremia may modify the expression of lipoprotein pattern as well as determine acute changes. In renal transplantation recipients, the main lipid alteration is hypercholesterolemia, while hypertriglyceridemia is less pronounced. In this review we have analyzed lipid and lipoprotein disturbances in CKD and also their relationship with progression of renal disease. Hypolipidemic treatments may also change the natural history of CVD in CKD patients and may represent important strategies in the management of CKD patients.
Collapse
Affiliation(s)
- Carlo Maria Barbagallo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties—University of Palermo, Via del Vespro, 127, 90127 Palermo, Italy; (C.M.B.); (A.B.C.); (A.G.); (D.N.); (R.C.); (M.R.A.)
| | - Angelo Baldassare Cefalù
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties—University of Palermo, Via del Vespro, 127, 90127 Palermo, Italy; (C.M.B.); (A.B.C.); (A.G.); (D.N.); (R.C.); (M.R.A.)
| | - Antonina Giammanco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties—University of Palermo, Via del Vespro, 127, 90127 Palermo, Italy; (C.M.B.); (A.B.C.); (A.G.); (D.N.); (R.C.); (M.R.A.)
| | - Davide Noto
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties—University of Palermo, Via del Vespro, 127, 90127 Palermo, Italy; (C.M.B.); (A.B.C.); (A.G.); (D.N.); (R.C.); (M.R.A.)
| | - Rosalia Caldarella
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties—University of Palermo, Via del Vespro, 127, 90127 Palermo, Italy; (C.M.B.); (A.B.C.); (A.G.); (D.N.); (R.C.); (M.R.A.)
| | - Marcello Ciaccio
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Section of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, 90127 Palermo, Italy;
| | - Maurizio Rocco Averna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties—University of Palermo, Via del Vespro, 127, 90127 Palermo, Italy; (C.M.B.); (A.B.C.); (A.G.); (D.N.); (R.C.); (M.R.A.)
| | - Emilio Nardi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties—University of Palermo, Via del Vespro, 127, 90127 Palermo, Italy; (C.M.B.); (A.B.C.); (A.G.); (D.N.); (R.C.); (M.R.A.)
- Correspondence: ; Tel.: +39-916-554-316
| |
Collapse
|
196
|
Liu J, Cao Y, Sheng Z. Effect of lipoprotein α on coronary stents expansion and its risk factors. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:249-256. [PMID: 33927071 PMCID: PMC10929932 DOI: 10.11817/j.issn.1672-7347.2021.200398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Indexed: 11/03/2022]
Abstract
OBJECTIVES To analyze the effect of hyperlipoproteinemia (α) on immediate expansion after coronary stent implantation guided by intravascular ultrasound (IVUS). METHODS A total of 160 patients (175 lesions) with coronary heart disease diagnosed by coronary artery angiography, who were performed percutaneous intervention guided by IVUS in the Department of Cardiology, Third Xiangya Hospital, Central South University, were enrolled retrospectively.According to the concentration of lipoproteina, the patients were divided into 2 groups: a hyperlipoproteinemia (α) group and a control group. Cardiac ejection fraction was measured with echocardiography. Logistic regression was used to analyze the influential factors for hyperlipoproteinemia (α). The target vessel was examined by IVUS to analyze the immediate expansion effect of hyperlipoproteinemia (α) after stent implantation. RESULTS The mean stent expansion index, lesion length, stent number, stent symmetry index and posterior balloon diameter were (94.73±18.9)%, (52.92±29.1) mm, (2.11±0.85), (83.62±13.07)%, and (9.46±2.00) mm in the hyperlipoproteinemia (α) group, respectively. Compared with the control group, there were significantly difference (all P<0.05). Multivariable regression analysis showed that the decreased creatinine clearance rate was an independent risk factor for hyperlipoproteinemia (α) (P<0.05). CONCLUSIONS Hyperlipoproteinemia (α) appears to be a predictor of stent underexpansion, and the decreased creatinine clearance rate is an independent risk factor for hyperlipoproteinemia (α).
Collapse
Affiliation(s)
- Jiajia Liu
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Yu Cao
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Zhe Sheng
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| |
Collapse
|
197
|
Swerdlow DI, Rider DA, Yavari A, Lindholm MW, Campion GV, Nissen SE. Treatment and prevention of lipoprotein(a)-mediated cardiovascular disease: the emerging potential of RNA interference therapeutics. Cardiovasc Res 2021; 118:1218-1231. [PMID: 33769464 PMCID: PMC8953457 DOI: 10.1093/cvr/cvab100] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 02/19/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022] Open
Abstract
Lipid- and lipoprotein-modifying therapies have expanded substantially in the last 25 years, resulting in reduction in the incidence of major adverse cardiovascular events. However, no specific lipoprotein(a) [Lp(a)]-targeting therapy has yet been shown to reduce cardiovascular disease risk. Many epidemiological and genetic studies have demonstrated that Lp(a) is an important genetically determined causal risk factor for coronary heart disease, aortic valve disease, stroke, heart failure, and peripheral vascular disease. Accordingly, the need for specific Lp(a)-lowering therapy has become a major public health priority. Approximately 20% of the global population (1.4 billion people) have elevated levels of Lp(a) associated with higher cardiovascular risk, though the threshold for determining ‘high risk’ is debated. Traditional lifestyle approaches to cardiovascular risk reduction are ineffective at lowering Lp(a). To address a lifelong risk factor unmodifiable by non-pharmacological means, Lp(a)-lowering therapy needs to be safe, highly effective, and tolerable for a patient population who will likely require several decades of treatment. N-acetylgalactosamine-conjugated gene silencing therapeutics, such as small interfering RNA (siRNA) and antisense oligonucleotide targeting LPA, are ideally suited for this application, offering a highly tissue- and target transcript-specific approach with the potential for safe and durable Lp(a) lowering with as few as three or four doses per year. In this review, we evaluate the causal role of Lp(a) across the cardiovascular disease spectrum, examine the role of established lipid-modifying therapies in lowering Lp(a), and focus on the anticipated role for siRNA therapeutics in treating and preventing Lp(a)-related disease.
Collapse
Affiliation(s)
| | | | - Arash Yavari
- Experimental Therapeutics, Radcliffe, Department of Medicine, University of Oxford, UK
| | | | | | - Steven E Nissen
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
198
|
Chang NC, Yeh CT, Lin YK, Kuo KT, Fong IH, Kounis NG, Hu P, Hung MY. Garcinol Attenuates Lipoprotein(a)-Induced Oxidative Stress and Inflammatory Cytokine Production in Ventricular Cardiomyocyte through α7-Nicotinic Acetylcholine Receptor-Mediated Inhibition of the p38 MAPK and NF-κB Signaling Pathways. Antioxidants (Basel) 2021; 10:antiox10030461. [PMID: 33809417 PMCID: PMC8000018 DOI: 10.3390/antiox10030461] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/27/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
Garcinol, a nicotinic acetylcholine receptor (nAChR) antagonist, has recently been established as an anti-inflammation agent. However, the molecular mechanism by which garcinol suppresses inflammation in the context of acute myocardial infarction (AMI) remains unclear. Hypothesis: We hypothesized that the administration of physiological doses of garcinol in mice with isoproterenol-induced AMI decreased the effect of lipoprotein(a) (Lp(a))-induced inflammation both in vivo and in vitro via the α7-nAChRs mediated p38 mitogen-activated protein kinase (MAPK)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) signaling pathway. We analyzed altered reactive oxygen species (ROS) generation, the production of superoxide by mitochondria, cytokine expression patterns, and the role of the p38 MAPK/NF-κB signaling pathway after Lp(a)-stimulated human ventricular cardiomyocyte AC16 cells were treated with increasing doses of garcinol. C-reactive protein (CRP), interleukin (IL)-1β, IL-6, or tumor necrosis factor (TNF)-α production were detected by enzyme-linked immunosorbent assay. The Cell Counting Kit-8 assay was used to evaluate drug cytotoxicity. Western blots and confocal fluorescence microscopy were used to determine altered expression patterns of inflammatory biomarkers. We also examined whether the therapeutic effect of garcinol in AMI was mediated in part by α7-nAChR. Lp(a)-induced inflammatory cardiomyocytes had increased expression of membrane-bound α7-nAChRs in vitro and in vivo. Low-dose garcinol did not affect cardiomyocyte viability but significantly reduced mitochondrial ROS, CRP, IL-1β, IL-6, and TNF-α production in Lp(a)-stimulated cardiomyocytes (p < 0.05). The Lp(a)-induced phosphorylation of p38 MAPKs, CamKII, and NFκB, as well as NFκB-p65 nuclear translocation, was also suppressed (p < 0.05) by garcinol, while the inhibition of p38 MAPK by the inhibitor SB203580 decreased the phosphorylation of extracellular signal-regulated kinase (ERK) and p38 MAPK. Garcinol protected cardiomyocytes by inhibiting apoptosis and inflammation in mice with AMI. Furthermore, garcinol also enhanced the expression of microRNA-205 that suppressed the α7-nAChR-induced p38 MAPK/NF-κB signaling pathway. Garcinol suppresses Lp(a)-induced oxidative stress and inflammatory cytokines by α7-nAChR-mediated inhibition of p38 MAPK/NF-κB signaling in cardiomyocyte AC16 cells and isoproterenol-induced AMI mice.
Collapse
Affiliation(s)
- Nen-Chung Chang
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan
| | - Chi-Tai Yeh
- Department of Medical Research and Education, Taipei Medical University—Shuang Ho Hospital, New Taipei City 23561, Taiwan; (C.-T.Y.); (I.-H.F.)
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu City 30015, Taiwan
| | - Yen-Kuang Lin
- Biostatistics Center, Office of Data Science, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Data Science, Taipei Medical University, Taipei 110, Taiwan
- Research Center of Big Data, College of Management, Taipei Medical University, Taipei 110, Taiwan;
| | - Kuang-Tai Kuo
- Division of Thoracic Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- Division of Thoracic Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Iat-Hang Fong
- Department of Medical Research and Education, Taipei Medical University—Shuang Ho Hospital, New Taipei City 23561, Taiwan; (C.-T.Y.); (I.-H.F.)
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu City 30015, Taiwan
| | - Nicholas G. Kounis
- Department of Internal Medicine, Division of Cardiology, University of Patras Medical School, 26221 Patras, Greece;
| | - Patrick Hu
- Department of Cardiology, University of California, Riverside, CA 92521, USA;
- Department of Cardiology, Riverside Medical Clinic, Riverside, CA 92506, USA
| | - Ming-Yow Hung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Correspondence: ; Tel.: +886-2-22490088
| |
Collapse
|
199
|
VLDL Cholesterol Accounts for One-Half of the Risk of Myocardial Infarction Associated With apoB-Containing Lipoproteins. J Am Coll Cardiol 2021; 76:2725-2735. [PMID: 33272366 DOI: 10.1016/j.jacc.2020.09.610] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/10/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Plasma apolipoprotein B (apoB) is a composite measure of all apoB-containing lipoproteins causing atherosclerotic cardiovascular disease; however, it is unclear which fraction of risk is explained by cholesterol and triglycerides, respectively, in very low-density lipoproteins (VLDLs). OBJECTIVES The authors tested the hypothesis that VLDL cholesterol and triglycerides each explain part of the myocardial infarction risk from apoB-containing lipoproteins. METHODS Nested within 109,751 individuals from the Copenhagen General Population Study, the authors examined 25,480 subjects free of lipid-lowering therapy and myocardial infarction at study entry. All had measurements of plasma apoB (quantitating number of apoB-containing lipoproteins) and cholesterol and triglyceride content of VLDL, intermediate-density lipoproteins (IDLs), and low-density lipoproteins (LDLs). RESULTS During a median 11 years of follow-up, 1,816 were diagnosed with myocardial infarction. Per 1-mmol/l higher levels, multivariable-adjusted hazard ratios for myocardial infarction were 2.07 (95% confidence interval [CI]: 1.81 to 2.36) for VLDL cholesterol, 1.19 (95% CI: 1.14 to 1.25) for VLDL triglycerides, 5.38 (95% CI: 3.73 to 7.75) for IDL cholesterol, and 1.86 (95% CI: 1.62 to 2.14) for LDL cholesterol. Per 1-g/l higher plasma apoB, the corresponding value was 2.21 (95% CI: 1.90 to 2.58). In a step-up Cox regression, risk factors for myocardial infarction entered by importance as VLDL cholesterol, systolic blood pressure, smoking, and IDL + LDL cholesterol, whereas VLDL triglycerides did not enter the model. VLDL cholesterol explained 50% and IDL + LDL cholesterol 29% of the risk of myocardial infarction from apoB-containing lipoproteins, whereas VLDL triglycerides did not explain risk. CONCLUSIONS VLDL cholesterol explained one-half of the myocardial infarction risk from elevated apoB-containing lipoproteins, whereas VLDL triglycerides did not explain risk.
Collapse
|
200
|
Ho JH, Adam S, Liu Y, Azmi S, Dhage S, Syed AA, Ammori BJ, Donn R, Heald A, Gibson MJ, Malik RA, Yang X, Durrington PN, Tsimikas S, Soran H. Effect of bariatric surgery on plasma levels of oxidised phospholipids, biomarkers of oxidised LDL and lipoprotein(a). J Clin Lipidol 2021; 15:320-331. [PMID: 33518459 DOI: 10.1016/j.jacl.2020.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Obesity is associated with adverse cardiovascular outcomes and this is improved following bariatric surgery. Oxidised phospholipids (OxPL) are thought to reflect the pro-inflammatory effects of lipoprotein(a) [Lp(a)], and both are independent predictors of cardiovascular disease. OBJECTIVE Our study sought to determine the impact of bariatric surgery on OxPL, biomarkers of oxidised LDL (OxLDL) and Lp(a). METHODS This is a prospective, observational study of 59 patients with severe obesity undergoing bariatric surgery. Blood samples were obtained prior to surgery and at 6 and 12 months after. Sixteen patients attending the tertiary medical weight management clinic at the same centre were also recruited for comparison. Lipid and metabolic blood parameters, OxLDL, OxPL on apolipoprotein B-100 (OxPL-apoB), IgG and IgM autoantibodies to MDA-LDL, IgG and IgM apoB-immune complexes and Lp(a) were measured. RESULTS Reduction in body mass index (BMI) was significant following bariatric surgery, from median 48 kg/m2 at baseline to 37 kg/m2 at 6 months and 33 kg/m2 at 12 months. OxPL-apoB levels decreased significantly at 12 months following surgery [5.0 (3.2-7.4) to 3.8 (3.0-5.5) nM, p = 0.001], while contrastingly, Lp(a) increased significantly [10.2 (3.8-31.9) to 16.9 (4.9-38.6) mg/dl, p = 0.002]. There were significant post-surgical decreases in IgG and IgM biomarkers, particularly at 12 months, while OxLDL remained unchanged. CONCLUSIONS Bariatric surgery results in a significant increase in Lp(a) but reductions in OxPL-apoB and other biomarkers of oxidised lipoproteins, suggesting increased synthetic capacity and reduced oxidative stress. These biomarkers might be clinically useful to monitor physiological effects of weight loss interventions.
Collapse
Affiliation(s)
- Jan Hoong Ho
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK; Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Safwaan Adam
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK; Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Yifen Liu
- Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Shazli Azmi
- Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Shaishav Dhage
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK; Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Akheel A Syed
- Department of Diabetes & Endocrinology, Salford Royal NHS Foundation Trust, Salford, UK
| | - Basil J Ammori
- Department of Surgery, Salford Royal NHS Foundation Trust, Salford, UK
| | - Rachelle Donn
- Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Adrian Heald
- Department of Diabetes & Endocrinology, Salford Royal NHS Foundation Trust, Salford, UK
| | - Martin J Gibson
- Department of Diabetes & Endocrinology, Salford Royal NHS Foundation Trust, Salford, UK
| | - Rayaz A Malik
- Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK; Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Xiaohong Yang
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California, San Diego, USA
| | - Paul N Durrington
- Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California, San Diego, USA.
| | - Handrean Soran
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK; Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK.
| |
Collapse
|