1
|
Luo D, Björnson E, Wang X, Zhang B, Xu C, Guan Y, Xiang S, Borén J, Hao X, Chen J. Distinct lipoprotein contributions to valvular heart disease: Insights from genetic analysis. Int J Cardiol 2025; 431:133218. [PMID: 40157612 DOI: 10.1016/j.ijcard.2025.133218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/26/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND The per-particle pathogenicity of very-low-density lipoprotein (VLDL) and lipoprotein(a) [Lp(a)] with risk of valvular heart diseases (VHD) other than aortic stenosis compared with low-density lipoprotein (LDL) remains unclear. METHODS Single-nucleotide polymorphism specific clusters associated with LDL cholesterol (LDL-C), VLDL cholesterol (VLDL-C) and Lp(a) were identified. The relationships of genetically predicted variation in apolipoprotein B (apoB) in these lipoproteins with risk of VHD and its major types (aortic stenosis, aortic regurgitation, and mitral regurgitation) were evaluated to determine the comparative pathogenicity by Mendelian randomization (MR) analyses. RESULTS The VHD odds ratio (OR) per 1 g/L higher apoB was 1.09 [95 % confidence interval (CI) 1.04-1.15] in LDL vs. 1.45 (95 % CI 1.25-1.69) in VLDL vs. 2.71 (95 % CI 1.92-3.82) in Lp(a) based on the cluster-based MR analyses. The polygenic scores for each lipoprotein weighted by apoB similarly showed a greater OR of VHD per 1 g/L apoB in VLDL [1.20 (95 % CI 1.06-1.37)] and in Lp(a) [2.54, (95 % CI 1.95-3.32)] compared with that in LDL [1.05 (95 % CI 1.01-1.08)]. Multivariable MR analyses further revealed the strong effects of VLDL-C and Lp(a) on VHD risk independent of LDL-C. In addition, significant associations between Lp(a) and all three major types of VHD were observed, while LDL and VLDL had no impact on aortic and mitral regurgitation. CONCLUSIONS VLDL and Lp(a) appear to have significantly greater per-particle pathogenicity in VHD compared to LDL. The distinct impacts of lipoproteins on different VHD subtypes suggest the inadequacy of just focusing on LDL-lowering treatment for valve disorders.
Collapse
Affiliation(s)
- Da Luo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Elias Björnson
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Xiaoying Wang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bofang Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Changwu Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yunlong Guan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shizhen Xiang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jing Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
2
|
Kramer AI, Iatan I, Brunham LR. The need for national and international registries of patients with elevated lipoprotein(a). Curr Opin Lipidol 2025; 36:105-110. [PMID: 40127096 DOI: 10.1097/mol.0000000000000982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
PURPOSE OF REVIEW Elevated lipoprotein(a) [Lp(a)] is a genetically determined independent risk factor for atherosclerotic cardiovascular disease (ASCVD). Current guidelines recommend universal testing of Lp(a) once in an individual's lifetime, with risk factor management intensification for those with elevated levels. However, there is a paucity of real-world data about how patients with elevated Lp(a) are managed and about their associated cardiovascular risk. The purpose of this review is to discuss recent progress in the establishment of registries of patients with elevated Lp(a). RECENT FINDINGS Multiple registries that include patients with elevated Lp(a) have been established in various countries. These studies will provide a snapshot of the global burden of this condition and the current patterns of treatment of this patient population. SUMMARY Elevated Lp(a) is a common but underdiagnosed risk factor for ASCVD. National and international registries are needed to expand our understanding and improve the treatment of this condition.
Collapse
Affiliation(s)
- Adam I Kramer
- Division of General Internal Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia
| | - Iulia Iatan
- Division of General Internal Medicine, Department of Medicine, McGill University Health Center, Montreal, Quebec
- Centre for Heart and Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Liam R Brunham
- Division of General Internal Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia
- Centre for Heart and Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
3
|
Anchouche K, Baass A, Thanassoulis G. Lp(a): A Clinical Review. Clin Biochem 2025; 137:110929. [PMID: 40258460 DOI: 10.1016/j.clinbiochem.2025.110929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
Elevated lipoprotein(a) (Lp[a]) is a genetically determined cardiovascular risk factor, linked to both atherosclerotic cardiovascular disease and aortic stenosis. Elevated Lp(a) is widely prevalent, and consequently, several cardiovascular societies now recommend performing Lp(a) screening at least once in all adults. While there are presently no approved drugs specifically aimed at lowering Lp(a), several promising candidates are currently in the drug development pipeline, and many of these are now undergoing late phase clinical trials. In this comprehensive review, we outline Lp(a) biology and genetics, describe Lp(a)'s relationship to various cardiovascular clinical phenotypes of interest, highlight novel Lp(a)-lowering therapies, and outline what role these may have in future clinical practice.
Collapse
Affiliation(s)
- Khalil Anchouche
- McGill University Health Centre and Research Institute, Montreal, QC, Canada; McGill University, Montreal, QC, Canada
| | - Alexis Baass
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montreal, QC, Canada; Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Montreal, QC, Canada
| | - George Thanassoulis
- McGill University Health Centre and Research Institute, Montreal, QC, Canada; McGill University, Montreal, QC, Canada.
| |
Collapse
|
4
|
Favereaux S, Durlach V, Vercoustre B. Patient Experience of Heart Disease with Elevated Lipoprotein(a): Views from a Patient, His Physician, and a Patient Association. Cardiol Ther 2025:10.1007/s40119-025-00416-6. [PMID: 40349269 DOI: 10.1007/s40119-025-00416-6] [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: 03/04/2025] [Accepted: 04/17/2025] [Indexed: 05/14/2025] Open
Abstract
This article presents three points of view on lipoprotein(a) [Lp(a)]: that of a patient, his endocrinologist, and a patient association, the Association Nationale des Hypercholestérolémies familiales et Lipoprotéines (a) (ANHET). By sharing his story, the patient reveals the severe impact his high Lp(a) levels had on his health, his daily life, and his family. The endocrinologist explains what Lp(a) is and its role as a risk factor for cardiovascular disease. As an expert in the field, he reviews the recommendations for the screening and management of Lp(a). The vice-president of ANHET describes the association's fight to increase awareness of this risk factor among patients, the medical profession, and even politicians, and to bring about changes in the healthcare system. Given the large number of people concerned, the perspectives of the patient, the physician, and the patient association converge in raising awareness of the negative impact of high levels of Lp(a) on health and the importance of intensifying Lp(a) screening.See the Supplementary Material for a French-language version of this abstract.
Collapse
Affiliation(s)
| | - Vincent Durlach
- Department of Cardiology, Reims University Hospital, Reims, France.
- Service de Cardiologie, Centre Hospitalier Universitaire de Reims, Bâtiment C, Christian Cabrol, 45 rue Cognacq-Jay, 51092, Reims, France.
| | - Bernard Vercoustre
- Association Nationale des Hypercholestérolémies familiales et Lipoprotéines (a) (AHNET), Chéry Les Pouilly, France
| |
Collapse
|
5
|
MacDougall DE, Tybjærg-Hansen A, Knowles JW, Stern TP, Hartsuff BK, McGowan MP, Baum SJ, Wilemon KA, Nordestgaard BG. Lipoprotein(a) and recurrent atherosclerotic cardiovascular events: the US Family Heart Database. Eur Heart J 2025:ehaf297. [PMID: 40331569 DOI: 10.1093/eurheartj/ehaf297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/05/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND AND AIMS Higher levels of lipoprotein(a) drive increasing risk of atherosclerotic cardiovascular disease (ASCVD) in otherwise healthy individuals regardless of sex and race/ethnicity. This study aimed to evaluate whether this is also true for recurrent ASCVD, and whether LDL cholesterol-lowering therapy possibly mitigates such a relationship. METHODS In US medical claims between 2012 and 2022 for 340 million individuals, 273 770 had diagnosed ASCVD and lipoprotein(a) measured in nmol/L. These women (n = 117 269; 43%) and men (n = 156 501; 57%) included Black (n = 22 451; 8%), Hispanic (n = 24 606; 9%), and White (n = 161 165; 59%) individuals. RESULTS Lipoprotein(a) levels were higher in women vs men and in Black vs Hispanic and White individuals. During a median follow-up of 5.4 years, 41 687 individuals (15%) experienced recurrent ASCVD. Higher lipoprotein(a) levels were associated with continuously increasing risk of recurrent ASCVD. Compared to individuals with lipoprotein(a) < 15 nmol/L, the adjusted hazard ratios for recurrent ASCVD events were 1.04 (95% confidence interval 1.01-1.07) for 15-79 nmol/L, 1.15 (1.12-1.19) for 80-179 nmol/L, 1.29 (1.25-1.33) for 180-299 nmol/L, and 1.45 (1.39-1.51) for ≥300 nmol/L. Results were similar for individual ASCVD components, and in sex, race/ethnicity, baseline ASCVD, and diabetes subgroups; however, high impact LDL cholesterol-lowering therapy possibly mitigates the deleterious effect of lipoprotein(a) ≥ 180 nmol/L, most pronounced in those on PCSK9 inhibitors. Interaction on recurrent ASCVD events between lipoprotein(a) categories and sex, race/ethnicity, baseline ASCVD, diabetes, and impact of LDL cholesterol-lowering therapy use had P-values of .61, .06, .33, .91, and 2 × 10-8, respectively. CONCLUSIONS In 273 770 individuals with ASCVD, higher lipoprotein(a) levels were associated with continuously increasing risk of recurrent ASCVD events regardless of sex and race/ethnicity that may have been partially mitigated by high impact LDL cholesterol-lowering therapy.
Collapse
Affiliation(s)
- Diane E MacDougall
- Department of Research, Family Heart Foundation, 5548 First Coast Hwy, Fernandina Beach, FL 32034, USA
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joshua W Knowles
- Division of Cardiovascular Medicine, Cardiovascular Institute and Prevention Research Center, Stanford University, Stanford, CA, USA
| | | | | | - Mary P McGowan
- Department of Research, Family Heart Foundation, 5548 First Coast Hwy, Fernandina Beach, FL 32034, USA
| | | | - Katherine A Wilemon
- Department of Research, Family Heart Foundation, 5548 First Coast Hwy, Fernandina Beach, FL 32034, USA
| | - Børge G Nordestgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| |
Collapse
|
6
|
Clarke R, Wright N, Lin K, Yu C, Walters RG, Lv J, Hill M, Kartsonaki C, Millwood IY, Bennett DA, Avery D, Yang L, Chen Y, Du H, Sherliker P, Yang X, Sun D, Li L, Qu C, Marcovina S, Collins R, Chen Z, Parish S. Causal Relevance of Lp(a) for Coronary Heart Disease and Stroke Types in East Asian and European Ancestry Populations: A Mendelian Randomization Study. Circulation 2025. [PMID: 40297899 DOI: 10.1161/circulationaha.124.072086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Elevated plasma levels of Lp(a) [lipoprotein(a)] are a causal risk factor for coronary heart disease and stroke in European individuals, but the causal relevance of Lp(a) for different stroke types and in East Asian individuals with different Lp(a) genetic architecture is uncertain. METHODS We measured plasma levels of Lp(a) in a nested case-control study of 18 174 adults (mean [SD] age, 57 [10] years; 49% female) in the China Kadoorie Biobank (CKB) and performed a genome-wide association analysis to identify genetic variants affecting Lp(a) levels, with replication in ancestry-specific subsets in UK Biobank. We further performed 2-sample Mendelian randomization analyses, associating ancestry-specific Lp(a)-associated instrumental variants derived from CKB or from published data in European individuals with risk of myocardial infarction (n=17 091), ischemic stroke (IS [n=29 233]) and its subtypes, or intracerebral hemorrhage (n=5845) in East Asian and European individuals using available data from CKB and genome-wide association analysis consortia. RESULTS In CKB observational analyses, plasma levels of Lp(a) were log-linearly and positively associated with higher risks of myocardial infarction and IS, but not with ICH. In genome-wide association analysis, we identified 29 single nucleotide polymorphisms independently associated with Lp(a) that together explained 33% of variance in Lp(a) in Chinese individuals. In UK Biobank, the lead Chinese variants identified in CKB were replicated in 1260 Chinese individuals, but explained only 10% of variance in Lp(a) in European individuals. In Mendelian randomization analyses, however, there were highly concordant effects of Lp(a) across both ancestries for all cardiovascular disease outcomes examined. In combined analyses of both ancestries, the proportional reductions in risk per 100 nmol/L lower genetically predicted Lp(a) levels for myocardial infarction were 3-fold greater than for total IS (rate ratio, 0.78 [95% CI, 0.76-0.81] versus 0.94 [0.92-0.96]), but were similar to those for large-artery IS (0.80 [0.73-0.87]; n=8134). There were weaker associations with cardioembolic IS (0.92 [95% CI, 0.86-0.98]; n=11 730), and no association with small-vessel IS (0.99 [0.91-1.07]; n=12 343) or with intracerebral hemorrhage (1.08 [0.96-1.21]; n=5845). CONCLUSIONS The effects of Lp(a) on risk of myocardial infarction and large-artery IS were comparable in East Asian and European individuals, suggesting that people with either ancestry could expect comparable proportional benefits for equivalent reductions in Lp(a), but there was little effect on other stroke types.
Collapse
Affiliation(s)
- Robert Clarke
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Neil Wright
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Kuang Lin
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Robin G Walters
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Michael Hill
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Christiana Kartsonaki
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Iona Y Millwood
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Derrick A Bennett
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Daniel Avery
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Ling Yang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Yiping Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Huaidong Du
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Paul Sherliker
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Xiaoming Yang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Chan Qu
- NCDs Prevention and Control Department, Liuyang CDC, China (C.Q.)
| | | | - Rory Collins
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Zhengming Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Sarah Parish
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| |
Collapse
|
7
|
Xu H, Wang Z, Chen D, Zhang H, Ge J, Li J. PCSK-9 Inhibitors Can Significantly Improve the Coronary Slow Flow Caused by Elevated Lipoprotein (a) in ST-Elevation Myocardial Infarction Patients With Chronic Kidney Disease. Catheter Cardiovasc Interv 2025. [PMID: 40277036 DOI: 10.1002/ccd.31543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Coronary slow flow and no reflow significantly predict poor prognosis in acute myocardial infarction (AMI) patients, especially those with chronic kidney disease (CKD). Early identification of factors contributing to these conditions can mitigate ischemic events and improve outcomes. AIMS This study aimed to investigate the association between elevated lipoprotein (a) [Lp(a)] levels and proprotein convertase subtilisin/kexin Type 9 (PCSK-9) inhibitor therapy with coronary slow flow or no reflow after percutaneous coronary intervention (PCI) in AMI patients with CKD. METHODS A total of 323 ST-elevation myocardial infarction (STEMI) patients who underwent PCI between October 2017 and June 2023 were included. Patients were divided into CKD (n = 132) and non-CKD (n = 191) groups. Lp(a) levels and the prevalence of coronary slow flow or no reflow after PCI were evaluated. STEMI patients with CKD were further categorized into elevated Lp(a) (n = 81) and normal Lp(a) (n = 51) subgroups. Logistic analysis identified risk factors for coronary slow flow/no reflow after PCI. The impact of PCSK-9 inhibitors on outcomes was also assessed in the elevated Lp(a) subgroup. RESULTS STEMI patients with CKD had significantly higher Lp(a) levels compared to those without CKD (median 36.75 vs. 15.90 mg/dL, p = 0.0001). CKD patients with elevated Lp(a) had a higher prevalence of coronary slow flow/no reflow after PCI than those with normal Lp(a) (38.3% vs. 13.7%, p = 0.002). Logistic regression analysis identified elevated Lp(a) as an independent risk factor for slow flow/no reflow after PCI in STEMI patients with CKD (OR = 2.985, p = 0.027). In CKD patients with elevated Lp(a), PCSK-9 inhibitors significantly improved post-PCI coronary flow and reduced composite cardiovascular events during 1-year follow-up (22.2% vs. 51.1%, p = 0.008). CONCLUSIONS Elevated Lp(a) is an independent risk factor for coronary slow flow or no reflow after PCI in STEMI patients with CKD. PCSK-9 inhibitors improve coronary blood flow and reduce cardiovascular events in these patients.
Collapse
Affiliation(s)
- Hao Xu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| | - Ziqing Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| | - Dan Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| | - Haojie Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| | - Junhua Ge
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| | - Jian Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| |
Collapse
|
8
|
Foffa I, Esposito A, Simonini L, Berti S, Vecoli C. Telomere Length and Clonal Hematopoiesis of Indeterminate Potential: A Loop Between Two Key Players in Aortic Valve Disease? J Cardiovasc Dev Dis 2025; 12:135. [PMID: 40278194 PMCID: PMC12027716 DOI: 10.3390/jcdd12040135] [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: 01/30/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Aortic valve stenosis (AVS) is the most common valvular heart disease that was considered, for a long time, a passive degenerative disease due to physiological aging. More recently, it has been recognized as an active, modifiable disease in which many cellular processes are involved. Nevertheless, since aging remains the major risk factor for AVS, a field of research has focused on the role of early (biological) aging and its dependent pathways in the initiation and progression of AVS. Telomeres are regions at the ends of chromosomes that are critical for maintaining genome stability in eukaryotic cells. Telomeres are the hallmarks and molecular drivers of aging and age-related degenerative pathologies. Clonal hematopoiesis of indeterminate potential (CHIP), a condition caused by somatic mutations of leukemia-associated genes in individuals without hematologic abnormalities or clonal disorders, has been reported to be associated with aging. CHIP represents a new and independent risk factor in cardiovascular diseases, including AVS. Interestingly, evidence suggests a causal link between telomere biology and CHIP in several pathological disorders. In this review, we discussed the current knowledge of telomere biology and CHIP as possible mechanisms of aortic valve degeneration. We speculated on how a better understanding of the complex relationship between telomere and CHIP might provide great potential for an early diagnosis and for developing novel medical therapies to reduce the constant increasing health burden of AVS.
Collapse
Affiliation(s)
- Ilenia Foffa
- CNR Institute of Clinical Physiology, 54100 Massa, Italy;
- Cardiology Unit, Fondazione Toscana Gabriele Monasterio, 54100 Massa, Italy; (A.E.); (S.B.)
| | - Augusto Esposito
- Cardiology Unit, Fondazione Toscana Gabriele Monasterio, 54100 Massa, Italy; (A.E.); (S.B.)
| | - Ludovica Simonini
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy;
| | - Sergio Berti
- Cardiology Unit, Fondazione Toscana Gabriele Monasterio, 54100 Massa, Italy; (A.E.); (S.B.)
| | - Cecilia Vecoli
- CNR Institute of Clinical Physiology, 54100 Massa, Italy;
- Cardiology Unit, Fondazione Toscana Gabriele Monasterio, 54100 Massa, Italy; (A.E.); (S.B.)
| |
Collapse
|
9
|
Durr MRR, Burwash IG, Lau L, Alfraidi H, Mulloy A, Tavoosi A, Arangalage D, Chan V, Lakhani M, Aljamaan A, Prosperi-Porta G, Gauda R, Beauchesne L, Mesana T, Messika-Zeitoun D. Lipoprotein (a) Distribution in Aortic Stenosis Patients: Are Lp(a) Reducing Agents the Ultimate Solution? JACC. ADVANCES 2025; 4:101662. [PMID: 40117697 PMCID: PMC11976252 DOI: 10.1016/j.jacadv.2025.101662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND There is currently no medical therapy that can slow down/stop the progression of aortic stenosis (AS). Novel drugs lowering lipoprotein(a) (Lp[a]), a proinflammatory particle linked to AS development, are currently under evaluation, but the proportion of patients with AS and elevated Lp(a) who might benefit from such therapy is not known. OBJECTIVES The authors sought to characterize the prevalence of high Lp(a) in patients with AS to determine the role of future Lp(a)-lowering therapies. METHODS In a nonselected Canadian population of AS patients seen in our specialized valve clinic, we assessed Lp(a) levels. High Lp(a) level was defined as an Lp(a) ≥100 nmol/L as per the Canadian Cardiovascular Society's Lipid Guidelines. RESULTS Lp(a) levels were measured in 162 patients (mean age: 75 ± 10 years, 43% female, mean pressure gradient: 29 ± 14 mm Hg). Mean Lp(a) was 69 ± 79 nmol/L (median: 24 nmol/L, IQR: 19-91 nmol/L), and 39 patients (24%) had a high Lp(a) level. There were no differences in the mean Lp(a) or in the proportion of high Lp(a) levels with respect to sex, age, or AS severity (all P > 0.20). CONCLUSIONS In this cross-sectional series of unselected AS patients followed in a valve clinic, only 1 in every 4 patients had a significantly elevated Lp(a) level. Novel Lp(a)-lowering therapies may have limited applicability to most patients with AS and highlight the need for further research into understanding the pathophysiology of AS and developing medical therapies targeting different pathways.
Collapse
Affiliation(s)
- Michael-Roy R Durr
- Division of Internal Medicine, Department of Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Ian G Burwash
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Lawrence Lau
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Hassan Alfraidi
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Andrew Mulloy
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Anahita Tavoosi
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Dimitri Arangalage
- Department of Cardiology, Assistance Publique-Hôpitaux de Paris, Bichat Hospital, Paris, France
| | - Vincent Chan
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Moiz Lakhani
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Alwaleed Aljamaan
- Division of Internal Medicine, Department of Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Graeme Prosperi-Porta
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Roja Gauda
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Luc Beauchesne
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Thierry Mesana
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - David Messika-Zeitoun
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada; School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
10
|
Qian X, Xu L, Geng B, Li F, Dong N. Navigating the Landscape of Translational Medicine of Calcific Aortic Valve Disease: Bridging Bench to Bedside. JACC. ASIA 2025; 5:503-515. [PMID: 40180541 DOI: 10.1016/j.jacasi.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/16/2024] [Accepted: 01/14/2025] [Indexed: 04/05/2025]
Abstract
Calcific aortic valve disease (CAVD) is a prevalent condition characterized by pathological thickening and calcification of the aortic valve, leading to increased pressure overload and cardiac remodeling, particularly in individuals aged 65 and older. This review synthesizes recent advances in understanding the pathogenesis of CAVD, focusing on key mechanisms including hemodynamic alterations, endothelial dysfunction, lipid deposition, inflammation, and fibrotic calcification. We evaluate emerging therapeutic targets based on pivotal basic research and clinical trials, highlighting the potential for mechanism-oriented interventions. Furthermore, we explore the implications of lipid-lowering therapies, anti-inflammatory strategies, and antifibrocalcific agents, as well as novel bioprosthetic designs aimed at enhancing patient outcomes. Additionally, we discuss the inherent genetic and molecular backgrounds influencing individual susceptibility to CAVD, emphasizing the promise of personalized therapy. By bridging the gap between basic science and clinical application, this review aims to guide future research efforts toward more effective prevention and treatment strategies for CAVD.
Collapse
Affiliation(s)
- Xingyu Qian
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingchuan Geng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
11
|
Khan TG, Bragazzi Cunha J, Raut C, Burroughs M, Vyas HS, Leix K, Goonewardena SN, Smrcka AV, Speliotes EK, Emmer BT. Functional interrogation of cellular Lp(a) uptake by genome-scale CRISPR screening. Atherosclerosis 2025; 403:119174. [PMID: 40174266 PMCID: PMC12011201 DOI: 10.1016/j.atherosclerosis.2025.119174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 02/04/2025] [Accepted: 03/07/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND AND AIMS An elevated level of lipoprotein(a), or Lp(a), in the bloodstream has been causally linked to the development of atherosclerotic cardiovascular disease and calcific aortic valve stenosis. Steady state levels of circulating lipoproteins are modulated by their rate of clearance, but the identity of the Lp(a) uptake receptor(s) has been controversial. METHODS We performed a genome-scale CRISPR screen to functionally interrogate all potential Lp(a) uptake regulators in HuH7 cells. Screen validation was performed by single gene disruption and overexpression. Direct binding between purified lipoproteins and recombinant protein was tested using biolayer interferometry. An association between human genetic variants and circulating Lp(a) levels was analyzed in the UK Biobank cohort. RESULTS The top positive and negative regulators of Lp(a) uptake in our screen were LDLR and MYLIP, encoding the LDL receptor and its ubiquitin ligase IDOL, respectively. We also found a significant correlation for other genes with established roles in LDLR regulation. No other gene products, including those previously proposed as Lp(a) receptors, exhibited a significant effect on Lp(a) uptake in our screen. We validated the functional influence of LDLR expression on HuH7 Lp(a) uptake, confirmed in vitro binding between the LDLR extracellular domain and purified Lp(a), and detected an association between loss-of-function LDLR variants and increased circulating Lp(a) levels in the UK Biobank cohort. CONCLUSIONS Our findings support a central role for the LDL receptor in mediating Lp(a) uptake by hepatocytes.
Collapse
Affiliation(s)
- Taslima G Khan
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, USA
| | - Juliana Bragazzi Cunha
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chinmay Raut
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael Burroughs
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Hitarthi S Vyas
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kyle Leix
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sascha N Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Elizabeth K Speliotes
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Brian T Emmer
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
12
|
Girard AS, Paulin A, Manikpurage HD, Lajeunesse E, Clavel MA, Pibarot P, Krege JH, Mathieu P, Thériault S, Arsenault BJ. Impact of Lipoprotein(a) on Valvular and Cardiovascular Outcomes in Patients With Calcific Aortic Valve Stenosis. J Am Heart Assoc 2025; 14:e038955. [PMID: 40079323 DOI: 10.1161/jaha.124.038955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/16/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Lp(a) (lipoprotein(a)) is an independent risk factor for calcific aortic valve stenosis (CAVS). Whether patients with CAVS and high Lp(a) levels are at higher risk of valvular or cardiovascular events is unknown. The aim of this study is to determine whether higher Lp(a) levels are associated with valvular and cardiovascular outcomes in patients with CAVS. METHODS AND RESULTS We identified 1962 patients from the UK Biobank with an electronic health record or self-reported CAVS diagnosis but who did not previously undergo aortic valve replacement (AVR) and had a minimal follow-up time of 2.5 years. Cox proportional hazard regression was used to evaluate the effect of Lp(a) on AVR, AVR or cardiac death, and valvular or cardiovascular events (AVR, cardiac death, myocardial infarction, stroke, heart failure, or coronary artery bypass grafting). The maximal follow-up time was set to 5 years. During the follow-up, 198 patients underwent AVR, 260 had AVR or cardiac death, and 435 had at least 1 valvular or cardiovascular event. Patients with Lp(a) levels ≥125 versus <125 nmol/L were at higher risk of AVR (hazard ratio [HR], 1.58 [95% CI, 1.17-2.12]), AVR or cardiac death (HR, 1.43 [95% CI, 1.10-1.86]), and cardiovascular or valvular events (HR, 1.36 [95% CI, 1.11-1.68]). Point estimates were comparable in men versus women, younger versus older patients, and in patients with higher versus lower plasma C-reactive protein levels. CONCLUSIONS In patients with CAVS, Lp(a) levels predicted a higher risk of valvular and cardiovascular outcomes. The impact of Lp(a)-lowering therapies on valvular and cardiovascular health should be assessed in a long-term randomized clinical trial.
Collapse
Affiliation(s)
- Arnaud S Girard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Audrey Paulin
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Hasanga D Manikpurage
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Emma Lajeunesse
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Marie-Annick Clavel
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Medicine, Faculty of Medicine Université Laval Québec QC Canada
| | - Philippe Pibarot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Medicine, Faculty of Medicine Université Laval Québec QC Canada
| | | | - Patrick Mathieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Surgery, Faculty of Medicine Université Laval Québec QC Canada
| | - Sébastien Thériault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine Université Laval Québec QC Canada
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Medicine, Faculty of Medicine Université Laval Québec QC Canada
| |
Collapse
|
13
|
Clair V, Zirille FM, Gill E. Rethinking cardiovascular risk: The emerging role of lipoprotein(a) screening. Am J Prev Cardiol 2025; 21:100945. [PMID: 40103685 PMCID: PMC11919408 DOI: 10.1016/j.ajpc.2025.100945] [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: 11/17/2024] [Revised: 01/09/2025] [Accepted: 02/07/2025] [Indexed: 03/20/2025] Open
Abstract
Lipoprotein(a) [Lp(a)] is a genetically inherited, independent risk factor for cardiovascular disease (CVD), affecting approximately 20-25% of the global population. Elevated Lp(a) levels are associated with a 2-3-fold increased risk of myocardial infarction and aortic valve stenosis, comparable to the risk seen in individuals with familial hypercholesterolemia. Despite its clinical relevance, the integration of Lp(a) screening into routine practice has been limited by inconsistent measurement techniques and a lack of targeted treatments. Recent advancements, including improved assays and the development of potential Lp(a)-lowering therapies, have renewed focus on the importance of Lp(a) screening. This review aims to clarify the role of Lp(a) in cardiovascular health by examining current evidence on who should be screened, when screening should occur, and the most accurate methods for measuring Lp(a). Key recommendations include universal, one-time screening for adults, selective screening for high-risk pediatric patients, and special considerations for individuals with conditions such as familial hypercholesterolemia and chronic kidney disease. Advances in assay technology now allow for more precise Lp(a) measurement, supporting better risk stratification. Additionally, emerging therapies that specifically target elevated Lp(a) levels could lead to more personalized management of CVD risk. Our findings support the integration of Lp(a) screening into routine cardiovascular risk assessment, highlighting its potential to improve early detection and prevention strategies across diverse patient populations.
Collapse
Affiliation(s)
- Victoria Clair
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Francis M Zirille
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Edward Gill
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
14
|
Martin SS, Aday AW, Allen NB, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Bansal N, Beaton AZ, Commodore-Mensah Y, Currie ME, Elkind MSV, Fan W, Generoso G, Gibbs BB, Heard DG, Hiremath S, Johansen MC, Kazi DS, Ko D, Leppert MH, Magnani JW, Michos ED, Mussolino ME, Parikh NI, Perman SM, Rezk-Hanna M, Roth GA, Shah NS, Springer MV, St-Onge MP, Thacker EL, Urbut SM, Van Spall HGC, Voeks JH, Whelton SP, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025; 151:e41-e660. [PMID: 39866113 DOI: 10.1161/cir.0000000000001303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2025 AHA Statistical Update is the product of a full year's worth of effort in 2024 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. This year's edition includes a continued focus on health equity across several key domains and enhanced global data that reflect improved methods and incorporation of ≈3000 new data sources since last year's Statistical Update. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
15
|
Ong JYS, Tan SML, Koh AS, Kong W, Sia CH, Yeo TC, Quek SC, Poh KK. Novel Circulating Biomarkers in Aortic Valve Stenosis. Int J Mol Sci 2025; 26:1902. [PMID: 40076529 PMCID: PMC11899762 DOI: 10.3390/ijms26051902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
The underlying pathophysiology of aortic stenosis and factors affecting its clinical progression remain poorly understood. Apart from B-type natriuretic peptide (BNP), novel and emerging biomarkers have been described in association with aortic stenosis, emphasising the potential for these biomarkers to illuminate on yet unknown mechanisms of its pathogenesis. In this review, we aimed to summarise what is known about aortic stenosis biomarkers, highlight the emerging ones, and provide a roadmap for translating these insights into clinical applications. Among the biomarkers studied, lipoprotein(a) [Lp(a)] has emerged as the most promising for risk stratification. Elevated Lp(a) levels are often associated with more rapid aortic stenosis progression. This detrimental effect is attributed to its role in promoting valve calcification. While other emerging biomarkers such as matrix metalloproteinases, monocytes, and metabolites show promises, their specific roles in aortic stenosis pathophysiology remain less clear. This may be due to their relatively recent discovery. Ongoing research aims to elucidate their mechanisms of action.
Collapse
Affiliation(s)
- Joy Yi-Shan Ong
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
| | - Sarah Ming Li Tan
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
| | - Angela S. Koh
- National Heart Centre Singapore, Singapore 169609, Singapore
- DUKE-NUS Medical School, Singapore 169857, Singapore
| | - William Kong
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Ching Hui Sia
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Tiong Cheng Yeo
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Swee Chye Quek
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Kian Keong Poh
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
16
|
Doherty S, Hernandez S, Rikhi R, Mirzai S, De Los Reyes C, McIntosh S, Block RC, Shapiro MD. Lipoprotein(a) as a Causal Risk Factor for Cardiovascular Disease. CURRENT CARDIOVASCULAR RISK REPORTS 2025; 19:8. [PMID: 39980866 PMCID: PMC11836235 DOI: 10.1007/s12170-025-00760-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 02/22/2025]
Abstract
Purpose of Review Lipoprotein(a) [Lp(a)], an atherogenic low-density lipoprotein cholesterol (LDL-C)-like molecule, has emerged as an important risk factor for the development of atherosclerotic cardiovascular disease (ASCVD). This review summarizes the evidence supporting Lp(a) as a causal risk factor for ASCVD and calcific aortic valve stenosis (CAVS). Recent Findings Lp(a) is largely (~ 90%) genetically determined and approximately 20% of the global population has elevated Lp(a). The unique structure of Lp(a) leads to proatherogenic, proinflammatory, and antifibrinolytic properties. Data from epidemiological, genome-wide association, Mendelian randomization, and meta-analyses have shown a clear association between Lp(a) and ASCVD, as well as CAVS. There are emerging data on the association between Lp(a) and ischemic stroke, peripheral arterial disease, and heart failure; however, the associations are not as strong. Summary Several lines of evidence support Lp(a) as a causal risk factor for ASCVD and CAVS. The 2024 National Lipid Association guidelines, 2022 European Atherosclerosis Society, and 2021 Canadian Cardiology Society guidelines recommend testing Lp(a) once in all adults to guide primary prevention efforts. Further studies on cardiovascular outcomes with Lp(a) targeted therapies will provide more insight on causal relationship between Lp(a) and cardiovascular disease.
Collapse
Affiliation(s)
- Sean Doherty
- Department of Internal Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Sebastian Hernandez
- Department of Internal Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Rishi Rikhi
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Department of Internal Medicine, Medical Center Boulevard, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Saeid Mirzai
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Department of Internal Medicine, Medical Center Boulevard, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Chris De Los Reyes
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, 14642 Rochester, NY USA
| | - Scott McIntosh
- Department of Public Health Sciences, Division of Public Health Sciences, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Robert C. Block
- Department of Public Health Sciences, Division of Epidemiology, Department of Medicine’s Cardiology Division, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642 USA
| | - Michael D. Shapiro
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Department of Internal Medicine, Medical Center Boulevard, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| |
Collapse
|
17
|
Blaser MC, Bäck M, Lüscher TF, Aikawa E. Calcific aortic stenosis: omics-based target discovery and therapy development. Eur Heart J 2025; 46:620-634. [PMID: 39656785 PMCID: PMC11825147 DOI: 10.1093/eurheartj/ehae829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/01/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Calcific aortic valve disease (CAVD) resulting in aortic stenosis (AS) is the most common form of valvular heart disease, affecting 2% of those over age 65. Those who develop symptomatic severe AS have an average further lifespan of <2 years without valve replacement, and three-quarters of these patients will develop heart failure, undergo valve replacement, or die within 5 years. There are no approved pharmaceutical therapies for AS, due primarily to a limited understanding of the molecular mechanisms that direct CAVD progression in the complex haemodynamic environment. Here, advances in efforts to understand the pathogenesis of CAVD and to identify putative drug targets derived from recent multi-omics studies [including (epi)genomics, transcriptomics, proteomics, and metabolomics] of blood and valvular tissues are reviewed. The recent explosion of single-cell omics-based studies in CAVD and the pathobiological and potential drug discovery insights gained from the application of omics to this disease area are a primary focus. Lastly, the translation of knowledge gained in valvular pathobiology into clinical therapies is addressed, with a particular emphasis on treatment regimens that consider sex-specific, renal, and lipid-mediated contributors to CAVD, and ongoing Phase I/II/III trials aimed at the prevention/treatment of AS are described.
Collapse
Affiliation(s)
- Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA
| | - Magnus Bäck
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Valvular and Coronary Disease, Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB 741, Boston, MA 02115, USA
| |
Collapse
|
18
|
Saboktakin Rizi S, Stamenkovic A, Ravandi A. Integrative Omics Approaches in Cardiovascular Disease Research: Current Trends and Future Directions. Can J Cardiol 2025:S0828-282X(25)00126-6. [PMID: 39952467 DOI: 10.1016/j.cjca.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/17/2025] Open
Affiliation(s)
- Shekoofeh Saboktakin Rizi
- Precision Cardiovascular Medicine Group, St Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada; Section of Cardiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Aleksandra Stamenkovic
- Precision Cardiovascular Medicine Group, St Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada; Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Amir Ravandi
- Precision Cardiovascular Medicine Group, St Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada; Section of Cardiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
19
|
Greco A, Finocchiaro S, Spagnolo M, Faro DC, Mauro MS, Raffo C, Sangiorgio G, Imbesi A, Laudani C, Mazzone PM, Ammirabile N, Giacoppo D, Landolina D, Capodanno D. Lipoprotein(a) as a Pharmacological Target: Premises, Promises, and Prospects. Circulation 2025; 151:400-415. [PMID: 39928714 DOI: 10.1161/circulationaha.124.069210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 10/29/2024] [Indexed: 02/12/2025]
Abstract
Atherosclerotic cardiovascular disease is a major health concern worldwide and requires effective preventive measures. Lp(a) (lipoprotein [a]) has recently garnered attention as an independent risk factor for astherosclerotic cardiovascular disease, with proinflammatory and prothrombotic mechanisms contributing to its atherogenicity. On an equimolar basis, Lp(a) is ~5 to 6 times more atherogenic than particles that have been widely associated with adverse cardiovascular outcomes, such as LDL (low-density lipoprotein). Lp(a) can enter the vessel wall, leading to the accumulation of oxidized phospholipids in the arterial intima, which are crucial for initiating plaque inflammation and triggering vascular disease progression. In addition, Lp(a) may cause atherothrombosis through interactions between apoA (apolipoprotein A) and the platelet PAR-1 (protease-activated receptor 1) receptor, as well as competitive inhibition of plasminogen. Because Lp(a) is mostly determined on genetic bases, a 1-time assessment in a lifetime can suffice to identify patients with elevated levels. Mendelian randomization studies and post hoc analyses of randomized trials of LDL cholesterol-lowering drugs showed a causal link between Lp(a) concentrations and cardiovascular outcomes, with therapeutic reduction of Lp(a) expected to contribute to estimated cardiovascular risk mitigation. Many Lp(a)-lowering drugs, including monoclonal antibodies, small interfering ribonucleic acids, antisense oligonucleotides, small molecules, and gene editing compounds, are at different stages of clinical investigation and show promise for clinical use. In particular, increased Lp(a) testing and treatment are expected to have a substantial impact at the population level, enabling the identification of high-risk individuals and the subsequent prevention of a large number of cardiovascular events. Ongoing phase 3 trials will further elucidate the cardiovascular benefits of Lp(a) reduction over the long term, offering potential avenues for targeted interventions and improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Antonio Greco
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Simone Finocchiaro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Marco Spagnolo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Denise Cristiana Faro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Maria Sara Mauro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Carmelo Raffo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Giuseppe Sangiorgio
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Antonino Imbesi
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Claudio Laudani
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Placido Maria Mazzone
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Nicola Ammirabile
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Daniele Giacoppo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Davide Landolina
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| |
Collapse
|
20
|
Lemešić DL, Šimičević L, Ganoci L, Gelemanović A, Šućur N, Pećin I. Association of rs3798220 Polymorphism with Cardiovascular Incidents in Individuals with Elevated Lp(a). Diagnostics (Basel) 2025; 15:404. [PMID: 40002555 PMCID: PMC11854902 DOI: 10.3390/diagnostics15040404] [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: 01/02/2025] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Lipoprotein (a) [Lp(a)] plays a significant role in atherosclerosis and cardiovascular disease (CVD). Genetic regulation of Lp(a) involves variations in the apo(a) LPA gene, as specific polymorphisms like rs10455872 and rs3798220, both linked to higher Lp(a) levels and CVD. CVD remains the leading global cause of death, with high Lp(a) levels increasingly recognized as a significant factor in younger patients with no other CVD risk factors. We aimed to evaluate the association of LPA genetic variations with Lp(a) levels and its effect on cardiovascular risk as there are existing inconsistent findings. Methods: This case-control study included 251 subjects with a median age of 52 years (interquartile range, IQR = 17) and elevated Lp(a) levels. Cases were subjects who experienced early cardiovascular incidents (women < 65, men < 55 years old), and the control group included subjects without such history. Genotyping of LPA gene polymorphisms (rs10455872 and rs3798220) was performed, and demographic data with Lp(a) levels were collected. To evaluate the association between the LPA genotypes and the risk of cardiovascular incidents (CVI), several logistic regression models were performed. The cut-off points for Lp(a) levels were determined using diagnostic test accuracy measures. Results: The rs3798220-C allele was associated with higher Lp(a) levels (288 ± 166 nmol/L in cases vs. 189 ± 102 nmol/L in controls, p < 0.001) and myocardial infarction (53% in cases vs. 36% in controls, p = 0.036). Among cases, 28.9% carried the rs3798220-C allele, compared to 18.7% in controls. The rs10455872-G allele was slightly more prevalent in controls (34.15% vs. 29.69%) but without further significant associations. In this study, the cut-off Lp(a) value of 151 nmol/L, for patients with a positive family history of early CVD, is associated with a higher chance of developing CVI. Conclusions: This study demonstrates an association between the LPA rs3798220-C allele and higher Lp(a) levels, as well as an increased risk of early onset myocardial infarction. However, the obtained association should further be evaluated at a much larger scale.
Collapse
Affiliation(s)
- Dunja Leskovar Lemešić
- Division for Metabolic Diseases, Department of Internal Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia; (N.Š.); (I.P.)
| | - Livija Šimičević
- Division for Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, 10000 Zagreb, Croatia;
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Lana Ganoci
- Division for Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, 10000 Zagreb, Croatia;
- Department of Basic and Clinical Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Andrea Gelemanović
- Mediterranean Institute for Life Sciences (MedILS), University of Split, 21000 Split, Croatia;
| | - Nediljko Šućur
- Division for Metabolic Diseases, Department of Internal Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia; (N.Š.); (I.P.)
| | - Ivan Pećin
- Division for Metabolic Diseases, Department of Internal Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia; (N.Š.); (I.P.)
- Department of Internal Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
21
|
Blaser MC, Aikawa E. Disease Drivers in Aortic Stenosis vs Atherosclerosis. JAMA Cardiol 2025; 10:109-111. [PMID: 39504008 DOI: 10.1001/jamacardio.2024.3749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Affiliation(s)
- Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
22
|
Govindarajan V, Wanna C, Johnson NP, Kolanjiyil AV, Kim H, Kitkungvan D, McPherson DM, Grande-Allen J, Chandran KB, Estrera A, Ramzy D, Prakash S. Unraveling aortic hemodynamics using fluid structure interaction: biomechanical insights into bicuspid aortic valve dynamics with multiple aortic lesions. Biomech Model Mechanobiol 2025; 24:17-27. [PMID: 39365514 DOI: 10.1007/s10237-024-01892-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
Aortic lesions, exemplified by bicuspid aortic valves (BAVs), can complicate congenital heart defects, particularly in Turner syndrome patients. The combination of BAV, dilated ascending aorta, and an elongated aortic arch presents complex hemodynamics, requiring detailed analysis for tailored treatment strategies. While current clinical decision-making relies on imaging modalities offering limited biomechanical insights, integrating high-performance computing and fluid-structure interaction algorithms with patient data enables comprehensive evaluation of diseased anatomy and planned intervention. In this study, a patient-specific workflow was utilized to biomechanically assess a Turner syndrome patient's BAV, dilated ascending aorta, and elongated arch. Results showed significant improvements in valve function (effective orifice area, EOA increased approximately twofold) and reduction in valve stress (~ 1.8-fold) following virtual commissurotomy, leading to enhanced flow dynamics and decreased viscous dissipation (~ twofold) particularly in the ascending aorta. However, increased viscous dissipation in the distal transverse aortic arch offset its local reduction in the AAo post-intervention, emphasizing the elongated arch's role in aortic hemodynamics. Our findings highlight the importance of comprehensive biomechanical evaluation and integrating patient-specific modeling with conventional imaging techniques for improved disease assessment, risk stratification, and treatment planning, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Vijay Govindarajan
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Road, Houston, TX, 77054, USA.
- Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Charles Wanna
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Road, Houston, TX, 77054, USA
| | - Nils P Johnson
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Road, Houston, TX, 77054, USA
| | | | | | - Danai Kitkungvan
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Road, Houston, TX, 77054, USA
| | - David M McPherson
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Road, Houston, TX, 77054, USA
| | | | - Krishnan B Chandran
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Road, Houston, TX, 77054, USA
- The University of Iowa, Iowa City, IA, USA
| | - Antony Estrera
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Road, Houston, TX, 77054, USA
| | - Danny Ramzy
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Road, Houston, TX, 77054, USA
| | - Siddharth Prakash
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Road, Houston, TX, 77054, USA
| |
Collapse
|
23
|
Hafiane A, Pisaturo A, Favari E, Bortnick AE. Atherosclerosis, calcific aortic valve disease and mitral annular calcification: same or different? Int J Cardiol 2025; 420:132741. [PMID: 39557087 DOI: 10.1016/j.ijcard.2024.132741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
There are similarities in the pathophysiologic mechanisms of atherosclerosis, calcific aortic valve disease (CAVD) and mitral annular calcification (MAC), however, medical treatment to slow or stop the progression of CAVD or MAC has been more elusive as compared to atherosclerosis. Atherosclerosis and CAVD share common demographic, clinical, protein, and genetic factors even more so than with MAC, which supports the possibility of shared medical therapies, though abrogating calcific extracellular vesicle shedding could be a common target for all three conditions. Herein, we summarize the overlapping and distinct pathways for further investigation, as well as key areas where additional research is needed.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Institute of the McGill University Health Centre, McGill University, Montreal, Canada.
| | | | - Elda Favari
- Department of Food and Drug, University of Parma, Parma, Italy.
| | - Anna E Bortnick
- Department of Medicine, Divisions of Cardiology and Geriatrics, and Department of Obstetrics & Gynecology and Women's Health, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, United States of America.
| |
Collapse
|
24
|
Halapas A, Cokkinos DV. Aortic Stenosis Prevention: Is a New Cardiovascular Disease Paradigm Coming of Age? J Clin Med 2025; 14:903. [PMID: 39941574 PMCID: PMC11818457 DOI: 10.3390/jcm14030903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Calcific aortic stenosis (CAS) is currently recognized as the third most frequent cardiovascular disorder in persons aged above 60 years, after atherosclerotic disease and hypertension, and together with its precursor aortic sclerosis it has been found in more than 30% of elderly individuals. CAS is an active multifactorial process characterized by a progressive fibro-calcific remodeling and thickening of the AV leaflets caused by hemodynamic flow factors, genetic factors, lipoprotein deposition, oxidation, chronic inflammation, immunomodulators, and finally osteoblastic transformation of cardiac. Herein a comprehensive state-of-the-art paper is presented regarding the underlying pathophysiological mechanisms of CAS and the potential preventive strategies as an alternative to surgical and interventional treatment.
Collapse
Affiliation(s)
- Antonios Halapas
- Department of Interventional Cardiologist and THV Program, Athens Medical Center, 11526 Athens, Greece
| | - Dennis V. Cokkinos
- Clinical, Translational and Experimental Surgery Research Centre, Biomedical Research Foundation Academy of Athens, 4, Soranou Ephessiou Str., 11527 Athens, Greece;
| |
Collapse
|
25
|
Bhatia HS, Dweck MR, Craig N, Capoulade R, Pibarot P, Trainor PJ, Whelton SP, Rikhi R, Lidani KCF, Post WS, Tsai MY, Criqui MH, Shapiro MD, Budoff MJ, DeFilippis AP, Thanassoulis G, Tsimikas S. Oxidized Phospholipids and Calcific Aortic Valvular Disease. J Am Coll Cardiol 2024; 84:2430-2441. [PMID: 39545902 DOI: 10.1016/j.jacc.2024.08.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Oxidized phospholipids (OxPLs) are carried by apolipoprotein B-100-containing lipoproteins (OxPL-apoB) including lipoprotein(a) (Lp[a]). Both OxPL-apoB and Lp(a) have been associated with calcific aortic valve disease (CAVD). OBJECTIVES This study aimed to evaluate the associations between OxPL-apoB, Lp(a) and the prevalence, incidence, and progression of CAVD. METHODS OxPL-apoB and Lp(a) were evaluated in MESA (Multi-Ethnic Study of Atherosclerosis) and a participant-level meta-analysis of 4 randomized trials of participants with established aortic stenosis (AS). In MESA, the association of OxPL-apoB and Lp(a) with aortic valve calcium (AVC) at baseline and 9.5 years was evaluated using multivariable ordinal regression models. In the meta-analysis, the association between OxPL-apoB and Lp(a) with AS progression (annualized change in peak aortic valve jet velocity) was evaluated using multivariable linear regression models. RESULTS In MESA, both OxPL-apoB and Lp(a) were associated with prevalent AVC (OR per SD: 1.19 [95% CI: 1.07-1.32] and 1.13 [95% CI: 1.01-1.27], respectively) with a significant interaction between the two (P < 0.01). Both OxPL-apoB and Lp(a) were associated with incident AVC at 9.5 years when evaluated individually (interaction P < 0.01). The OxPL-apoB∗Lp(a) interaction demonstrated higher odds of prevalent and incident AVC for OxPL-apoB with increasing Lp(a) levels. In the meta-analysis, when analyzed separately, both OxPL-apoB and Lp(a) were associated with faster increase in peak aortic valve jet velocity, but when evaluated together, only OxPL-apoB remained significant (ß: 0.07; 95% CI: 0.01-0.12). CONCLUSIONS OxPL-apoB is a predictor of the presence, incidence, and progression of AVC and established AS, particularly in the setting of elevated Lp(a) levels, and may represent a novel therapeutic target for CAVD.
Collapse
Affiliation(s)
- Harpreet S Bhatia
- Division of Cardiology, Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil Craig
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Romain Capoulade
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Philippe Pibarot
- Department of Cardiology, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Québec, Québec, Canada
| | - Patrick J Trainor
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, USA
| | - Seamus P Whelton
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Rishi Rikhi
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Karita C F Lidani
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael H Criqui
- Division of Cardiology, Department of Medicine, University of California-San Diego, La Jolla, California, USA; Division of Preventive Medicine, Department of Family Medicine, University of California-San Diego, La Jolla, California, USA
| | - Michael D Shapiro
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Matthew J Budoff
- Division of Cardiology, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Andrew P DeFilippis
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - George Thanassoulis
- Department of Medicine, Division of Experimental Medicine, McGill University Health Center, Montreal, Québec, Canada
| | - Sotirios Tsimikas
- Division of Cardiology, Department of Medicine, University of California-San Diego, La Jolla, California, USA.
| |
Collapse
|
26
|
Otto CM, Newby DE, Hillis GS. Calcific Aortic Stenosis: A Review. JAMA 2024; 332:2014-2026. [PMID: 39527048 DOI: 10.1001/jama.2024.16477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Importance Calcific aortic stenosis (AS) restricts the aortic valve opening during systole due to calcification and fibrosis of either a congenital bicuspid or a normal trileaflet aortic valve. In the US, AS affects 1% to 2% of adults older than 65 years and approximately 12% of adults older than 75 years. Worldwide, AS leads to more than 100 000 deaths annually. Observations Calcific AS is characterized by aortic valve leaflet lipid infiltration and inflammation with subsequent fibrosis and calcification. Symptoms due to severe AS, such as exercise intolerance, exertional dyspnea, and syncope, are associated with a 1-year mortality rate of up to 50% without aortic valve replacement. Echocardiography can detect AS and measure the severity of aortic valve dysfunction. Although progression rates vary, once aortic velocity is higher than 2 m/s, progression to severe AS occurs typically within 10 years. Severe AS is defined by an aortic velocity 4 m/s or higher, a mean gradient 40 mm Hg or higher, or a valve area less than or equal to 1.0 cm2. Management of mild to moderate AS and asymptomatic severe AS consists of patient education about the typical progression of disease; clinical and echocardiographic surveillance at intervals of 3 to 5 years for mild AS, 1 to 2 years for moderate AS, and 6 to 12 months for severe AS; and treatment of hypertension, hyperlipidemia, and cigarette smoking as indicated. When a patient with severe AS develops symptoms, surgical aortic valve replacement (SAVR) or transcatheter aortic valve implantation (TAVI) is recommended, which restores an average life expectancy; in patients aged older than 70 years with a low surgical risk, 10-year all-cause mortality was 62.7% with TAVI and 64.0% with SAVR. TAVI is associated with decreased length of hospitalization, more rapid return to normal activities, and less pain compared with SAVR. However, evidence supporting TAVI for patients aged younger than 65 years and long-term outcomes of TAVI are less well defined than for SAVR. For patients with symptomatic severe AS, the 2020 American College of Cardiology/American Heart Association guideline recommends SAVR for individuals aged 65 years and younger, SAVR or TAVI for those aged 66 to 79 years, and TAVI for individuals aged 80 years and older or those with an estimated surgical mortality of 8% or higher. Conclusions Calcific AS is a common chronic progressive condition among older adults and is diagnosed via echocardiography. Symptomatic patients with severe AS have a mortality rate of up to 50% after 1 year, but treatment with SAVR or TAVI reduces mortality to that of age-matched control patients. The type and timing of valve replacement should be built on evidence-based guidelines, shared decision-making, and involvement of a multidisciplinary heart valve team.
Collapse
Affiliation(s)
- Catherine M Otto
- Division of Cardiology, University of Washington School of Medicine, Seattle
| | - David E Newby
- University of Edinburgh, British Heart Foundation Centre of Research Excellence, Royal Infirmary, Edinburgh, United Kingdom
| | - Graham S Hillis
- Department of Cardiology, Royal Perth Hospital and Medical School, University of Western Australia, Perth
| |
Collapse
|
27
|
Konieczyńska M, Natorska J, Ząbczyk M, Undas A. Lipoprotein(a) and thromboembolism: current state of knowledge and unsolved issues. Arch Med Sci 2024; 20:1770-1783. [PMID: 39967936 PMCID: PMC11831339 DOI: 10.5114/aoms/197357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 02/20/2025] Open
Abstract
Lipoprotein(a) [Lp(a)], a low-density lipoprotein-like particle containing a highly polymorphic apolipoprotein(a) [apo(a)] homologous in > 80% to plasminogen, was identified as a genetically determined independent risk factor for cardiovascular disease. Elevated Lp(a) levels, found in about 20% of Europeans, are strongly linked to higher rates of myocardial infarction, major adverse cardiac events, accelerated plaque progression, ischemic stroke (especially in younger adults), and calcific aortic valve disease. However, its role in venous thromboembolism, including atypical locations like cerebral and retinal vein thrombosis, remains controversial despite several shared mechanisms underlying arterial and venous thromboembolism. The most robust evidence supports antifibrinolytic properties of elevated Lp(a), particularly smaller apo(a) isoforms, which inhibit plasminogen activation mainly by interacting with the tissue-type plasminogen activator, plasminogen, and fibrin. Other prothrombotic mechanisms include increased synthesis of plasminogen activator inhibitor (PAI-1), formation of denser fibrin networks composed of thinner fibers, less susceptible to lysis, increased platelet activation, enhanced oxidation of phospholipids leading to a low-grade proinflammatory state, upregulated tissue factor expression, and suppression of tissue factor pathway inhibitor. Targeted Lp(a) lowering therapies are currently being tested in randomized clinical trials and could potentially have clinically relevant antithrombotic effects, evidenced by the reduced risk of thromboembolism. This review summarizes the available data on the prothrombotic and antifibrinolytic actions of Lp(a), along with clinical evidence for the increased risk of thromboembolic events related to elevated Lp(a). It also introduces new concepts to explain discrepant clinical results regarding venous events, highlighting the impact of oxidized phospholipids on a prothrombotic state under conditions of high Lp(a).
Collapse
Affiliation(s)
- Małgorzata Konieczyńska
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- The John Paul II Hospital, Krakow, Poland
| | - Joanna Natorska
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- The John Paul II Hospital, Krakow, Poland
| | - Michał Ząbczyk
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- The John Paul II Hospital, Krakow, Poland
| | - Anetta Undas
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- The John Paul II Hospital, Krakow, Poland
| |
Collapse
|
28
|
Nicholls SJ, Nelson AJ, Michael LF. Oral agents for lowering lipoprotein(a). Curr Opin Lipidol 2024; 35:275-280. [PMID: 39329200 DOI: 10.1097/mol.0000000000000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
PURPOSE OF REVIEW To review the development of oral agents to lower Lp(a) levels as an approach to reducing cardiovascular risk, with a focus on recent advances in the field. RECENT FINDINGS Extensive evidence implicates Lp(a) in the causal pathway of atherosclerotic cardiovascular disease and calcific aortic stenosis. There are currently no therapies approved for lowering of Lp(a). The majority of recent therapeutic advances have focused on development of injectable agents that target RNA and inhibit synthesis of apo(a). Muvalaplin is the first, orally administered, small molecule inhibitor of Lp(a), which acts by disrupting binding of apo(a) and apoB, in clinical development. Nonhuman primate and early human studies have demonstrated the ability of muvalaplin to produce dose-dependent lowering of Lp(a). Ongoing clinical trials will evaluate the impact of muvalaplin in high cardiovascular risk and will ultimately need to determine whether this strategy lowers the rate of cardiovascular events. SUMMARY Muvalaplin is the first oral agent, developed to lower Lp(a) levels. The ability of muvalaplin to reduce cardiovascular risk remains to be investigated, in order to determine whether it will be a useful agent for the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Stephen J Nicholls
- From the Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Adam J Nelson
- From the Victorian Heart Institute, Monash University, Melbourne, Australia
| | | |
Collapse
|
29
|
Jose A, Fernando JJ, Kienesberger PC. Lysophosphatidic acid metabolism and signaling in heart disease. Can J Physiol Pharmacol 2024; 102:685-696. [PMID: 38968609 DOI: 10.1139/cjpp-2024-0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid that is mainly produced by the secreted lysophospholipase D, autotaxin (ATX), and signals through at least six G protein-coupled receptors (LPA1-6). Extracellular LPA is degraded through lipid phosphate phosphatases (LPP1, LPP2, and LPP3) at the plasmamembrane, terminating LPA receptor signaling. The ATX-LPA-LPP3 pathway is critically involved in a wide range of physiological processes, including cell survival, migration, proliferation, angiogenesis, and organismal development. Similarly, dysregulation of this pathway has been linked to many pathological processes, including cardiovascular disease. This review summarizes and interprets current literature examining the regulation and role of the ATX-LPA-LPP3 axis in heart disease. Specifically, the contribution of altered LPA metabolism via ATX and LPP3 and resulting changes to LPA receptor signaling in obesity cardiomyopathy, cardiac mitochondrial dysfunction, myocardial infarction/ischemia-reperfusion injury, hypertrophic cardiomyopathy, and aortic valve stenosis is discussed.
Collapse
Affiliation(s)
- Anu Jose
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Jeffy J Fernando
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Petra C Kienesberger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| |
Collapse
|
30
|
Sohlman M, Jauhiainen R, Vangipurapu J, Laakso A, Ala-Korpela M, Kuulasmaa T, Kuusisto J. Biomarkers reflecting insulin resistance increase the risk of aortic stenosis in a population-based study of 10,144 Finnish men. Ann Med 2024; 56:2419996. [PMID: 39593205 DOI: 10.1080/07853890.2024.2419996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 11/28/2024] Open
Abstract
AIMS To investigate a comprehensive panel of biomarkers and risk of aortic stenosis (AS) in a prospective population-based study. METHODS Anthropometric, metabolic, and inflammatory biomarkers were measured in the Metabolic Syndrome in the Men Study of 10,144 Finnish men without AS at baseline. Cases of AS were identified from the medical records. Cox regression analysis was used to identify variables predicting AS over a follow-up time of 10.8 years. Principal component (PC) analysis was applied to the biomarkers that predicted AS. Cox regression analysis was used to investigate the resulting PCs as AS predictors. RESULTS AS was diagnosed in 116 men (1.1%), with a median age of 62 years. In Cox regression analyses, fasting, 30 min, and 120 min plasma insulin, and proinsulin, with hazard ratios (HR) ranging from 1.38 (1.12-1.69, p = 2.1E-3) to 1.44 (1.23-1.68, p = 4.0E-6), Matsuda index [HR 0.68 (0.56-0.82, p = 6.9E-5)], and serum C-peptide [HR 1.47 (1.22-1.77, p = 5.0E-5)] were associated with incident AS, in addition to age, systolic blood pressure, BMI, waist circumference, waist/hip ratio, height, body fat mass, fat-free mass, and hs-CRP, and remained significant after adjustments, or if diabetic subjects were excluded. PC 1, consisting of fasting plasma insulin, C-peptide, Matsuda index, waist/hip ratio, and urine albumin excretion, and PC 2, consisting of age, body fat mass, and systolic blood pressure, were significantly associated with AS [HRs 1.37(1.09-1.73) and 1.77 (1.45-2.17), respectively]. CONCLUSION Biomarkers reflecting insulin resistance are risk factors for AS, a novel finding indicating that insulin resistance is important in the pathogenesis of AS.
Collapse
Affiliation(s)
- Maija Sohlman
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Raimo Jauhiainen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Annamaria Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Center for Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Mika Ala-Korpela
- Systems Epidemiology, University of Oulu, Oulu, Finland
- Research Unit of Population Health, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Teemu Kuulasmaa
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Center for Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
31
|
Anchouche K, Thanassoulis G. Lp(a): A Rapidly Evolving Therapeutic Landscape. Curr Atheroscler Rep 2024; 27:7. [PMID: 39576403 DOI: 10.1007/s11883-024-01252-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2024] [Indexed: 11/24/2024]
Abstract
PURPOSE OF REVIEW Elevated lipoprotein(a) (Lp[a]) is a genetically determined cardiovascular risk factor, causally linked to both atherosclerotic coronary artery disease and aortic stenosis. Elevated Lp(a) is widely prevalent, and several cardiovascular societies now recommend performing Lp(a) screening at least once in all adults. However, there are currently no approved drugs aimed specifically at lowering Lp(a). In this review, we describe several promising Lp(a)-lowering therapies in the drug development pipeline and outline what role these may have in future clinical practice. RECENT FINDINGS Pelacarsen and olpasiran are two novel RNA-based injectable therapies which are being studied in ongoing phase 3 clinical trials, with the earliest of these to be concluded in 2025. These drugs act by degrading transcribed LPA mRNA, which would normally yield the apolipoprotein(a) constituent of Lp(a). Other candidate drugs, such as Lepodisiran, Zerlasiran, and Muvalaplin, are also in early-stage development. While there are presently no Lp(a)-lowering drugs available for routine clinical use, several promising candidates are currently under investigation. If these prove to be effective in randomized clinical trials, they will expand the cardiovascular care armamentarium and will allow clinicians to treat a presently unmitigated cardiovascular risk factor.
Collapse
Affiliation(s)
- Khalil Anchouche
- McGill University Health Centre and Research Institute, Montréal, Québec, Canada
- McGill University, Montréal, Québec, Canada
| | - George Thanassoulis
- McGill University Health Centre and Research Institute, Montréal, Québec, Canada.
- McGill University, Montréal, Québec, Canada.
| |
Collapse
|
32
|
O'Toole T, Shah NP, Giamberardino SN, Kwee LC, Voora D, McGarrah RW, Ferencik M, Lu MT, Kraus WE, Foldyna B, Douglas PS, Shah SH, Pagidipati NJ. Association Between Lipoprotein(a) and Obstructive Coronary Artery Disease and High-Risk Plaque: Insights From the PROMISE Trial. Am J Cardiol 2024; 231:40-47. [PMID: 39245334 PMCID: PMC11524762 DOI: 10.1016/j.amjcard.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
The role of lipoprotein (a) (Lp[a]) in the development of obstructive coronary artery disease (CAD) and high-risk plaque (HRP) in primary prevention patients with stable chest pain is unknown. We sought to evaluate the relation of Lp(a), independent of low-density lipoprotein cholesterol (LDL-C), with the presence of obstructive CAD and HRP to improve understanding of the residual risk imparted by Lp(a) on CAD. We performed a secondary analysis in Prospective Multicenter Imaging Study for Evaluation of Chest Pain (PROMISE) Trial participants who had coronary computed tomographic angiography (CTA) performed and Lp(a) data available. Lp(a) concentration was analyzed as a binary variable, with elevated Lp(a) defined as ≥50 mg/100 ml. "Stenosis ≥50%" was defined as ≥50% coronary artery stenosis in any epicardial vessel, and "stenosis ≥70%" was defined as ≥70% coronary artery stenosis in any epicardial vessel and/or ≥50% left main coronary artery stenosis. HRP was defined as presence of plaque on CTA imaging with evidence of positive remodeling, low computed tomography attenuation, or napkin-ring sign. Multivariate logistic regression models were constructed to evaluate the association between Lp(a) and the outcomes of obstructive CAD and HRP stratified by LDL-C ≥100 versus <100 mg/100 ml. Of the 1,815 patients who underwent CTA and had Lp(a) data available, those with elevated Lp(a) were more commonly women and Black than those with lower Lp(a). Elevated Lp(a) was associated with stenosis ≥50% (odds ratio 1.57, 95% confidence interval 1.14 to 2.15, p = 0.005) and stenosis ≥70% (odds ratio 2.05, 95% confidence interval 1.34 to 3.11, p = 0.0008) in the multivariate models, and this relation was not modified by LDL-C ≥100 versus <100 mg/100 ml (interaction p >0.4). Elevated Lp(a) was not associated with HRP when adjusted for obstructive CAD. This study of patients without known CAD found that elevated Lp(a) ≥50 mg/100 ml was independently associated with the presence of obstructive CAD regardless of controlled versus uncontrolled LDL-C but was not independently associated with HRP when stenosis ≥50% or ≥70% was accounted for. Further research is warranted to delineate the role of Lp(a) in the residual risk for atherosclerotic cardiovascular disease that patients may have despite optimal LDL-C lowering.
Collapse
Affiliation(s)
- Thomas O'Toole
- Duke Clinical Research Institute; Department of Medicine
| | - Nishant P Shah
- Duke Clinical Research Institute; Department of Medicine
| | | | - Lydia Coulter Kwee
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | | | - Robert W McGarrah
- Department of Medicine; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | - Maros Ferencik
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Dept. of Radiology, Massachusetts General Hospital - Harvard Medical School, Boston, Massachusetts
| | - William E Kraus
- Department of Medicine; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Dept. of Radiology, Massachusetts General Hospital - Harvard Medical School, Boston, Massachusetts
| | | | - Svati H Shah
- Duke Clinical Research Institute; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | | |
Collapse
|
33
|
Morita H, Hoshiga M. Fibroblast Growth Factors in Cardiovascular Disease. J Atheroscler Thromb 2024; 31:1496-1511. [PMID: 39168622 PMCID: PMC11537794 DOI: 10.5551/jat.rv22025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 08/23/2024] Open
Abstract
Despite advancements in managing traditional cardiovascular risk factors, many cardiovascular diseases (CVDs) persist. Fibroblast growth factors (FGFs) have emerged as potential diagnostic markers and therapeutic targets for CVDs. FGF1, FGF2, and FGF4 are primarily used for therapeutic angiogenesis. Clinical applications are being explored based on animal studies using approaches such as recombinant protein administration and adenovirus-mediated gene delivery, targeting patients with coronary artery disease and lower extremity arterial disease. Although promising results have been observed in animal models and early-stage clinical trials, further studies are required to assess their therapeutic potential. The FGF19 subfamily, consisting of FGF19, FGF21, and FGF23, act via endocrine signaling in various organs. FGF19, primarily expressed in the small intestine, plays important roles in glucose, lipid, and bile acid metabolism and has therapeutic potential for metabolic disorders. FGF21, found in various tissues, improves glucose metabolism and insulin sensitivity, suggesting potential for treating obesity and diabetes. FGF23, primarily secreted by osteocytes, regulates vitamin D and phosphate metabolism and serves as an important biomarker for chronic kidney disease and CVDs. Thus, FGFs holds promise for both therapeutic and diagnostic applications in metabolic and cardiovascular diseases. Understanding the mechanisms of FGF may pave the way for novel strategies to prevent and manage CVDs, potentially addressing the limitations of current treatments. This review explores the roles of FGF1, FGF2, FGF4, and the FGF19 subfamily in maintaining cardiovascular health. Further research and clinical trials are crucial to fully understand the therapeutic potential of FGFs in managing cardiovascular health.
Collapse
Affiliation(s)
- Hideaki Morita
- Department of Cardiology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Masaaki Hoshiga
- Department of Cardiology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
34
|
Thomas PE, Vedel-Krogh S, Kamstrup PR. High lipoprotein(a) is a risk factor for peripheral artery disease, abdominal aortic aneurysms, and major adverse limb events. Curr Opin Cardiol 2024; 39:511-519. [PMID: 39356276 DOI: 10.1097/hco.0000000000001168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
PURPOSE OF REVIEW To summarize evidence from recent studies of high lipoprotein(a) as a risk factor for peripheral artery disease (PAD), abdominal aortic aneurysms (AAA), and major adverse limb events (MALE). Additionally, provide clinicians with 10-year absolute risk charts enabling risk prediction of PAD and AAA by lipoprotein(a) levels and conventional risk factors. RECENT FINDINGS Numerous studies support high lipoprotein(a) as an independent risk factor for PAD, AAA, and MALE. The strongest evidence is from the Copenhagen General Population Study (CGPS) and the UK Biobank, two large general population-based cohorts. In the CGPS, a 50 mg/dl higher genetically determined lipoprotein(a) associated with hazard ratios of 1.39 (1.24-1.56) for PAD and 1.21 (1.01-1.44) for AAA. Corresponding hazard ratio in the UK Biobank were 1.38 (1.30-1.46) and 1.42 (1.28-1.59). In CGPS participants with levels at least 99th (≥143 mg/dl) vs, less than 50th percentile (≤9 mg/dl), hazard ratios were 2.99 (2.09-4.30) for PAD and 2.22 (1.21-4.07) for AAA, with a corresponding incidence rate ratio for MALE of 3.04 (1.55-5.98) in participants with PAD. SUMMARY Evidence from both observational and genetic studies support high lipoprotein(a) as a causal risk factor for PAD, AAA, and MALE, and highlight the potential of future lipoprotein(a)-lowering therapy to reduce the substantial morbidity and mortality associated with these diseases.
Collapse
Affiliation(s)
- Peter E Thomas
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Signe Vedel-Krogh
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Pia R Kamstrup
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
35
|
Ma Z, Zhong J, Tu W, Li S, Chen J. The functions of apolipoproteins and lipoproteins in health and disease. MOLECULAR BIOMEDICINE 2024; 5:53. [PMID: 39465476 PMCID: PMC11513782 DOI: 10.1186/s43556-024-00218-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
Lipoproteins and apolipoproteins are crucial in lipid metabolism, functioning as essential mediators in the transport of cholesterol and triglycerides and being closely related to the pathogenesis of multiple systems, including cardiovascular. Lipoproteins a (Lp(a)), as a unique subclass of lipoproteins, is a low-density lipoprotein(LDL)-like particle with pro-atherosclerotic and pro-inflammatory properties, displaying high heritability. More and more strong evidence points to a possible link between high amounts of Lp(a) and cardiac conditions like atherosclerotic cardiovascular disease (ASCVD) and aortic stenosis (AS), making it a risk factor for heart diseases. In recent years, Lp(a)'s role in other diseases, including neurological disorders and cancer, has been increasingly recognized. Although therapies aimed at low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) have achieved significant success, elevated Lp(a) levels remain a significant clinical management problem. Despite the limited efficacy of current lipid-lowering therapies, major clinical advances in new Lp(a)-lowering therapies have significantly advanced the field. This review, grounded in the pathophysiology of lipoproteins, seeks to summarize the wide-ranging connections between lipoproteins (such as LDL-C and HDL-C) and various diseases, alongside the latest clinical developments, special emphasis is placed on the pivotal role of Lp(a) in cardiovascular disease, while also examining its future potential and mechanisms in other conditions. Furthermore, this review discusses Lp(a)-lowering therapies and highlights significant recent advances in emerging treatments, advocates for further exploration into Lp(a)'s pathogenic mechanisms and its potential as a therapeutic target, proposing new secondary prevention strategies for high-risk individuals.
Collapse
Affiliation(s)
- Zijun Ma
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, 430030, Hubei, China
| | - Wei Tu
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Shiliang Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun Chen
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China.
| |
Collapse
|
36
|
Karakasis P, Patoulias D, Giannakoulas G, Sagris M, Theofilis P, Fragakis N, Biondi-Zoccai G. Effect of Glucagon-like Peptide-1 Receptor Agonism on Aortic Valve Stenosis Risk: A Mendelian Randomization Analysis. J Clin Med 2024; 13:6411. [PMID: 39518550 PMCID: PMC11546526 DOI: 10.3390/jcm13216411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Aortic valve repair is currently the only effective treatment for calcific aortic valve stenosis (CAVS), as no pharmacological therapies exist to prevent or slow its progression. Recent promising results showed that glucagon-like peptide-1 (GLP-1) attenuates the calcification of aortic valve interstitial cells. Therefore, we conducted a two-sample Mendelian randomization analysis to investigate the effect of GLP-1 receptor agonism (GLP-1Ra) on the risk of CAVS. Methods: The inverse variance weighted (IVW) method was used to obtain the primary causal inference, and several sensitivity analyses, including MR-Egger, were performed to assess the robustness of the results. Results: Based on the IVW estimates, the GLP-1Ra showed a neutral effect on the risk of CAVS (odds ratio [OR] per 1 mmol/mol decrease in glycated hemoglobin = 0.87, 95% CI = [0.69, 1.11], p = 0.259; I2 = 4.5%, Cohran's Q = 2.09, heterogeneity p = 0.35; F statistic = 16.8). A non-significant effect was also derived by the sensitivity analyses. No evidence of horizontal pleiotropy was identified. Conclusions: GLP-1Ra was not significantly associated with the risk of CAVS. Furthermore, pragmatically designed studies are required to evaluate the effect of GLP-1Ra on the clinical course of CAVS in different patient subgroups.
Collapse
Affiliation(s)
- Paschalis Karakasis
- Second Department of Cardiology, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
- Department of Hygiene, Social-Preventive Medicine & Medical Statistics, Medical School, Aristotle University of Thessaloniki, University Campus, 54642 Thessaloniki, Greece
| | - Dimitrios Patoulias
- Second Propedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - George Giannakoulas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Marios Sagris
- School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (M.S.); (P.T.)
| | - Panagiotis Theofilis
- School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (M.S.); (P.T.)
| | - Nikolaos Fragakis
- Second Department of Cardiology, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy;
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy
| |
Collapse
|
37
|
Vassiliou VS, Johnson N, Langlands K, Tsampasian V. Genetics of Calcific Aortic Stenosis: A Systematic Review. Genes (Basel) 2024; 15:1309. [PMID: 39457433 PMCID: PMC11508093 DOI: 10.3390/genes15101309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Calcific aortic stenosis is the most prevalent valvular abnormality in the Western world. Factors commonly associated with calcific aortic stenosis include advanced age, male sex, hypertension, diabetes and impaired renal function. This review synthesises the existing literature on genetic associations with calcific aortic stenosis. Methods: A systematic search was conducted in the PubMed, Ovid and Cochrane libraries from inception to 21 July 2024 to identify human studies investigating the genetic factors involved in calcific aortic stenosis. From an initial pool of 1392 articles, 78 were selected for full-text review and 31 were included in the final qualitative synthesis. The risk of bias in these studies was assessed using the Newcastle Ottawa Scale. Results: Multiple genes have been associated with calcific aortic stenosis. These genes are involved in different biological pathways, including the lipid metabolism pathway (PLA, LDL, APO, PCSK9, Lp-PLA2, PONS1), the inflammatory pathway (IL-6, IL-10), the calcification pathway (PALMD, TEX41) and the endocrine pathway (PTH, VIT D, RUNX2, CACNA1C, ALPL). Additional genes such as NOTCH1, NAV1 and FADS1/2 influence different pathways. Mechanistically, these genes may promote a pro-inflammatory and pro-calcific environment in the aortic valve itself, leading to increased osteoblastic activity and subsequent calcific degeneration of the valve. Conclusions: Numerous genetic associations contribute to calcific aortic stenosis. Recognition of these associations can enhance risk stratification for individuals and their first-degree relatives, facilitate family screening, and importantly, pave the way for targeted therapeutic interventions focusing on the identified genetic factors. Understanding these genetic factors can also lead to gene therapy to prevent calcific aortic stenosis in the future.
Collapse
Affiliation(s)
- Vassilios S. Vassiliou
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK; (N.J.); (V.T.)
- Fitzwilliam College, University of Cambridge, Cambridge CB3 0DG, UK
| | - Nicholas Johnson
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK; (N.J.); (V.T.)
| | - Kenneth Langlands
- Institute of Continuing Education, University of Cambridge, Cambridge CB23 8AQ, UK;
| | - Vasiliki Tsampasian
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK; (N.J.); (V.T.)
| |
Collapse
|
38
|
Boffa MB, Koschinsky ML. Lipoprotein(a) and cardiovascular disease. Biochem J 2024; 481:1277-1296. [PMID: 39302109 PMCID: PMC11555715 DOI: 10.1042/bcj20240037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
Elevated plasma levels of lipoprotein(a) (Lp(a)) are a prevalent, independent, and causal risk factor for atherosclerotic cardiovascular disease and calcific aortic valve disease. Lp(a) consists of a lipoprotein particle resembling low density lipoprotein and the covalently-attached glycoprotein apolipoprotein(a) (apo(a)). Novel therapeutics that specifically and potently lower Lp(a) levels are currently in advanced stages of clinical development, including in large, phase 3 cardiovascular outcomes trials. However, fundamental unanswered questions remain concerning some key aspects of Lp(a) biosynthesis and catabolism as well as the true pathogenic mechanisms of the particle. In this review, we describe the salient biochemical features of Lp(a) and apo(a) and how they underlie the disease-causing potential of Lp(a), the factors that determine plasma Lp(a) concentrations, and the mechanism of action of Lp(a)-lowering drugs.
Collapse
Affiliation(s)
- Michael B. Boffa
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Marlys L. Koschinsky
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
39
|
Loewenstein I, Lichtenstein D, Goldiner I, Ben-Shoshan J, Halkin A, Konigstein M, Banai S, Arbel Y, Finkelstein A, Steinvil A. Lipoprotein(a) Levels in Severe Aortic Stenosis Referred for Transcatheter Aortic Valve Implantation Compared to Controls. JACC. ADVANCES 2024; 3:101264. [PMID: 39296820 PMCID: PMC11408374 DOI: 10.1016/j.jacadv.2024.101264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024]
Abstract
Background Limited observational reports link elevated lipoprotein(a) (Lp[a]) levels to aortic stenosis (AS) or to disease progression. Data on large cohorts of verified severe AS patients are lacking. Objectives The purpose of the study was to characterize Lp(a) levels of severe AS patients referred to transcatheter aortic valve implantation (TAVI) and compare them to a large cohort of Lp(a) samples derived from the general population. Methods Lp(a) levels obtained from frozen serum samples of TAVI patients between 2012 and 2017 were compared to a control group for whom Lp(a) levels were obtained for any reason and stratified by gender. Multivariable binary logistic regression analyses were conducted to investigate associations between younger age at TAVI and an Lp(a) cutoff of 50 mg/dL. Results Lp(a) levels of 503 TAVI were compared to 25,343 controls. Patients in the AS group had mildly higher median Lp(a) levels compared to controls (20.5 vs 18.7 mg/dL, P = 0.04). Lp(a) levels in males with severe AS were higher than controls (19.9 vs 16.6 mg/dL, P = 0.04). Females had a nonsignificant difference (22.1 vs 21.3 mg/dL, P = 0.87). In multivariable analysis, an Lp(a) cutoff of above 50 mg/dL was not associated with an earlier age at TAVI (beta: 1.04; 95% CI: 0.42-2.57; P = 0.94). Conclusions Median Lp(a) levels were only mildly higher in severe AS patients undergoing TAVI in comparison to a large control group, mainly driven by higher Lp(a) levels in males. Higher Lp(a) levels were not associated with an earlier age at TAVI, rejecting its association with an accelerated disease progression.
Collapse
Affiliation(s)
- Itamar Loewenstein
- Cardiology Department, Tel Aviv Sourasky Medical Center, Israel, Affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daniel Lichtenstein
- Cardiology Department, Tel Aviv Sourasky Medical Center, Israel, Affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilana Goldiner
- Division of Clinical Laboratories, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Jeremy Ben-Shoshan
- Cardiology Department, Tel Aviv Sourasky Medical Center, Israel, Affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amir Halkin
- Cardiology Department, Tel Aviv Sourasky Medical Center, Israel, Affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Maayan Konigstein
- Cardiology Department, Tel Aviv Sourasky Medical Center, Israel, Affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shmuel Banai
- Cardiology Department, Tel Aviv Sourasky Medical Center, Israel, Affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yaron Arbel
- Cardiology Department, Tel Aviv Sourasky Medical Center, Israel, Affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Finkelstein
- Cardiology Department, Tel Aviv Sourasky Medical Center, Israel, Affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arie Steinvil
- Cardiology Department, Tel Aviv Sourasky Medical Center, Israel, Affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
40
|
Kim AR, Ahn JM, Kang DY, Jun TJ, Sun BJ, Kim HJ, Kim JB, Kim DH, Park DW, Kim YH, Han KH, Park SJ. Association of Lipoprotein(a) With Severe Degenerative Aortic Valve Stenosis. JACC. ASIA 2024; 4:751-760. [PMID: 39553905 PMCID: PMC11561479 DOI: 10.1016/j.jacasi.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 11/19/2024]
Abstract
Background Lipoprotein(a) (Lp[a]) is associated with the development of aortic valve calcification. Objectives The aim of this study was to evaluate the association between the serum level of Lp(a) and the development of severe degenerative aortic stenosis (AS) and subsequent aortic valve replacement (AVR). Methods A total of 44,742 patients with Lp(a) measurements and echocardiography at baseline evaluation between 2000 and 2020 were included from a single tertiary heart center. The primary outcome was the development of severe degenerative AS, defined as a transaortic maximal velocity of ≥4.0 m/s. Results During a median follow-up period of 6.8 years (Q1-Q3: 2.3-12.4 years), severe degenerative AS was diagnosed in 472 patients (1.1%), and subsequent AVR was performed in 387 patients (0.9%). Lp(a) levels were associated with risk for severe degenerative AS, with levels of 30 to 50, 50 to 100, and >100 mg/dL demonstrating adjusted HRs of 1.02 (95% CI: 0.78-1.34; P = 0.88), 1.18 (95% CI: 0.91-1.53; P = 0.22), and 1.96 (95% CI: 1.31-2.94; P = 0.001) compared to <30 mg/dL. Similarly, the risk for AVR due to severe degenerative AS was significantly associated with higher levels of Lp(a) (>100 mg/dL) (adjusted HR: 2.05; 95% CI: 1.31-3.19; P = 0.002). Such associations were not observed in the development of severe bicuspid (P = 0.63) or rheumatic (P = 0.96) AS. Conclusions Lp(a) levels >100 mg/dL were significantly associated with risk for severe degenerative AS and subsequent AVR, regardless of the baseline severity of AS. Such associations were not observed in other etiologies of severe AS.
Collapse
Affiliation(s)
- Ah-Ram Kim
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jung-Min Ahn
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Do-Yoon Kang
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tae Joon Jun
- Big Data Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Byung Joo Sun
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ho Jin Kim
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Joon Bum Kim
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Dae-Hee Kim
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Duk-Woo Park
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Hak Kim
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ki Hoon Han
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Jung Park
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
41
|
Tsimikas S. Lp(a) and Accelerated Progression of Aortic Stenosis: A Rationale for Universal Measurement and Therapeutic Targeting. JACC. ASIA 2024; 4:761-763. [PMID: 39553910 PMCID: PMC11561483 DOI: 10.1016/j.jacasi.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
|
42
|
Takeji Y, Tada H, Taniguchi T, Sakata K, Kitai T, Shirai S, Takamura M. Current Management and Therapy of Severe Aortic Stenosis and Future Perspective. J Atheroscler Thromb 2024; 31:1353-1364. [PMID: 39111841 PMCID: PMC11456350 DOI: 10.5551/jat.rv22023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 10/04/2024] Open
Abstract
Intervention for severe aortic stenosis (AS) has dramatically progressed since the introduction of transcatheter aortic valve replacement (TAVR). Decades ago, controversies existed regarding comparing clinical outcomes between TAVR and surgical aortic valve replacement (SAVR) in various risk profiles. Recently, we discussed the durability of transcatheter heart valves and their lifetime management after aortic valve replacement (AVR). Regarding the management of AS, we discuss the appropriate timing of intervention for severe aortic stenosis, especially in asymptomatic patients. In spite of dramatic progression of intervention for AS, there are no established medications available to prevent or slow the progression of AS at present. Basic research and genome studies have suggested several targets associated with the progression of aortic valve calcification. Randomized controlled trials evaluating the efficacy of medications to prevent AS progression are ongoing, which might lead to new strategies for AS management. In this review, we summarize the current management of AS and the drugs expected to prevent the progression of AS.
Collapse
Affiliation(s)
- Yasuaki Takeji
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Hayato Tada
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tomohiko Taniguchi
- Department of Cardiovascular Medicine, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Kenji Sakata
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Takeshi Kitai
- Department of Heart Failure and Transplantation, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Shinichi Shirai
- Division of Cardiology, Kokura Memorial Hospital, Fukuoka, Japan
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
43
|
Fauvel C, Coisne A, Capoulade R, Bourg C, Diakov C, Ribeyrolles S, Jouan J, Folliguet T, Kibler M, Dreyfus J, Magne J, Bohbot Y, Pezel T, Modine T, Donal E. Unmet needs and knowledge gaps in aortic stenosis: A position paper from the Heart Valve Council of the French Society of Cardiology. Arch Cardiovasc Dis 2024; 117:590-600. [PMID: 39353805 DOI: 10.1016/j.acvd.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/19/2024] [Accepted: 06/30/2024] [Indexed: 10/04/2024]
Abstract
Nowadays, valvular heart disease remains a significant challenge among cardiovascular diseases, affecting millions of people worldwide and exerting substantial pressure on healthcare systems. Within the spectrum of valvular heart disease, aortic stenosis is the most common valvular lesion in developed countries. Despite notable advances in understanding its pathophysiological processes, improved cardiovascular imaging techniques and expanding therapeutic options in recent years, there are still unmet needs and knowledge gaps regarding aortic stenosis pathophysiology, severity assessment, management and decision-making strategy. This review, prepared on behalf of the Heart Valve Council of the French Society of Cardiology, describes these gaps and future research perspectives to improve the outcome of patients with aortic stenosis.
Collapse
Affiliation(s)
- Charles Fauvel
- Cardiology Department, Rouen University Hospital, 76000 Rouen, France
| | - Augustin Coisne
- Institut Pasteur de Lille, CHU Lille, Lille University, INSERM, 59000 Lille, France
| | - Romain Capoulade
- L'Institut du Thorax, CHU Nantes, Nantes University, CNRS, INSERM, 44007 Nantes, France
| | - Corentin Bourg
- Department of Cardiology, CHU Rennes, University of Rennes, INSERM, LTSI - UMR 1099, 35000 Rennes, France
| | | | | | - Jérome Jouan
- Department of Cardiac and Thoracic Surgery, Limoges University Teaching Hospital, 87000 Limoges, France
| | - Thierry Folliguet
- Department of Cardiac Surgery, Henri Mondor University Hospital, AP-HP, 94000 Créteil, France
| | - Marion Kibler
- Department of Cardiovascular Surgery and Medicine, New Civil Hospital, CHU Strasbourg, Strasbourg University, 67000 Strasbourg, France
| | - Julien Dreyfus
- Cardiology Department, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| | - Julien Magne
- Department of Cardiology, Dupuytren Hospital, CHU Limoges, 87000 Limoges, France; INSERM 1094, Limoges Faculty of Medicine, 87025 Limoges, France
| | - Yohann Bohbot
- Department of Cardiology, Amiens University Hospital, 80054 Amiens, France
| | - Théo Pezel
- Department of Radiology and Department of Cardiology, Lariboisière Hospital, AP-HP, Paris Cité University, 75010 Paris, France
| | - Thomas Modine
- Department of Cardiology and Cardiovascular Surgery, Haut-Lévêque Cardiological Hospital, Bordeaux University Hospital, 33604 Pessac, France
| | - Erwan Donal
- Department of Cardiology, CHU Rennes, University of Rennes, INSERM, LTSI - UMR 1099, 35000 Rennes, France.
| |
Collapse
|
44
|
Nordestgaard BG, Langsted A. Lipoprotein(a) and cardiovascular disease. Lancet 2024; 404:1255-1264. [PMID: 39278229 DOI: 10.1016/s0140-6736(24)01308-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 09/18/2024]
Abstract
One in five people are at high risk for atherosclerotic cardiovascular disease and aortic valve stenosis due to high lipoprotein(a). Lipoprotein(a) concentrations are lowest in people from east Asia, Europe, and southeast Asia, intermediate in people from south Asia, the Middle East, and Latin America, and highest in people from Africa. Concentrations are more than 90% genetically determined and 17% higher in post-menopausal women than in men. Individuals at a higher cardiovascular risk should have lipoprotein(a) concentrations measured once in their lifetime to inform those with high concentrations to adhere to a healthy lifestyle and receive medication to lower other cardiovascular risk factors. With no approved drugs to lower lipoprotein(a) concentrations, it is promising that at least five drugs in development lower concentrations by 65-98%, with three currently being tested in large cardiovascular endpoint trials. This Review covers historical perspectives, physiology and pathophysiology, genetic evidence of causality, epidemiology, role in familial hypercholesterolaemia and diabetes, management, screening, diagnosis, measurement, prevention, and future lipoprotein(a)-lowering drugs.
Collapse
Affiliation(s)
- Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Anne Langsted
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
45
|
Giussani M, Orlando A, Tassistro E, Torresani E, Lieti G, Patti I, Colombrita C, Bulgarelli I, Antolini L, Parati G, Genovesi S. Is lipoprotein(a) measurement important for cardiovascular risk stratification in children and adolescents? Ital J Pediatr 2024; 50:161. [PMID: 39227973 PMCID: PMC11373248 DOI: 10.1186/s13052-024-01732-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/17/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Elevated lipoprotein (Lp(a)) levels are associated with increased risk of atherosclerotic processes and cardiovascular events in adults. The amount of Lp(a) is mainly genetically determined. Therefore, it is important to identify individuals with elevated Lp(a) as early as possible, particularly if other cardiovascular risk factors are present. The purpose of the study was to investigate whether, in a population of children and adolescents already followed for the presence of one or more cardiovascular risk factors (elevated blood pressure (BP), and/or excess body weight, and/or dyslipidemia), the measurement of Lp(a) can be useful for better stratifying their risk profile. METHODS In a sample of 195 children and adolescents, height, body weight, waist circumference and systolic (SBP) and diastolic (DBP) BP were measured. Body Mass Index (BMI) and SBP and DBP z-scores were calculated. Plasma Lp(a), total cholesterol, high-density lipoprotein (HDL), triglycerides, glucose, insulin, uric acid and creatinine were assessed. Low-density lipoprotein (LDL) cholesterol was calculated with the Friedewald formula. High Lp(a) was defined as ≥ 75 nmol/L and high LDL cholesterol as ≥ 3.37 mmol/L. RESULTS Our sample of children and adolescents (54.4% males, mean age 11.5 years) had median LDL cholesterol and Lp(a) values equal to 2.54 (interquartile range, IQR: 2.07-3.06) mmol/L and 22 (IQR: 7.8-68.6) nmol/L respectively. 13.8% of children had LDL cholesterol ≥ 3.37 mmol/L and 22.6 Lp(a) values ≥ 75 nmol/L. Lp(a) values were higher in children of normal weight than in those with excess weight (p = 0.007), but the difference disappeared if normal weight children referred for dyslipidemia only were excluded from the analysis (p = 0.210). 69.4% of children had normal Lp(a) and LDL cholesterol values and only 6.2% showed both elevated Lp(a) and LDL cholesterol levels. However, 16.6% of the sample, despite having normal LDL cholesterol, had elevated Lp(a) values. Multivariable analyses showed a significant association of LDL cholesterol both with Lp(a) values, and with the presence of elevated Lp(a) levels. For each mmol/L increase in LDL cholesterol the risk of having an elevated Lp(a) value increased by 73%. There was an inverse correlation between BMI z-score and Lp(a). Neither BP z-scores, nor other biochemical parameters were associated with Lp(a). CONCLUSIONS In our population more than one out of five children had elevated Lp(a) values, and in about 17% of children elevated Lp(a) values were present in the absence of increased LDL cholesterol. Our results suggest that Lp(a) measurement can be useful to better define the cardiovascular risk profile in children and adolescents already followed for the presence of other cardiovascular risk factors such as elevated BP, excess body weight and high LDL cholesterol.
Collapse
Affiliation(s)
- Marco Giussani
- Istituto Auxologico Italiano, IRCCS, Via L. Ariosto 13, Milano, 20145, Italy.
| | - Antonina Orlando
- Istituto Auxologico Italiano, IRCCS, Via L. Ariosto 13, Milano, 20145, Italy
| | - Elena Tassistro
- Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Erminio Torresani
- Istituto Auxologico Italiano, IRCCS, Via L. Ariosto 13, Milano, 20145, Italy
| | - Giulia Lieti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Ilenia Patti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Claudia Colombrita
- Istituto Auxologico Italiano, IRCCS, Via L. Ariosto 13, Milano, 20145, Italy
| | - Ilaria Bulgarelli
- Istituto Auxologico Italiano, IRCCS, Via L. Ariosto 13, Milano, 20145, Italy
| | - Laura Antolini
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Bicocca Center of Bioinformatics, Biostatistics and Bioimaging (B4 Center), University of Milano-Bicocca, Monza, Italy
| | - Gianfranco Parati
- Istituto Auxologico Italiano, IRCCS, Via L. Ariosto 13, Milano, 20145, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Simonetta Genovesi
- Istituto Auxologico Italiano, IRCCS, Via L. Ariosto 13, Milano, 20145, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
46
|
Le Nezet E, Marqueze-Pouey C, Guisle I, Clavel MA. Molecular Features of Calcific Aortic Stenosis in Female and Male Patients. CJC Open 2024; 6:1125-1137. [PMID: 39525825 PMCID: PMC11544188 DOI: 10.1016/j.cjco.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/06/2024] [Indexed: 11/16/2024] Open
Abstract
Over the past 15 years, sex-related differences in aortic valve (AV) stenosis (AS) have been highlighted, affecting various aspects of AS, such as the pathophysiology, AV lesions, left ventricle remodelling, and outcomes. Female patients were found to present a more profibrotic pattern of leaflet remodelling and/or thickening, whereas male patients have a preponderance of calcification within stenosed leaflets. The understanding of these sex differences is still limited, owing to the underrepresentation of female patients in many basic and clinical research studies and trials. A better understanding of sex differences in the pathophysiology of AS may highlight new therapeutic targets that potentially could be sex-specific. This review aims to summarize sex-related differences in AS, as discovered from basic research experiments, covering aspects of the disease ranging from leaflet composition to signalling pathways, sex hormones, genetics and/or transcriptomics, and potential sex-adapted medical treatments.
Collapse
Affiliation(s)
- Emma Le Nezet
- Institut universitaire de cardiologie et pneumologie de Québec [Quebec Heart & Lung Institute], Université Laval, Québec City, Québec, Canada
| | - Chloé Marqueze-Pouey
- Institut universitaire de cardiologie et pneumologie de Québec [Quebec Heart & Lung Institute], Université Laval, Québec City, Québec, Canada
| | - Isabelle Guisle
- Institut universitaire de cardiologie et pneumologie de Québec [Quebec Heart & Lung Institute], Université Laval, Québec City, Québec, Canada
| | - Marie-Annick Clavel
- Institut universitaire de cardiologie et pneumologie de Québec [Quebec Heart & Lung Institute], Université Laval, Québec City, Québec, Canada
| |
Collapse
|
47
|
Arsenault BJ, Loganath K, Girard A, Botezatu S, Zheng KH, Tzolos E, Abdoun K, Tastet L, Capoulade R, Côté N, Craig N, Chan KL, Tam JW, Teo KK, Couture C, Clavel MA, Mathieu P, Thériault S, Stroes ESG, Newby DE, Tsimikas S, Pibarot P, Dweck MR. Lipoprotein(a) and Calcific Aortic Valve Stenosis Progression: A Systematic Review and Meta-Analysis. JAMA Cardiol 2024; 9:835-842. [PMID: 39018080 PMCID: PMC11255972 DOI: 10.1001/jamacardio.2024.1882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/17/2024] [Indexed: 07/18/2024]
Abstract
Importance There are currently no pharmacological treatments available to slow hemodynamic progression of aortic stenosis. Plasma lipoprotein(a) concentrations predict incident aortic stenosis but its association with hemodynamic progression is controversial. Objective To determine the association between plasma lipoprotein(a) concentrations and hemodynamic progression in patients with aortic stenosis. Design, Settings and Participants The study included patients with aortic stenosis from 5 longitudinal clinical studies conducted from March 2001 to March 2023 in Canada and the UK. Of 757 total patients, data on plasma lipoprotein(a) concentrations and rates of hemodynamic progression assessed by echocardiography were available for 710, who were included in this analysis. Data were analyzed from March 2023 to April 2024. Exposure Cohort-specific plasma lipoprotein(a) concentration tertiles. Main Outcomes and Measures Hemodynamic aortic stenosis progression on echocardiography as assessed by annualized change in peak aortic jet velocity, mean transvalvular gradient, and aortic valve area. Results Among the included patients, 497 (70%) were male and 213 (30%) were female. The mean (SD) age was 65.2 (13.1) years. Patients in the top lipoprotein(a) tertile demonstrated 41% (estimate, 1.41; 95% CI, 1.13-1.75) faster progression of peak aortic jet velocity and 57% (estimate, 1.57; 95% CI, 1.18-2.10) faster progression of mean transvalvular gradient than patients in the bottom tertile. There was no evidence of heterogeneity across the individual cohorts. Progression of aortic valve area was comparable between groups (estimate, 1.23; 95% CI, 0.71-2.12). Similar results were observed when plasma lipoprotein(a) concentrations were treated as a continuous variable. Conclusions and Relevance In this study, higher plasma lipoprotein(a) concentrations were associated with faster rates of hemodynamic progression in patients with aortic stenosis. Lowering plasma lipoprotein(a) concentrations warrants further investigation in the prevention and treatment of aortic stenosis.
Collapse
Affiliation(s)
- Benoit J. Arsenault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Krithika Loganath
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Arnaud Girard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
| | - Simona Botezatu
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- University of Medicine and Pharmacy Carol Davila, Cardiology Department, Euroecolab, Bucharest, Romania
| | - Kang H. Zheng
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Cardiology, Onze Lieve Vrouwe Gasthuis Hospital, Amsterdam, the Netherlands
| | - Evangelos Tzolos
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Kathia Abdoun
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
| | - Lionel Tastet
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
| | - Romain Capoulade
- Nantes Université, Centre hospitalier universitaire Nantes, Centre national de recherche scientifique, Institut national de la santé et de la recherche médicale, l’institut du thorax, Nantes, France
| | - Nancy Côté
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
| | - Neil Craig
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Kwan L. Chan
- Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - James W. Tam
- Department of Medicine, St Boniface General Hospital, Winnipeg, Manitoba, Canada
| | - Koon K. Teo
- Department of Medicine (Cardiology), McMaster University, Hamilton, Ontario, Canada
| | - Christian Couture
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
| | - Marie-Annick Clavel
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Patrick Mathieu
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Sébastien Thériault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Erik S. G. Stroes
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - David E. Newby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Sotirios Tsimikas
- Division of Cardiovascular Diseases, Department of Medicine, University of California, San Diego, La Jolla
| | - Philippe Pibarot
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Marc R. Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
48
|
Zhu L, Li N, Shi H, Shao G, Sun L. Genetic causal association between lipidomic profiles, inflammatory proteomics, and aortic stenosis: a Mendelian randomization investigation. Eur J Med Res 2024; 29:446. [PMID: 39217396 PMCID: PMC11365128 DOI: 10.1186/s40001-024-02014-z] [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: 03/25/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Aortic stenosis (AS) is a prevalent and serious valvular heart disease with a complex etiology involving genetic predispositions, lipid dysregulation, and inflammation. The specific roles of lipid and protein biomarkers in AS development are not fully elucidated. This study aimed to elucidate the causal relationships between lipidome, inflammatory proteins, and AS using Mendelian randomization (MR), identifying potential therapeutic targets. METHODS Utilizing data from large-scale genome-wide association studies (GWAS) and genome-wide protein quantitative trait loci (pQTL) studies, we conducted MR analyses on 179 plasma lipidome and 91 inflammatory proteins to assess their causal associations with AS. Our approach included Inverse Variance Weighting (IVW), Wald ratio, and robust adjusted profile score (RAPS) analyses to refine these associations. MR-Egger regression was used to address directional horizontal pleiotropy. RESULTS Our MR analysis showed that genetically predicted 50 lipids were associated with AS, including 38 as risk factors [(9 Sterol ester, 18 Phosphatidylcholine, 4 Phosphatidylethanolamine, 1 Phosphatidylinositol and 6 Triacylglycerol)] and 12 as protective. Sterol ester (27:1/17:1) emerged as the most significant risk factor with an odds ratio (OR) of 3.11. Additionally, two inflammatory proteins, fibroblast growth factor 19 (FGF19) (OR = 0.830, P = 0.015), and interleukin 6 (IL-6) (OR = 0.729, P = 1.79E-04) were significantly associated with reduced AS risk. However, a two-step MR analysis showed no significant mediated correlations between these proteins and the lipid-AS pathway. CONCLUSION This study reveals complex lipid and protein interactions in AS, identifying potential molecular targets for therapy. These results go beyond traditional lipid profiling and significantly advance our genetic and molecular understanding of AS, highlighting potential pathways for intervention and prevention.
Collapse
Affiliation(s)
- Linwen Zhu
- Department of Cardiovascular Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315041, Zhejiang, China
| | - Ni Li
- Department of Cardiovascular Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315041, Zhejiang, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Huoshun Shi
- Department of Cardiovascular Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315041, Zhejiang, China
| | - Guofeng Shao
- Department of Cardiovascular Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315041, Zhejiang, China.
| | - Lebo Sun
- Department of Cardiovascular Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315041, Zhejiang, China.
| |
Collapse
|
49
|
O'Donoghue ML, Rosenson RS, López JAG, Lepor NE, Baum SJ, Stout E, Gaudet D, Knusel B, Kuder JF, Murphy SA, Wang H, Wu Y, Shah T, Wang J, Wilmanski T, Sohn W, Kassahun H, Sabatine MS. The Off-Treatment Effects of Olpasiran on Lipoprotein(a) Lowering: OCEAN(a)-DOSE Extension Period Results. J Am Coll Cardiol 2024; 84:790-797. [PMID: 39168564 DOI: 10.1016/j.jacc.2024.05.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Olpasiran, a small interfering RNA (siRNA), blocks lipoprotein(a) (Lp(a)) production by preventing translation of apolipoprotein(a) mRNA. In phase 2, higher doses of olpasiran every 12 weeks (Q12W) reduced circulating Lp(a) by >95%. OBJECTIVES This study sought to assess the timing of return of Lp(a) to baseline after discontinuation of olpasiran, as well as longer-term safety. METHODS OCEAN(a)-DOSE (Olpasiran Trials of Cardiovascular Events And LipoproteiN[a] Reduction-DOSE Finding Study) was a phase 2, dose-finding trial that enrolled 281 participants with atherosclerotic cardiovascular disease and Lp(a) >150 nmol/L to 1 of 4 active doses of olpasiran vs placebo (10 mg, 75 mg, 225 mg Q12W, or an exploratory dose of 225 mg Q24W given subcutaneously). The last dose of olpasiran was administered at week 36; after week 48, there was an extended off-treatment follow-up period for a minimum of 24 weeks. RESULTS A total of 276 (98.2%) participants entered the off-treatment follow-up period. The median study exposure (treatment combined with off-treatment phases) was 86 weeks (Q1-Q3: 79-99 weeks). For the 75 mg Q12W dose, the off-treatment placebo-adjusted mean percent change from baseline in Lp(a) was -76.2%, -53.0%, -44.0%, and -27.9% at 60, 72, 84, and 96 weeks, respectively (all P < 0.001). The respective off-treatment changes in Lp(a) for the 225 mg Q12W dose were -84.4%, -61.6%, -52.2%, and -36.4% (all P < 0.001). During the extension follow-up phase, no new safety concerns were identified. CONCLUSIONS Olpasiran is a potent siRNA with prolonged effects on Lp(a) lowering. Participants receiving doses ≥75 mg Q12W sustained a ∼40% to 50% reduction in Lp(a) levels close to 1 year after the last dose. (Olpasiran Trials of Cardiovascular Events And LipoproteiN[a] Reduction-DOSE Finding Study [OCEAN(a)-DOSE]; NCT04270760).
Collapse
Affiliation(s)
- Michelle L O'Donoghue
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| | - Robert S Rosenson
- Icahn School of Medicine, Mount Sinai Hospital, New York, New York, USA
| | | | - Norman E Lepor
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Seth J Baum
- Flourish Research, Boca Raton, Florida, USA; Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Elmer Stout
- Crossroads Clinical Research Inc, Mooresville, North Carolina, USA
| | - Daniel Gaudet
- Department of Medicine, University of Montréal, Chicoutimi, Quebec, Canada
| | - Beat Knusel
- Global Development, Amgen, Thousand Oaks, California, USA
| | - Julia F Kuder
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sabina A Murphy
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Huei Wang
- Global Development, Amgen, Thousand Oaks, California, USA
| | - You Wu
- Global Development, Amgen, Thousand Oaks, California, USA
| | - Trupti Shah
- Global Development, Amgen, Thousand Oaks, California, USA
| | - Jingying Wang
- Global Development, Amgen, Thousand Oaks, California, USA
| | | | - Winnie Sohn
- Global Development, Amgen, Thousand Oaks, California, USA
| | | | - Marc S Sabatine
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
50
|
Alradwan I, AL Fayez N, Alomary MN, Alshehri AA, Aodah AH, Almughem FA, Alsulami KA, Aldossary AM, Alawad AO, Tawfik YMK, Tawfik EA. Emerging Trends and Innovations in the Treatment and Diagnosis of Atherosclerosis and Cardiovascular Disease: A Comprehensive Review towards Healthier Aging. Pharmaceutics 2024; 16:1037. [PMID: 39204382 PMCID: PMC11360443 DOI: 10.3390/pharmaceutics16081037] [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: 05/26/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular diseases (CVDs) are classed as diseases of aging, which are associated with an increased prevalence of atherosclerotic lesion formation caused by such diseases and is considered as one of the leading causes of death globally, representing a severe health crisis affecting the heart and blood vessels. Atherosclerosis is described as a chronic condition that can lead to myocardial infarction, ischemic cardiomyopathy, stroke, and peripheral arterial disease and to date, most pharmacological therapies mainly aim to control risk factors in patients with cardiovascular disease. Advances in transformative therapies and imaging diagnostics agents could shape the clinical applications of such approaches, including nanomedicine, biomaterials, immunotherapy, cell therapy, and gene therapy, which are emerging and likely to significantly impact CVD management in the coming decade. This review summarizes the current anti-atherosclerotic therapies' major milestones, strengths, and limitations. It provides an overview of the recent discoveries and emerging technologies in nanomedicine, cell therapy, and gene and immune therapeutics that can revolutionize CVD clinical practice by steering it toward precision medicine. CVD-related clinical trials and promising pre-clinical strategies that would significantly impact patients with CVD are discussed. Here, we review these recent advances, highlighting key clinical opportunities in the rapidly emerging field of CVD medicine.
Collapse
Affiliation(s)
- Ibrahim Alradwan
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Nojoud AL Fayez
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Mohammad N. Alomary
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Abdullah A. Alshehri
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Alhassan H. Aodah
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Fahad A. Almughem
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Khulud A. Alsulami
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Ahmad M. Aldossary
- Wellness and Preventative Medicine Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Abdullah O. Alawad
- Healthy Aging Research Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Yahya M. K. Tawfik
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Essam A. Tawfik
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| |
Collapse
|