51
|
Reyes-Soffer G, Yeang C, Michos ED, Boatwright W, Ballantyne CM. High lipoprotein(a): Actionable strategies for risk assessment and mitigation. Am J Prev Cardiol 2024; 18:100651. [PMID: 38646021 PMCID: PMC11031736 DOI: 10.1016/j.ajpc.2024.100651] [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: 12/08/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 04/23/2024] Open
Abstract
High levels of lipoprotein(a) [Lp(a)] are causal for atherosclerotic cardiovascular disease (ASCVD). Lp(a) is the most prevalent inherited dyslipidemia and strongest genetic ASCVD risk factor. This risk persists in the presence of at target, guideline-recommended, LDL-C levels and adherence to lifestyle modifications. Epidemiological and genetic evidence supporting its causal role in ASCVD and calcific aortic stenosis continues to accumulate, although various facets regarding Lp(a) biology (genetics, pathophysiology, and expression across race/ethnic groups) are not yet fully understood. The evolving nature of clinical guidelines and consensus statements recommending universal measurements of Lp(a) and the scientific data supporting its role in multiple disease states reinforce the clinical merit to start population screening for Lp(a) now. There is a current gap in the implementation of recommendations for primary and secondary cardiovascular disease (CVD) prevention in those with high Lp(a), in part due to a lack of protocols for management strategies. Importantly, targeted apolipoprotein(a) [apo(a)]-lowering therapies that reduce Lp(a) levels in patients with high Lp(a) are in phase 3 clinical development. This review focuses on the identification and clinical management of patients with high Lp(a). Specifically, we highlight the clinical value of measuring Lp(a) and its use in determining Lp(a)-associated CVD risk by providing actionable guidance, based on scientific knowledge, that can be utilized now to mitigate risk caused by high Lp(a).
Collapse
Affiliation(s)
| | - Calvin Yeang
- Department of Medicine, UC San Diego Health, CA, USA
| | - Erin D Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, MD, USA
| | | | | |
Collapse
|
52
|
Shiyovich A, Berman AN, Besser SA, Biery DW, Kaur G, Divakaran S, Singh A, Huck DM, Weber B, Plutzky J, Di Carli MF, Nasir K, Cannon C, Januzzi JL, Bhatt DL, Blankstein R. Association of Lipoprotein (a) and Standard Modifiable Cardiovascular Risk Factors With Incident Myocardial Infarction: The Mass General Brigham Lp(a) Registry. J Am Heart Assoc 2024; 13:e034493. [PMID: 38761082 PMCID: PMC11179826 DOI: 10.1161/jaha.123.034493] [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: 01/23/2024] [Accepted: 03/29/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is a robust predictor of coronary heart disease outcomes, with targeted therapies currently under investigation. We aimed to evaluate the association of high Lp(a) with standard modifiable risk factors (SMuRFs) for incident first acute myocardial infarction (AMI). METHODS AND RESULTS This retrospective study used the Mass General Brigham Lp(a) Registry, which included patients aged ≥18 years with an Lp(a) measurement between 2000 and 2019. Exclusion criteria were severe kidney dysfunction, malignant neoplasm, and prior known atherosclerotic cardiovascular disease. Diabetes, dyslipidemia, hypertension, and smoking were considered SMuRFs. High Lp(a) was defined as >90th percentile, and low Lp(a) was defined as <50th percentile. The primary outcome was fatal or nonfatal AMI. A combination of natural language processing algorithms, International Classification of Diseases (ICD) codes, and laboratory data was used to identify the outcome and covariates. A total of 6238 patients met the eligibility criteria. The median age was 54 (interquartile range, 43-65) years, and 45% were women. Overall, 23.7% had no SMuRFs, and 17.8% had ≥3 SMuRFs. Over a median follow-up of 8.8 (interquartile range, 4.2-12.8) years, the incidence of AMI increased gradually, with higher number of SMuRFs among patients with high (log-rank P=0.031) and low Lp(a) (log-rank P<0.001). Across all SMuRF subgroups, the incidence of AMI was significantly higher for patients with high Lp(a) versus low Lp(a). The risk of high Lp(a) was similar to having 2 SMuRFs. Following adjustment for confounders and number of SMuRFs, high Lp(a) remained significantly associated with the primary outcome (hazard ratio, 2.9 [95% CI, 2.0-4.3]; P<0.001). CONCLUSIONS Among patients with no prior atherosclerotic cardiovascular disease, high Lp(a) is associated with significantly higher risk for first AMI regardless of the number of SMuRFs.
Collapse
Affiliation(s)
- Arthur Shiyovich
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
- Department of RadiologyBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Adam N. Berman
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Stephanie A. Besser
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - David W. Biery
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Gurleen Kaur
- Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Sanjay Divakaran
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
- Department of RadiologyBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Avinainder Singh
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Daniel M. Huck
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
- Department of RadiologyBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Brittany Weber
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Jorge Plutzky
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Marcelo F. Di Carli
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
- Department of RadiologyBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Khurram Nasir
- Division of Cardiovascular Prevention and Wellness, Department of Cardiovascular MedicineHouston Methodist DeBakey Heart and Vascular CenterHoustonTX
| | - Christopher Cannon
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - James L. Januzzi
- Cardiology DivisionMassachusetts General Hospital, Harvard Medical School, Baim Institute for Clinical ResearchBostonMA
| | - Deepak L. Bhatt
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
- Mount Sinai HeartIcahn School of Medicine at Mount Sinai Health SystemNew YorkNY
| | - Ron Blankstein
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
- Department of RadiologyBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| |
Collapse
|
53
|
Nissen SE, Wolski K, Watts GF, Koren MJ, Fok H, Nicholls SJ, Rider DA, Cho L, Romano S, Melgaard C, Rambaran C. Single Ascending and Multiple-Dose Trial of Zerlasiran, a Short Interfering RNA Targeting Lipoprotein(a): A Randomized Clinical Trial. JAMA 2024; 331:1534-1543. [PMID: 38587822 PMCID: PMC11002768 DOI: 10.1001/jama.2024.4504] [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: 01/25/2024] [Accepted: 03/06/2024] [Indexed: 04/09/2024]
Abstract
Importance Lipoprotein(a) is a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and calcific aortic stenosis, with no pharmacological treatments approved by regulatory authorities. Objectives To assess the safety and tolerability of zerlasiran, a short interfering RNA targeting hepatic synthesis of apolipoprotein(a), and effects on serum concentrations of lipoprotein(a). Design, Setting, and Participants Single- and multiple-dose study in healthy participants and patients with stable ASCVD, respectively, with lipoprotein(a) serum concentrations greater than 150 nmol/L, conducted at 7 research sites in the US, the Netherlands, UK, and Australia between November 18, 2020, and February 8, 2023, with last follow-up on August 23, 2023. Interventions Participants were randomized to receive (1) a single subcutaneous dose of placebo (n = 8), zerlasiran 300 mg (n = 6) or 600 mg (n = 6); or (2) 2 doses of placebo (n = 9), zerlasiran 200 mg (n = 9) at a 4-week interval or 300 mg (n = 9) or 450 mg (n = 9) at an 8-week interval. Main Outcomes Measures The primary outcome was safety and tolerability. Secondary outcomes included serum levels of zerlasiran and effects on lipoprotein(a) serum concentrations. Results Among 37 patients in the multiple-dose group (mean age, 56 [SD, 10.4] years; 15 [42%] women), 36 completed the trial. Among 14 participants with extended follow-up after single doses, 13 completed the trial. There were no serious adverse events. Median baseline lipoprotein(a) concentrations in the multiple-dose group were 288 (IQR, 199-352) nmol/L. Median changes in lipoprotein(a) concentration at 365 days after single doses were 14% (IQR, 13% to 15%) for the placebo group, -30% (IQR, -51% to -18%) for the 300 mg of zerlasiran group, and -29% (IQR, -39% to -7%) for the 600-mg dose group. After 2 doses, maximal median changes in lipoprotein(a) concentration were 19 (IQR, -17 to 28) nmol/L for the placebo group, -258 (IQR, -289 to -188) nmol/L for the 200 mg of zerlasiran group, -310 (IQR, -368 to -274) nmol/L for the 300-mg dose group, and -242 (IQR, -343 to -182) nmol/L for the 450-mg dose group, with maximal median percent change of 7% (IQR, -4% to 21%), -97% (IQR, -98% to -95%), -98% (IQR, -99% to -97%), and -99% (IQR, -99% to -98%), respectively, attenuating to 0.3% (IQR, -2% to 21%), -60% (IQR, -71% to -40%), -90% (IQR, -91% to -74%), and -89% (IQR, -91% to -76%) 201 days after administration. Conclusions Zerlasiran was well tolerated and reduced lipoprotein(a) concentrations with infrequent administration. Trial Registration ClinicalTrials.gov Identifier: NCT04606602.
Collapse
Affiliation(s)
| | - Kathy Wolski
- Cleveland Clinic Center for Clinical Research, Cleveland, Ohio
| | - Gerald F. Watts
- Department of Cardiology, Royal Perth Hospital and School of Medicine, University of Western Australia, Perth, Australia
| | - Michael J. Koren
- Jacksonville Center for Clinical Research, Jacksonville, Florida
| | - Henry Fok
- Silence Therapeutics, London, United Kingdom
| | | | | | - Leslie Cho
- Cleveland Clinic Center for Clinical Research, Cleveland, Ohio
| | | | - Carrie Melgaard
- Cleveland Clinic Center for Clinical Research, Cleveland, Ohio
| | | |
Collapse
|
54
|
Pavlyha M, Li Y, Crook S, Anderson BR, Reyes-Soffer G. Race/ethnicity and socioeconomic status affect the assessment of lipoprotein(a) levels in clinical practice. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.14.24307362. [PMID: 38798532 PMCID: PMC11118621 DOI: 10.1101/2024.05.14.24307362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background and Objective High Lp(a) levels are a risk factor for ASCVD, however Lp(a) ordering in clinical practice is low. This study examines how race/ethnicity and socioeconomic status influence Lp(a) ordering. Methods This is a single center, retrospective study (2/1/2020-6/30/2023) using electronic medical records of adults with at least one ICD-10 diagnosis of ASCVD or resistant hyperlipidemia (LDL-C >160 mg/dL on statin therapy). We evaluated Lp(a) level differences among racial/ethnic groups and sexes. We also assessed associations between diagnosis type, diagnosis number, age at diagnosis, race, socioeconomic score (based on zip codes), public health coverage and presence of Lp(a) orders. Results 4% of our cohort (N=56,833) had an Lp(a) order (17.3% Hispanic, 8.7% non-Hispanic Black, 47.5% non-Hispanic White and, 27% Asian/others). Non-Hispanic Black and Hispanic patients had lower rates of Lp(a) orders (0.17%, 0.28%, respectively) when compared to non-Hispanic White patients (2.35%), p<0.001, however, their median Lp(a) levels were higher. Individuals belonging to deprived socioeconomic groups or on Medicaid, were less likely to have an Lp(a) order (RR=0.39, p<0.001 and RR=0.40, p<0.001 respectively). Certain diagnoses (carotid stenosis, family history of ASCVD and FH) and multiple diagnoses (>2) resulted in more Lp(a) orders compared to those with only one diagnosis (p<0.001). Conclusions Lp(a) ordering is low in patients with ASCVD. Non-Hispanic Black and Hispanic patients at risk are less likely to have an Lp(a) order. Individuals residing in socioeconomically deprived neighborhoods and on Medicaid are also less like have Lp(a) order. Lp(a) orders depend on the type and number of patients' diagnoses.
Collapse
|
55
|
Leistner DM, Laguna-Fernandez A, Haghikia A, Abdelwahed YS, Schatz AS, Erbay A, Roehle R, Fonseca AF, Ferber P, Landmesser U. Impact of elevated lipoprotein(a) on coronary artery disease phenotype and severity. Eur J Prev Cardiol 2024; 31:856-865. [PMID: 38348689 DOI: 10.1093/eurjpc/zwae007] [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: 06/22/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 03/19/2024]
Abstract
AIMS A thorough characterization of the relationship between elevated lipoprotein(a) [Lp(a)] and coronary artery disease (CAD) is lacking. This study aimed to quantitatively assess the association of increasing Lp(a) levels and CAD severity in a real-world population. METHODS AND RESULTS This non-interventional, cross-sectional, LipidCardio study included patients aged ≥21 years undergoing angiography (October 2016-March 2018) at a tertiary cardiology centre, who have at least one Lp(a) measurement. The association between Lp(a) and CAD severity was determined by synergy between PCI with taxus and cardiac surgery (SYNTAX)-I and Gensini scores and angiographic characteristics. Overall, 975 patients (mean age: 69.5 years) were included; 70.1% were male, 97.5% had Caucasian ancestry, and 33.2% had a family history of premature atherosclerotic cardiovascular disease. Median baseline Lp(a) level was 19.3 nmol/L. Patients were stratified by baseline Lp(a): 72.9% had < 65 nmol/L, 21.0% had ≥100 nmol/L, 17.2% had ≥125 nmol/L, and 12.9% had ≥150 nmol/L. Compared with the normal (Lp(a) < 65 nmol/L) group, elevated Lp(a) groups (e.g. ≥ 150 nmol/L) had a higher proportion of patients with prior CAD (48.4% vs. 62.7%; P < 0.01), prior coronary revascularization (39.1% vs. 51.6%; P = 0.01), prior coronary artery bypass graft (6.0% vs. 15.1%; P < 0.01), vessel(s) with lesions (68.5% vs. 81.3%; P = 0.03), diffusely narrowed vessels (10.9% vs. 16.5%; P = 0.01) or chronic total occlusion lesions (14.3% vs. 25.2%; P < 0.01), and higher median SYNTAX-I (3.0 vs. 5.5; P = 0.01) and Gensini (10.0 vs. 16.0; P < 0.01) scores. CONCLUSION Elevated Lp(a) was associated with a more severe presentation of CAD. Awareness of Lp(a) levels in patients with CAD may have implications in their clinical management.
Collapse
Affiliation(s)
- David M Leistner
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
- University Hospital Frankfurt and Wolfgang Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | | | - Arash Haghikia
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
| | - Youssef S Abdelwahed
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Anne-Sophie Schatz
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Aslihan Erbay
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- University Hospital Frankfurt and Wolfgang Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Robert Roehle
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ana F Fonseca
- Novartis Pharma AG, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Philippe Ferber
- Novartis Pharma AG, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Ulf Landmesser
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
| |
Collapse
|
56
|
Nicholls SJ. Integrating lipoprotein(a) into preventive cardiology: probably important to get the measurement right. Eur J Prev Cardiol 2024; 31:901-902. [PMID: 38296811 DOI: 10.1093/eurjpc/zwae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 01/27/2024] [Indexed: 02/02/2024]
Affiliation(s)
- Stephen J Nicholls
- Victorian Heart Institute, Monash University, 631 Blackburn Road, Melbourne, VIC 3168, Australia
| |
Collapse
|
57
|
Yu MM, Wang ML, Wang JJ, Lin BL, Zhao X, Tao XW, Chen YY, Li PY, Zhang JK, Ge JB, Jin H, Zeng MS. Association of Lipoprotein(a) Levels With Myocardial Infarction in Patients With Low-Attenuation Plaque. J Am Coll Cardiol 2024; 83:1743-1755. [PMID: 38692827 DOI: 10.1016/j.jacc.2024.03.367] [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: 01/11/2024] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Lipoprotein(a) (Lp[a]) is associated with an increased risk of myocardial infarction (MI). However, the mechanism underlying this association has yet to be fully elucidated. OBJECTIVES This multicenter study aimed to investigate whether association between Lp(a) and MI risk is reinforced by the presence of low-attenuation plaque (LAP) identified by coronary computed tomography angiography (CCTA). METHODS In a derivation cohort, a total of 5,607 patients with stable chest pain suspected of coronary artery disease who underwent CCTA and Lp(a) measurement were prospectively enrolled. In validation cohort, 1,122 patients were retrospectively collected during the same period. High Lp(a) was defined as Lp(a) ≥50 mg/dL. The primary endpoint was a composite of time to fatal or nonfatal MI. Associations were estimated using multivariable Cox proportional hazard models. RESULTS During a median follow-up of 8.2 years (Q1-Q3: 7.2-9.3 years), the elevated Lp(a) levels were associated with MI risk (adjusted HR [aHR]: 1.91; 95% CI: 1.46-2.49; P < 0.001). There was a significant interaction between Lp(a) and LAP (Pinteraction <0.001) in relation to MI risk. When stratified by the presence or absence of LAP, Lp(a) was associated with MI in patients with LAP (aHR: 3.03; 95% CI: 1.92-4.76; P < 0.001). Mediation analysis revealed that LAP mediated 73.3% (P < 0.001) for the relationship between Lp(a) and MI. The principal findings remained unchanged in the validation cohort. CONCLUSIONS Elevated Lp(a) augmented the risk of MI during 8 years of follow-up, especially in patients with LAP identified by CCTA. The presence of LAP could reinforce the relationship between Lp(a) and future MI occurrence.
Collapse
Affiliation(s)
- Meng-Meng Yu
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, China
| | - Ming-Liang Wang
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin-Jin Wang
- Department of Radiology, Suzhou Ninth People's Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Bo-Li Lin
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | | | - Yin-Yin Chen
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, China
| | - Peng-Yang Li
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jing-Kun Zhang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Jun-Bo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Hang Jin
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, China; Department of Radiology, Zhongshan Hospital (Minhang Meilong Branch), Fudan University and Shanghai Geriatric Medical Center, Shanghai, China.
| | - Meng-Su Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, Shanghai, China.
| |
Collapse
|
58
|
Bhatia HS, Becker RC, Leibundgut G, Patel M, Lacaze P, Tonkin A, Narula J, Tsimikas S. Lipoprotein(a), platelet function and cardiovascular disease. Nat Rev Cardiol 2024; 21:299-311. [PMID: 37938756 PMCID: PMC11216952 DOI: 10.1038/s41569-023-00947-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 11/09/2023]
Abstract
Lipoprotein(a) (Lp(a)) is associated with atherothrombosis through several mechanisms, including putative antifibrinolytic properties. However, genetic association studies have not demonstrated an association between high plasma levels of Lp(a) and the risk of venous thromboembolism, and studies in patients with highly elevated Lp(a) levels have shown that Lp(a) lowering does not modify the clotting properties of plasma ex vivo. Lp(a) can interact with several platelet receptors, providing biological plausibility for a pro-aggregatory effect. Observational clinical studies suggest that elevated plasma Lp(a) concentrations are associated with worse long-term outcomes in patients undergoing revascularization. Furthermore, in these patients, those with elevated plasma Lp(a) levels derive more benefit from prolonged dual antiplatelet therapy than those with normal Lp(a) levels. The ASPREE trial in healthy older individuals treated with aspirin showed a reduction in ischaemic events in those who had a single-nucleotide polymorphism in LPA that is associated with elevated Lp(a) levels in plasma, without an increase in bleeding events. In this Review, we re-examine the role of Lp(a) in the regulation of platelet function and suggest areas of research to define further the clinical relevance to cardiovascular disease of the observed associations between Lp(a) and platelet function.
Collapse
Affiliation(s)
- Harpreet S Bhatia
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA
| | - Richard C Becker
- Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Gregor Leibundgut
- Division of Cardiology, University Hospital of Basel, Basel, Switzerland
| | - Mitul Patel
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA
| | - Paul Lacaze
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Andrew Tonkin
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jagat Narula
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
59
|
Bess C, Mehta A, Joshi PH. All we need to know about lipoprotein(a). Prog Cardiovasc Dis 2024; 84:27-33. [PMID: 38759878 DOI: 10.1016/j.pcad.2024.05.007] [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: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Lipoprotein(a) [Lp(a)], a genetically determined macromolecular complex, is independently and causally associated with atherosclerotic cardiovascular disease (ASCVD) and calcific aortic stenosis via proposed proinflammatory, prothrombotic, and proatherogenic mechanisms. While Lp(a) measurement standardization issues are being resolved, several guidelines now support testing Lp(a) at least once in each adult's lifetime for ASCVD risk prediction which can foster implementation of more aggressive primary or secondary prevention therapies. Currently, there are several emerging targeted Lp(a) lowering therapies in active clinical investigation for safety and cardiovascular benefit among both primary and secondary prevention populations. First degree relatives of patients with high Lp(a) should be encouraged to undergo cascade screening. Primary prevention patients with high Lp(a) should consider obtaining a coronary calcium score for further risk estimation and to guide further ASCVD risk factor management including consideration of low dose aspirin therapy. Secondary prevention patients with high Lp(a) levels should consider adding PCSK9 inhibition to statin therapy.
Collapse
Affiliation(s)
- Courtney Bess
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern, Dallas, TX, United States of America; Parkland Health and Hospital System, Dallas, TX, United States of America
| | - Anurag Mehta
- VCU Health Pauley Health Center, Richmond, VA, United States of America
| | - Parag H Joshi
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern, Dallas, TX, United States of America; Parkland Health and Hospital System, Dallas, TX, United States of America.
| |
Collapse
|
60
|
Simistiras A, Georgiopoulos G, Delialis D, Mavraganis G, Oikonomou E, Maneta E, Loutos C, Evangelou E, Stamatelopoulos K. Association of Lipoprotein(a) with arterial stiffness: A Mendelian randomization study. Eur J Clin Invest 2024; 54:e14168. [PMID: 38239089 DOI: 10.1111/eci.14168] [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: 07/19/2023] [Accepted: 01/09/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND In this study we used Mendelian randomization (MR) to investigate the potential causal association of lipoprotein (a) [Lp(a)] levels with pulse wave velocity (PWV). METHODS Genetic variants associated with Lp(a) were retrieved from the UK Biobank GWAS (N = 290,497). A non- overlapping GWAS based on a European cohort (N = 7,000) was used to obtain genetic associations with PWV (outcome) and utilized two different measures for the same trait, brachial-ankle (baPWV) and carotid-femoral (cfPWV) PWV. We applied a two-sample MR using the inverse variance weighting method (IVW) and a series of sensitivity analyses for 170 SNPs that were selected as instrumental variables (IVs). RESULTS Our analyses do not support a causal association between Lp(a) and PWV for neither measurement [βiwv(baPWV) = -.0005, p = .8 and βiwv(cfPWV) = -.006, p = .16]. The above findings were consistent across sensitivity analyses including weighted median, mode-based estimation, MR-Egger regression and MR-PRESSO. CONCLUSION We did not find evidence indicating that Lp(a) is causally associated with PWV, the gold standard marker of arterial stiffness.
Collapse
Affiliation(s)
- Alexandros Simistiras
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, UK
| | - Dimitrios Delialis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Georgios Mavraganis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Ermioni Oikonomou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Eleni Maneta
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Christos Loutos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| |
Collapse
|
61
|
Aminorroaya A, Dhingra LS, Oikonomou EK, Saadatagah S, Thangaraj P, Vasisht Shankar S, Spatz ES, Khera R. Development and multinational validation of an algorithmic strategy for high Lp(a) screening. NATURE CARDIOVASCULAR RESEARCH 2024; 3:558-566. [PMID: 39195936 DOI: 10.1038/s44161-024-00469-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/02/2024] [Indexed: 08/29/2024]
Abstract
Elevated lipoprotein (a) (Lp(a)) is associated with premature atherosclerotic cardiovascular disease. However, fewer than 0.5% of individuals undergo Lp(a) testing, limiting the evaluation and use of novel targeted therapeutics currently under development. Here we describe the development of a machine learning model for targeted screening for elevated Lp(a) (≥150 nmol l-1) in the UK Biobank (N = 456,815), the largest cohort with protocolized Lp(a) testing. We externally validated the model in 3 large cohort studies, ARIC (N = 14,484), CARDIA (N = 4,124) and MESA (N = 4,672). The model, Algorithmic Risk Inspection for Screening Elevated Lp(a) (ARISE), reduced the number needed to test to find one individual with elevated Lp(a) by up to 67.3%, based on the probability threshold, with consistent performance across external validation cohorts. ARISE could be used to optimize screening for elevated Lp(a) using commonly available clinical features, with the potential for its deployment in electronic health records to enhance the yield of Lp(a) testing in real-world settings.
Collapse
Affiliation(s)
- Arya Aminorroaya
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Lovedeep S Dhingra
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Evangelos K Oikonomou
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Phyllis Thangaraj
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Sumukh Vasisht Shankar
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Erica S Spatz
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Center for Outcomes Research and Evaluation (CORE), Yale New Haven Hospital, New Haven, CT, USA
| | - Rohan Khera
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- Center for Outcomes Research and Evaluation (CORE), Yale New Haven Hospital, New Haven, CT, USA.
- Section of Health Informatics, Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA.
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
62
|
Sinha T, Guntha M, Mayow AH, Zin AK, Chaudhari SS, Khan MW, Kholoki S, Khan A. Impact of Elevated Lipoprotein A on Clinical Outcomes in Patients Undergoing Percutaneous Coronary Intervention: A Systematic Review and Meta-analysis. Cureus 2024; 16:e61069. [PMID: 38915979 PMCID: PMC11195316 DOI: 10.7759/cureus.61069] [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] [Accepted: 05/25/2024] [Indexed: 06/26/2024] Open
Abstract
Lipoprotein(a) (Lp(a)) is an inherited lipoprotein particle associated with increased risk of atherosclerotic cardiovascular (CV) diseases. However, its impact on outcomes after percutaneous coronary intervention (PCI) remains unclear. The objective of this study was to assess the relationship between elevated Lp(a) levels and major adverse cardiovascular events (MACEs) and other outcomes in patients undergoing PCI. We systematically searched Embase, MEDLINE/PubMed, and Web of Science for studies published from 2015 to 2024 comparing CV outcomes between patients with elevated versus non-elevated Lp(a) levels after PCI. Primary outcome was MACE. Secondary outcomes included all-cause mortality, CV mortality, stroke, myocardial infarction, and revascularization. Risk ratios (RRs) were pooled using a random-effect model. Fifteen studies with 45,059 patients were included. Patients with elevated Lp(a) had a significantly higher risk of MACE (RR 1.38, 95% confidence interval (CI) 1.23-1.56). Elevated Lp(a) was also associated with increased risks of all-cause death (RR 1.26), CV death (RR 1.58), myocardial infarction (RR 1.44), revascularization (RR 1.38), and stroke (RR 1.18). Heterogeneity was considerable for some outcomes. This meta-analysis demonstrates that elevated Lp(a) levels are associated with worse CV outcomes, including higher rates of MACE, mortality, and recurrent ischemic events in patients undergoing PCI. Novel therapeutic approaches specifically targeting Lp(a) reduction may help mitigate residual CV risk in this high-risk population.
Collapse
Affiliation(s)
- Tanya Sinha
- Internal Medicine, Tribhuvan University, Kathmandu, NPL
| | | | | | - Aung K Zin
- Internal Medicine, University of Medicine, Mandalay, Mandalay, MMR
| | - Sandipkumar S Chaudhari
- Cardiothoracic Surgery, University of Alabama at Birmingham, Birmingham, USA
- Family Medicine, University of North Dakota School of Medicine and Health Sciences, Fargo, USA
| | | | - Samer Kholoki
- Internal Medicine, La Grange Memorial Hospital, Chicago, USA
| | - Areeba Khan
- Critical Care Medicine, United Medical and Dental College, Karachi, PAK
| |
Collapse
|
63
|
Anderson JL, Knight S, Dong L, May HT, Le VT, Bair TL, Knowlton KU. Coronary Calcium Is Elevated in Patients with Myocardial Infarction without Standard Modifiable Risk Factors. J Clin Med 2024; 13:2569. [PMID: 38731098 PMCID: PMC11084599 DOI: 10.3390/jcm13092569] [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: 03/08/2024] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Objectives: Recent reports have highlighted myocardial infarction (MI) patients without standard modifiable risk factors (SMRF), noting them to be surprisingly common and to have a substantial risk of adverse outcomes. The objective of this study was to address the challenge of identifying at-risk patients without SMRF and providing preventive therapy. Methods: Patients presenting between 2001 and 2021 to Intermountain Health catheterization laboratories with a diagnosis of MI were included if they also had a coronary artery calcium (CAC) scan by computed tomography within 2 years. SMRF were defined as a clinical diagnosis or treatment of hypertension, hyperlipidemia, diabetes, or smoking. The co-primary endpoints in SMRF-less patients were: (1) proportion of patients with an elevated (>50%ile) CAC score, and (2) an indication for statin therapy (i.e., CAC ≥ 100 AU or ≥75%ile). The 60-day and long-term major adverse cardiovascular events were determined. A comparison set included MI patients with SMRF. Results: We identified 429 MI patients with a concurrent CAC scan, of which 60 had no SMRF. SMRF status did not distinguish most risk factors or interventions. No-SMRF patients had a high CAC prevalence and percentile (82% ≥ 50%ile; median, 80%ile), and 77% met criteria for preventive therapy. As expected, patients with SMRF had high CAC scores and percentiles. Outcomes were more favorable for No-SMRF status and for lower CAC scores. Conclusions: Patients without SMRF presenting with an MI have a high prevalence and percentile of CAC. Wider application of CAC scans, including in those without SMRF, is promising as a method to identify an additional at-risk population for MI and to provide primary preventive therapy.
Collapse
Affiliation(s)
- Jeffrey L. Anderson
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (S.K.); (L.D.); (H.T.M.); (V.T.L.); (T.L.B.); (K.U.K.)
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Stacey Knight
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (S.K.); (L.D.); (H.T.M.); (V.T.L.); (T.L.B.); (K.U.K.)
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Li Dong
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (S.K.); (L.D.); (H.T.M.); (V.T.L.); (T.L.B.); (K.U.K.)
| | - Heidi T. May
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (S.K.); (L.D.); (H.T.M.); (V.T.L.); (T.L.B.); (K.U.K.)
| | - Viet T. Le
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (S.K.); (L.D.); (H.T.M.); (V.T.L.); (T.L.B.); (K.U.K.)
- Rocky Mountain University of Health Professions, Provo, UT 84606, USA
| | - Tami L. Bair
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (S.K.); (L.D.); (H.T.M.); (V.T.L.); (T.L.B.); (K.U.K.)
| | - Kirk U. Knowlton
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (S.K.); (L.D.); (H.T.M.); (V.T.L.); (T.L.B.); (K.U.K.)
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
64
|
Szarek M, Bhatt DL, Miller M, Brinton EA, Jacobson TA, Tardif JC, Ballantyne CM, Mason RP, Ketchum SB, Lira Pineda A, Doyle RT, Steg PG. Lipoprotein(a) Blood Levels and Cardiovascular Risk Reduction With Icosapent Ethyl. J Am Coll Cardiol 2024; 83:1529-1539. [PMID: 38530686 DOI: 10.1016/j.jacc.2024.02.016] [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: 01/12/2024] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Elevated lipoprotein(a) (Lp[a]) concentrations are associated with increased cardiovascular event risk even in the presence of well-controlled low-density lipoprotein cholesterol levels, but few treatments are documented to reduce this residual risk. OBJECTIVES The aim of this post hoc analysis of REDUCE-IT (Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial) was to explore the cardiovascular benefit of icosapent ethyl (IPE) across a range of Lp(a) levels. METHODS A total of 8,179 participants receiving statin therapy with established cardiovascular disease or age ≥50 years with diabetes and ≥1 additional risk factor, fasting triglyceride 1.69 to 5.63 mmol/L, and low-density lipoprotein cholesterol 1.06 to 2.59 mmol/L were randomized to receive 2 g twice daily of IPE or matching placebo. Relationships between continuous baseline Lp(a) mass concentration and risk for first and total (first and subsequent) major adverse cardiovascular events (MACE) were analyzed, along with the effects of IPE on first MACE among those with Lp(a) concentrations ≥50 or <50 mg/dL. RESULTS Among 7,026 participants (86% of those randomized) with baseline Lp(a) assessments, the median concentration was 11.6 mg/dL (Q1-Q3: 5.0-37.4 mg/dL). Lp(a) had significant relationships with first and total MACE (P < 0.0001), while event reductions with IPE did not vary across the range of Lp(a) (interaction P > 0.10). IPE significantly reduced first MACE in subgroups with concentrations ≥50 and <50 mg/dL. CONCLUSIONS Baseline Lp(a) concentration was prognostic for MACE among participants with elevated triglyceride levels receiving statin therapy. Importantly, IPE consistently reduced MACE across a range of Lp(a) levels, including among those with clinically relevant elevations.
Collapse
Affiliation(s)
- Michael Szarek
- Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA; CPC Clinical Research, Aurora, Colorado, USA; State University of New York, Downstate Health Sciences University, Brooklyn, New York, USA.
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Miller
- Department of Medicine, Crescenz Veterans Affairs Medical Center and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Terry A Jacobson
- Lipid Clinic and Cardiovascular Risk Reduction Program, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Christie M Ballantyne
- Department of Medicine, Baylor College of Medicine, and the Texas Heart Institute, Houston, Texas, USA
| | | | | | | | | | - Ph Gabriel Steg
- Université Paris-Cité, INSERM-UMR1148, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, French Alliance for Cardiovascular Trials, and Institut Universitaire de France, Paris, France
| |
Collapse
|
65
|
Wang J, Xu J, Liu T, Yu C, Xu F, Wang G, Li S, Dai X. Biomechanics-mediated endocytosis in atherosclerosis. Front Cardiovasc Med 2024; 11:1337679. [PMID: 38638885 PMCID: PMC11024446 DOI: 10.3389/fcvm.2024.1337679] [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: 11/13/2023] [Accepted: 03/04/2024] [Indexed: 04/20/2024] Open
Abstract
Biomechanical forces, including vascular shear stress, cyclic stretching, and extracellular matrix stiffness, which influence mechanosensitive channels in the plasma membrane, determine cell function in atherosclerosis. Being highly associated with the formation of atherosclerotic plaques, endocytosis is the key point in molecule and macromolecule trafficking, which plays an important role in lipid transportation. The process of endocytosis relies on the mobility and tension of the plasma membrane, which is sensitive to biomechanical forces. Several studies have advanced the signal transduction between endocytosis and biomechanics to elaborate the developmental role of atherosclerosis. Meanwhile, increased plaque growth also results in changes in the structure, composition and morphology of the coronary artery that contribute to the alteration of arterial biomechanics. These cross-links of biomechanics and endocytosis in atherosclerotic plaques play an important role in cell function, such as cell phenotype switching, foam cell formation, and lipoprotein transportation. We propose that biomechanical force activates the endocytosis of vascular cells and plays an important role in the development of atherosclerosis.
Collapse
Affiliation(s)
- Jinxuan Wang
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
- Department of Cardiology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jianxiong Xu
- School of Health Management, Xihua University, Chengdu, China
| | - Tianhu Liu
- Department of Cardiology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Cardiology and Vascular Health Research Center, Chengdu Medical College, Chengdu, China
| | - Chaoping Yu
- Department of Cardiology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Cardiology and Vascular Health Research Center, Chengdu Medical College, Chengdu, China
| | - Fengcheng Xu
- Department of Cardiology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Cardiology and Vascular Health Research Center, Chengdu Medical College, Chengdu, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Shun Li
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Xiaozhen Dai
- Department of Cardiology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Cardiology and Vascular Health Research Center, Chengdu Medical College, Chengdu, China
- School of Biosciences and Technology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
66
|
Rudin S, Kriemler L, Dittrich TD, Zietz A, Schweizer J, Arnold M, Peters N, Barinka F, Jung S, Arnold M, Fischer U, Rentsch K, Christ-Crain M, Katan M, De Marchis GM. Lipoprotein(a) as a blood marker for large artery atherosclerosis stroke etiology: validation in a prospective cohort from a swiss stroke center. Swiss Med Wkly 2024; 154:3633. [PMID: 38579294 DOI: 10.57187/s.3633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] serum levels are highly genetically determined and promote atherogenesis. High Lp(a) levels are associated with increased cardiovascular morbidity. Serum Lp(a) levels have recently been associated with large artery atherosclerosis (LAA) stroke. We aimed to externally validate this association in an independent cohort. METHODS This study stems from the prospective multicentre CoRisk study (CoPeptin for Risk Stratification in Acute Stroke patients [NCT00878813]), conducted at the University Hospital Bern, Switzerland, between 2009 and 2011, in which Lp(a) plasma levels were measured within the first 24 hours after stroke onset. We assessed the association of Lp(a) with LAA stroke using multivariable logistic regression and performed interaction analyses to identify potential effect modifiers. RESULTS Of 743 patients with ischaemic stroke, 105 (14%) had LAA stroke aetiology. Lp(a) levels were higher for LAA stroke than non-LAA stroke patients (23.0 nmol/l vs 16.3 nmol/l, p = 0.01). Multivariable regression revealed an independent association of log10and#xA0;Lp(a) with LAA stroke aetiology (aOR 1.47 [95% CI 1.03and#x2013;2.09], p = 0.03). The interaction analyses showed that Lp(a) was not associated with LAA stroke aetiology among patients with diabetes. CONCLUSIONS In a well-characterised cohort of patients with ischaemic stroke, we validated the association of higher Lp(a) levels with LAA stroke aetiology, independent of traditional cardiovascular risk factors. These findings may inform randomised clinical trials investigating the effect of Lp(a) lowering agents on cardiovascular outcomes. The CoRisk (CoPeptin for Risk Stratification in Acute Patients) study is registered on ClinicalTrials.gov. REGISTRATION NUMBER NCT00878813.
Collapse
Affiliation(s)
- Salome Rudin
- Department of Neurology and Stroke Center, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Department of Internal Medicine, Hospital of Zweisimmen, Zweisimmen, Switzerland
| | - Lilian Kriemler
- Department of Neurology and Stroke Center, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Clinic for Internal Medicine, Cantonal Hospital Schaffhausen, Schaffhausen, Switzerland
| | - Tolga D Dittrich
- Department of Neurology and Stroke Center, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Department of Neurology and Stroke Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Annaelle Zietz
- Department of Neurology and Stroke Center, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Juliane Schweizer
- Department of Neurology, Stadtspital Zürich, Triemli, Zurich, Switzerland
| | - Markus Arnold
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Nils Peters
- Department of Neurology and Stroke Center, Hirslanden Hospital Zurich, Zurich, Switzerland
| | - Filip Barinka
- Department of Neurology and Stroke Center, Hirslanden Hospital Zurich, Zurich, Switzerland
| | - Simon Jung
- Department of Neurology, Inselspital, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Marcel Arnold
- Department of Neurology, Inselspital, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Urs Fischer
- Department of Neurology and Stroke Center, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Department of Neurology, Inselspital, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Katharina Rentsch
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Department of Laboratory Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Mirjam Christ-Crain
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Department of Endocrinology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Mira Katan
- Department of Neurology and Stroke Center, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Gian Marco De Marchis
- Department of Neurology and Stroke Center, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Department of Neurology and Stroke Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
67
|
Singh S, Baars DP, Aggarwal K, Desai R, Singh D, Pinto-Sietsma SJ. Association between lipoprotein (a) and risk of heart failure: A systematic review and meta-analysis of Mendelian randomization studies. Curr Probl Cardiol 2024; 49:102439. [PMID: 38301917 DOI: 10.1016/j.cpcardiol.2024.102439] [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: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Rising incidence of heart failure (HF) in the Western world despite advanced clinical care necessitate exploration of further preventive tools and strategies. Lipoprotein(a) [Lp(a)], recognized as one of the major cardiovascular risk factors has also been implicated as a risk factor for HF. However, existing evidence remains inconclusive and that has led us to perform this meta-analysis. METHODS PubMed/Medline, EMBASE and Scopus were systematically searched for studies evaluating an association of Lp(a) with occurrence of HF from inception-till November 2023. Random effects models and I2 statistics were used for pooled odds ratio (OR) and heterogeneity assessment. We performed leave one out sensitivity analyses by sequentially removing one study at a time and recalculating the pooled effect size. RESULT Our search rendered in total 360 studies and after final screening this resulted in 7 Mendelian randomization (MR) design. According to the MR analysis, increasing Lp(a) level were significantly associated with increased risk of HF (OR 1.064, 95 % CI: 1.043-1.086, I2= 97.59 %, P < 0.001). In addition, Leave-one-out sensitivity analysis showed that the effect size did not change substantially by removal of any particular study in MR studies and ORs ranged from 1.051 (when excluding Levin) to a maximum of 1.111 (when excluding Wang or Jiang), hereby confirming the association. CONCLUSION We were able to show that by meta-analysis of MR data, increasing lipoprotein (a) levels are associated with an increased risk of HF. Whether this is due to a direct effect on heart muscle contraction or whether this is due to an increased risk of ischemic cardiac disease remains to be elucidated.
Collapse
Affiliation(s)
- Sandeep Singh
- Departments of Clinical Epidemiology, Biostatistics and Bio-informatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands; Department of Vascular Medicine, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Daniël P Baars
- Department of Vascular Medicine, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | | | - Rupak Desai
- Independent Researcher, Atlanta, Georgia, United States
| | - Dyutima Singh
- Department of Cardiology, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Sara-Joan Pinto-Sietsma
- Departments of Clinical Epidemiology, Biostatistics and Bio-informatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands; Department of Vascular Medicine, Amsterdam UMC, location AMC, Amsterdam, The Netherlands.
| |
Collapse
|
68
|
Pavlyha M, Hunter M, Nowygrod R, Patel V, Morrissey N, Bajakian D, Li Y, Reyes-Soffer G. Small apolipoprotein(a) isoforms may predict primary patency following peripheral arterial revascularization. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.18.24304485. [PMID: 38562737 PMCID: PMC10984047 DOI: 10.1101/2024.03.18.24304485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background High lipoprotein (a) [Lp(a)] is associated with adverse limb events in patients undergoing lower extremity revascularization. Lp(a) levels are genetically pre-determined, with LPA gene encoding for two apolipoprotein (a) [apo(a)] isoforms. Isoform size variations are driven by the number of kringle IV type 2 (KIV-2) repeats. Lp(a) levels are inversely correlated with isoform size. In this study, we examined the role of Lp(a) levels, apo(a) size and inflammatory markers with lower extremity revascularization outcomes. Methods 25 subjects with chronic peripheral arterial disease (PAD), underwent open or endovascular lower extremity revascularization (mean age of 66.7±9.7 years; F=12, M=13; Black=8, Hispanic=5, and White=12). Pre- and post-operative medical history, self-reported symptoms, ankle brachial indices (ABIs), and lower extremity duplex ultrasounds were obtained. Plasma Lp(a), apoB100, lipid panel, and pro-inflammatory markers (IL-6, IL-18, hs-CRP, TNFα) were assayed preoperatively. Isoform size was estimated using gel electrophoresis and weighted isoform size ( wIS ) calculated based on % isoform expression. Firth logistic regression was used to examine the relationship between Lp(a) levels, and wIS with procedural outcomes: symptoms (better/worse), primary patency at 2-4 weeks, ABIs, and re-intervention within 3-6 months. We controlled for age, sex, history of diabetes, smoking, statin, antiplatelet and anticoagulation use. Results Median plasma Lp(a) level was 108 (44, 301) nmol/L. The mean apoB100 level was 168.0 ± 65.8 mg/dL. These values were not statistically different among races. We found no association between Lp(a) levels and w IS with measured plasma pro-inflammatory markers. However, smaller apo(a) wIS was associated with occlusion of the treated lesion(s) in the postoperative period [OR=1.97 (95% CI 1.01 - 3.86, p<0.05)]. The relationship of smaller apo(a) wIS with re-intervention was not as strong [OR=1.57 (95% CI 0.96 - 2.56), p=0.07]. We observed no association between wIS with patient reported symptoms or change in ABIs. Conclusions In this small study, subjects with smaller apo(a) isoform size undergoing peripheral arterial revascularization were more likely to experience occlusion in the perioperative period and/or require re-intervention. Larger cohort studies identifying the mechanism and validating these preliminary data are needed to improve understanding of long-term peripheral vascular outcomes. Key Findings 25 subjects with symptomatic PAD underwent open or endovascular lower extremity revascularization in a small cohort. Smaller apo(a) isoforms were associated with occlusion of the treated lesion(s) within 2-4 weeks [OR=1.97 (95% CI 1.01 - 3.86, p<0.05)], suggesting apo(a) isoform size as a predictor of primary patency in the early period after lower extremity intervention. Take Home Message Subjects with high Lp(a) levels, generally have smaller apo(a) isoform sizes. We find that, in this small cohort, patients undergoing peripheral arterial revascularization subjects with small isoforms are at an increased risk of treated vessel occlusion in the perioperative period. Table of Contents Summary Subjects with symptomatic PAD requiring lower extremity revascularization have high median Lp(a) levels. Individuals with smaller apo(a) weighted isoform size (wIS) have lower primary patency rates and/or require re-intervention.
Collapse
|
69
|
Berman AN, Biery DW, Besser SA, Singh A, Shiyovich A, Weber BN, Huck DM, Divakaran S, Hainer J, Kaur G, Blaha MJ, Cannon CP, Plutzky J, Januzzi JL, Booth JN, López JAG, Kent ST, Nasir K, Di Carli MF, Bhatt DL, Blankstein R. Lipoprotein(a) and Major Adverse Cardiovascular Events in Patients With or Without Baseline Atherosclerotic Cardiovascular Disease. J Am Coll Cardiol 2024; 83:873-886. [PMID: 38418000 DOI: 10.1016/j.jacc.2023.12.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/14/2023] [Accepted: 12/22/2023] [Indexed: 03/01/2024]
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is associated with an increased risk of atherosclerotic cardiovascular disease (ASCVD). However, whether the optimal Lp(a) threshold for risk assessment should differ based on baseline ASCVD status is unknown. OBJECTIVES The purpose of this study was to assess the association between Lp(a) and major adverse cardiovascular events (MACE) among patients with and without baseline ASCVD. METHODS We studied a retrospective cohort of patients with Lp(a) measured at 2 medical centers in Boston, Massachusetts, from 2000 to 2019. To assess the association of Lp(a) with incident MACE (nonfatal myocardial infarction [MI], nonfatal stroke, coronary revascularization, or cardiovascular mortality), Lp(a) percentile groups were generated with the reference group set at the first to 50th Lp(a) percentiles. Cox proportional hazards modeling was used to assess the association of Lp(a) percentile group with MACE. RESULTS Overall, 16,419 individuals were analyzed with a median follow-up of 11.9 years. Among the 10,181 (62%) patients with baseline ASCVD, individuals in the 71st to 90th percentile group had a 21% increased hazard of MACE (adjusted HR: 1.21; P < 0.001), which was similar to that of individuals in the 91st to 100th group (adjusted HR: 1.26; P < 0.001). Among the 6,238 individuals without established ASCVD, there was a continuously higher hazard of MACE with increasing Lp(a), and individuals in the 91st to 100th Lp(a) percentile group had the highest relative risk with an adjusted HR of 1.93 (P < 0.001). CONCLUSIONS In a large, contemporary U.S. cohort, elevated Lp(a) is independently associated with long-term MACE among individuals with and without baseline ASCVD. Our results suggest that the threshold for risk assessment may be different in primary vs secondary prevention cohorts.
Collapse
Affiliation(s)
- Adam N Berman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/adambermanMD
| | - David W Biery
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephanie A Besser
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Avinainder Singh
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Arthur Shiyovich
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brittany N Weber
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel M Huck
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sanjay Divakaran
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jon Hainer
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gurleen Kaur
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael J Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, Maryland, USA
| | - Christopher P Cannon
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jorge Plutzky
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | - John N Booth
- Center for Observational Research, Amgen Inc, Thousand Oaks, California, USA
| | | | - Shia T Kent
- Center for Observational Research, Amgen Inc, Thousand Oaks, California, USA
| | - Khurram Nasir
- Department of Cardiovascular Medicine, Division of Cardiovascular Prevention and Wellness, Houston Methodist DeBakey Heart and Vascular Center, Houston, Texas, USA
| | - Marcelo F Di Carli
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai Health System, New York, New York, USA. https://twitter.com/DLBHATTMD
| | - Ron Blankstein
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
70
|
Wong ND. Lipoprotein(a): Ready for Prime Time? J Am Coll Cardiol 2024; 83:887-889. [PMID: 38418001 DOI: 10.1016/j.jacc.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 03/01/2024]
Affiliation(s)
- Nathan D Wong
- Division of Cardiology, Heart Disease Prevention Program, University of California, Irvine, California USA.
| |
Collapse
|
71
|
Marco-Benedí V, Cenarro A, Laclaustra M, Calmarza P, Bea AM, Vila À, Morillas-Ariño C, Puzo J, Mediavilla Garcia JD, Fernández Alamán AI, Suárez Tembra M, Civeira F. Influence of triglyceride concentration in lipoprotein (a) as a function of dyslipidemia. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:71-77. [PMID: 38161102 DOI: 10.1016/j.arteri.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Recently, an inverse relationship between the blood concentration of lipoprotein(a) (Lp(a)) and triglycerides (TG) has been demonstrated. The larger the VLDL particle size, the greater the presence of VLDL rich in apoliprotein E and in subjects with the apoE2/E2 genotype, the lower Lp(a) concentration. The mechanism of this inverse association is unknown. The objective of this analysis was to evaluate the Lp(a)-TG association in patients treated at the lipid units included in the registry of the Spanish Society of Atherosclerosis (SEA) by comparing the different dyslipidemias. PATIENTS AND METHODS Five thousand two hundred and seventy-five subjects ≥18 years of age registered in the registry before March 31, 2023, with Lp(a) concentration data and complete lipid profile information without treatment were included. RESULTS The mean age was 53.0 ± 14.0 years, with 48% women. The 9.5% of subjects (n = 502) had diabetes and the 22.4% (n = 1184) were obese. The median TG level was 130 mg/dL (IQR 88.0-210) and Lp(a) 55.0 nmol/L (IQR 17.9-156). Lp(a) concentration showed a negative association with TG concentration when TG values exceeded 300 mg/dL. Subjects with TG > 1000 mg/dL showed the lowest level of Lp(a), 17.9 nmol/L, and subjects with TG < 300 mg/dL had a mean Lp(a) concentration of 60.1 nmol/L. In subjects without diabetes or obesity, the inverse association of Lp(a)-TG was especially important (p < 0.001). The median Lp(a) was 58.3 nmol/L in those with TG < 300 mg/dL and 22.0 nmol/L if TG > 1000 mg/dL. No association was found between TG and Lp(a) in subjects with diabetes and obesity, nor in subjects with familial hypercholesterolemia. In subjects with multifactorial combined hyperlipemia with TG < 300 mg/dL, Lp(a) was 64.6 nmol/L; in the range of 300-399 mg/dL of TG, Lp(a) decreased to 38. 8 nmol/L, and up to 22.3 nmol/L when TG > 1000 mg/dL. CONCLUSIONS Our results show an inverse Lp(a)-TG relationship in TG concentrations > 300 mg/dL in subjects without diabetes, obesity and without familial hypercholesterolemia. Our results suggest that, in those hypertriglyceridemias due to hepatic overproduction of VLDL, the formation of Lp(a) is reduced, unlike those in which the peripheral catabolism of TG-rich lipoproteins is reduced.
Collapse
Affiliation(s)
- Victoria Marco-Benedí
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España; Universidad de Zaragoza, Zaragoza, España.
| | - Ana Cenarro
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España; Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, España
| | - Martín Laclaustra
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España; Universidad de Zaragoza, Zaragoza, España
| | - Pilar Calmarza
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España
| | - Ana M Bea
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España
| | - Àlex Vila
- Unidad de Lípidos, Servicio de Medicina Interna, Hospital de Figueres, Figueres, España
| | - Carlos Morillas-Ariño
- Sección de Endocrinología y Nutrición, Hospital Universitario Dr. Peset, Valencia, España; Departamento de Medicina, Universidad de Valencia, Valencia, España
| | - José Puzo
- Unidad de Lípidos, Servicio de Análisis y Bioquímica Clínica, Hospital San Jorge, Huesca, España
| | | | | | - Manuel Suárez Tembra
- Unidad de Lípidos y Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital San Rafael, A Coruña, España
| | - Fernando Civeira
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España; Universidad de Zaragoza, Zaragoza, España
| |
Collapse
|
72
|
Yuen T, Mancini GJ, Hegele RA, Pearson GJ. Consideration and Application of Lipoprotein(a) in the Risk Assessment of Atherosclerotic Cardiovascular Disease Risk in Adults. CJC Open 2024; 6:597-606. [PMID: 38559332 PMCID: PMC10980900 DOI: 10.1016/j.cjco.2023.11.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 04/04/2024] Open
Abstract
Lipoprotein(a) (Lp[a]) is an low-density lipoprotein (LDL)-like particle in which apolipoprotein (apo) B is covalently bound to a plasminogen-like molecule called apo(a). A High level of Lp(a) has been demonstrated to be an independent, causal, and prevalent risk factor for atherosclerotic cardiovascular disease (ASCVD), as well as aortic valve disease, through mechanisms that promote atherogenesis, inflammation, and thrombosis. With reliable and accessible assays, Lp(a) level has been established to be associated linearly with the risk for ASCVD. The 2021 Canadian Cardiovascular Society Dyslipidemia Guidelines recommend measuring an Lp(a) level once in a person's lifetime as part of the initial lipid screening. The aim of this review is to provide an update and overview of the utility and application of Lp(a) level in the assessment and treatment of adults at risk for ASCVD, consistent with this guideline recommendation.
Collapse
Affiliation(s)
- Tiffany Yuen
- Faculty of Medicine & Dentistry, University of Alberta, Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - G.B. John Mancini
- University of British Columbia, Department of Medicine, Division of Cardiology, Vancouver, British Columbia, Canada
| | - Robert A. Hegele
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Glen J. Pearson
- Faculty of Medicine & Dentistry, University of Alberta, Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| |
Collapse
|
73
|
Thau H, Neuber S, Emmert MY, Nazari-Shafti TZ. Targeting Lipoprotein(a): Can RNA Therapeutics Provide the Next Step in the Prevention of Cardiovascular Disease? Cardiol Ther 2024; 13:39-67. [PMID: 38381282 PMCID: PMC10899152 DOI: 10.1007/s40119-024-00353-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/12/2024] [Indexed: 02/22/2024] Open
Abstract
Numerous genetic and epidemiologic studies have demonstrated an association between elevated levels of lipoprotein(a) (Lp[a]) and cardiovascular disease. As a result, lowering Lp(a) levels is widely recognized as a promising strategy for reducing the risk of new-onset coronary heart disease, stroke, and heart failure. Lp(a) consists of a low-density lipoprotein-like particle with covalently linked apolipoprotein A (apo[a]) and apolipoprotein B-100, which explains its pro-thrombotic, pro-inflammatory, and pro-atherogenic properties. Lp(a) serum concentrations are genetically determined by the apo(a) isoform, with shorter isoforms having a higher rate of particle synthesis. To date, there are no approved pharmacological therapies that effectively reduce Lp(a) levels. Promising treatment approaches targeting apo(a) expression include RNA-based drugs such as pelacarsen, olpasiran, SLN360, and lepodisiran, which are currently in clinical trials. In this comprehensive review, we provide a detailed overview of RNA-based therapeutic approaches and discuss the recent advances and challenges of RNA therapeutics specifically designed to reduce Lp(a) levels and thus the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Henriette Thau
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Sebastian Neuber
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Maximilian Y Emmert
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353, Berlin, Germany.
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353, Berlin, Germany.
- Institute for Regenerative Medicine, University of Zurich, 8044, Zurich, Switzerland.
| | - Timo Z Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353, Berlin, Germany
- BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353, Berlin, Germany
| |
Collapse
|
74
|
Reijnders E, van der Laarse A, Ruhaak LR, Cobbaert CM. Closing the gaps in patient management of dyslipidemia: stepping into cardiovascular precision diagnostics with apolipoprotein profiling. Clin Proteomics 2024; 21:19. [PMID: 38429638 PMCID: PMC10908091 DOI: 10.1186/s12014-024-09465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/14/2024] [Indexed: 03/03/2024] Open
Abstract
In persons with dyslipidemia, a high residual risk of cardiovascular disease remains despite lipid lowering therapy. Current cardiovascular risk prediction mainly focuses on low-density lipoprotein cholesterol (LDL-c) levels, neglecting other contributing risk factors. Moreover, the efficacy of LDL-c lowering by statins resulting in reduced cardiovascular risk is only partially effective. Secondly, from a metrological viewpoint LDL-c falls short as a reliable measurand. Both direct and calculated LDL-c tests produce inaccurate test results at the low end under aggressive lipid lowering therapy. As LDL-c tests underperform both clinically and metrologically, there is an urging need for molecularly defined biomarkers. Over the years, apolipoproteins have emerged as promising biomarkers in the context of cardiovascular disease as they are the functional workhorses in lipid metabolism. Among these, apolipoprotein B (ApoB), present on all atherogenic lipoprotein particles, has demonstrated to clinically outperform LDL-c. Other apolipoproteins, such as Apo(a) - the characteristic apolipoprotein of the emerging risk factor lipoprotein(a) -, and ApoC-III - an inhibitor of triglyceride-rich lipoprotein clearance -, have attracted attention as well. To support personalized medicine, we need to move to molecularly defined risk markers, like the apolipoproteins. Molecularly defined diagnosis and molecularly targeted therapy require molecularly measured biomarkers. This review provides a summary of the scientific validity and (patho)physiological role of nine serum apolipoproteins, Apo(a), ApoB, ApoC-I, ApoC-II, ApoC-III, ApoE and its phenotypes, ApoA-I, ApoA-II, and ApoA-IV, in lipid metabolism, their association with cardiovascular disease, and their potential as cardiovascular risk markers when measured in a multiplex apolipoprotein panel.
Collapse
Affiliation(s)
- Esther Reijnders
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - Arnoud van der Laarse
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
75
|
Zhang Z, Rodriguez M, Zheng Z. Clot or Not? Reviewing the Reciprocal Regulation Between Lipids and Blood Clotting. Arterioscler Thromb Vasc Biol 2024; 44:533-544. [PMID: 38235555 PMCID: PMC10922732 DOI: 10.1161/atvbaha.123.318286] [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] [Indexed: 01/19/2024]
Abstract
Both hyperlipidemia and thrombosis contribute to the risks of atherosclerotic cardiovascular diseases, which are the leading cause of death and reduced quality of life in survivors worldwide. The accumulation of lipid-rich plaques on arterial walls eventually leads to the rupture or erosion of vulnerable lesions, triggering excessive blood clotting and leading to adverse thrombotic events. Lipoproteins are highly dynamic particles that circulate in blood, carry insoluble lipids, and are associated with proteins, many of which are involved in blood clotting. A growing body of evidence suggests a reciprocal regulatory relationship between blood clotting and lipid metabolism. In this review article, we summarize the observations that lipoproteins and lipids impact the hemostatic system, and the clotting-related proteins influence lipid metabolism. We also highlight the gaps that need to be filled in this area of research.
Collapse
Affiliation(s)
- Ziyu Zhang
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Maya Rodriguez
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, Wisconsin 53226, USA
- College of Arts and Sciences, Marquette University, Milwaukee, Wisconsin 53233, USA
| | - Ze Zheng
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| |
Collapse
|
76
|
Karp A, Jacobs M, Barris B, Labkowsky A, Frishman WH. Lipoprotein(a): A Review of Risk Factors, Measurements, and Novel Treatment Modalities. Cardiol Rev 2024:00045415-990000000-00218. [PMID: 38415744 DOI: 10.1097/crd.0000000000000667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
The study of lipoprotein(a) [Lp(a)] has long been a source of interest as a possible independent risk factor for atherosclerotic cardiovascular disease (ASCVD). The results of large sample observational studies, genome-wide association studies, and Mendelian randomization studies have been strong indicators supporting the link between ASCVD and Lp(a) despite early studies, with less sensitive assays, failing to show a connection. The recommendations for the indications and frequency of testing Lp(a) levels vary between US, Canadian, and European organizations due to the uncertain role of Lp(a) in ASCVD. The innovation of recent therapies, such as antisense oligonucleotides and small interfering RNA, designed to specifically target and reduce Lp(a) levels by targeting mRNA translation have once more thrust LP(a) into the spotlight of inquiry. These emerging modalities serve the dual purpose of definitively elucidating the connection between elevated Lp(a) levels and atherosclerotic cardiovascular risk, as well as the possibility of providing clinicians with the tools necessary to manage elevated Lp(a) levels in vulnerable populations. This review seeks to examine the mechanisms of atherogenicity of Lp(a) and explore the most current pharmacologic therapies currently in development.
Collapse
Affiliation(s)
- Avrohom Karp
- From the Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| | - Menachem Jacobs
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Ben Barris
- From the Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| | - Alexander Labkowsky
- From the Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| | - William H Frishman
- From the Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| |
Collapse
|
77
|
Yang R, Wu S, Zhao Z, Deng X, Deng Q, Wang D, Liu Q. Causal association between lipoproteins and risk of coronary artery disease-a systematic review and meta-analysis of Mendelian randomization studies. Clin Res Cardiol 2024:10.1007/s00392-024-02420-7. [PMID: 38407584 DOI: 10.1007/s00392-024-02420-7] [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: 12/10/2023] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
OBJECTIVE To systematically evaluate the causal effect of lipoproteins to the risk of coronary artery disease (CAD) by systematic review and meta-analysis of the associated Mendelian randomization (MR) studies. METHODS This systematic review was registered in PROSPERO (ID CRD42023465430). Searches from the databases (e.g., PubMed, Embase, Cochrane, Web of Science) and non-database sources to collect MR studies. The search time frame was from the database inception to August 2023. After data extraction, quality evaluation was performed, and the meta-analysis with bias evaluation was carried out with RevMan software. RESULTS A total of 5,828,409 participants from 21 records were included. Quality and bias assessment was performed by evaluating the internal three assumptions of MR studies. Meta-analysis for the causal association between non-HDL lipoproteins and CAD showed a significantly positive association between LDL and CAD (OR 1.37, 95% CI 1.26-1.49; P < 0.001, I2 = 95%), apoB and CAD (OR 1.38, 95% CI 1.11-1.71; P = 0.003, I2 = 98%), and Lp(a) and CAD (OR 1.21, 95% CI 1.12-1.31; P < 0.001, I2 = 99%). Interestingly, although there was no statistical significance in the association between VLDL/apoA1 and CAD (both P > 0.05), the pooled non-HDL lipoproteins showed a significantly positive association with CAD (OR 1.28, 95% CI 1.22-1.34; P < 0.001, I2 = 99%). For the HDL lipoproteins, the pooled OR showed a significantly negative association with CAD (OR 0.84, 95% CI 0.72-0.98; P = 0.002, I2 = 72%). However, the protective effect of HDL on CAD diminished when analyzed together with apoA1 and/or apoB (both P > 0.05). The funnel plot did not show serious publication bias, and sensitivity analysis performed relatively well robustness of the causal association of LDL, apoB, Lp(a), and total cholesterol with CAD. CONCLUSION The present meta-analysis suggests an overall effect of causal association between lipoproteins and CAD. Most of the non-HDL lipoproteins (LDL, apoB, Lp(a)) promote CAD, while the protective effect of HDL in CAD still needs to be verified in the future.
Collapse
Affiliation(s)
- Rongyuan Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Shirong Wu
- The Second Clinical School of Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Zhen Zhao
- The Second Clinical School of Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Xuanxuan Deng
- The Second Clinical School of Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Qiuying Deng
- The Second Clinical School of Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Dawei Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, 510120, People's Republic of China
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Qing Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, 510120, People's Republic of China.
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, #111 Dade Road, Yuexiu District, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
78
|
Agouridis AP, Filippatos TD, Kostapanos M, Kostara C, Tsimihodimos V. The effect of rosuvastatin alone or in combination with fenofibrate or omega-3 fatty acids on lipoprotein(a) levels in patients with mixed hyperlipidemia. Arch Med Sci Atheroscler Dis 2024; 9:e26-e32. [PMID: 38434941 PMCID: PMC10905261 DOI: 10.5114/amsad/178441] [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: 10/17/2023] [Accepted: 01/08/2024] [Indexed: 03/05/2024] Open
Abstract
Introduction Lipoprotein(a) [Lp(a)] is a strong, genetically determined, pathogenetic factor of atherosclerotic cardiovascular disease (ASCVD). The aim of this post-hoc analysis was to compare the effect of hypolipidemic treatment on Lp(a) levels of patients with mixed hyperlipidemia. Material and methods We previously randomized patients with mixed hyperlipidemia (low-density lipoprotein [LDL-C] > 160 mg/dl and triglycerides > 200 mg/dl) to rosuvastatin monotherapy 40 mg/day (R group, n = 30) or rosuvastatin 10 mg/day combined with fenofibrate 200 mg/day (RF group, n = 30) or omega-3 fatty acids 2 g/day (RΩ group, n = 30). In the present post-hoc analysis, we included only the patients whose Lp(a) levels were assessed (16, 16 and 15 in the R, RF and RΩ groups, respectively). Lipid profile and Lp(a) were measured at baseline and after 3 months of treatment. Results Significant reductions in total cholesterol, LDL-C, non-high-density lipoprotein-cholesterol (non-HDL-C) and triglyceride levels were observed in all groups. A significant increase in Lp(a) levels was noted in the R (p = 0.017) and RF (p = 0.029) groups, while no significant difference was seen in the RΩ group (p = NS). Regarding Lp(a) elevations, no differences were found between groups. In the R group, a strong negative correlation between the changes in Lp(a) and LDL-C (r = -0.500, p = 0.049) was observed, while a significant negative correlation between the changes in Lp(a) and triglycerides (r = -0.531, p = 0.034) was noted in the RF group. Conclusions Rosuvastatin and/or fenofibrate treatment increases Lp(a) levels in patients with mixed hyperlipidemia. Novel therapies should target Lp(a) level reduction to decrease the residual ASCVD risk in patients with mixed hyperlipidemia.
Collapse
Affiliation(s)
- Aris P Agouridis
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Department of Internal Medicine, German Oncology Center, Limassol, Cyprus
- Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
| | - Theodosios D Filippatos
- Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
- Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Michael Kostapanos
- Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
- Lipid Clinic, Department of General Medicine, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Christina Kostara
- Laboratory of Clinical Chemistry, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Vasilis Tsimihodimos
- Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
| |
Collapse
|
79
|
Mormone A, Tortorella G, Esposito F, Caturano A, Marrone A, Cozzolino D, Galiero R, Marfella R, Sasso FC, Rinaldi L. Advances in Pharmacological Approaches for Managing Hypercholesterolemia: A Comprehensive Overview of Novel Treatments. Biomedicines 2024; 12:432. [PMID: 38398034 PMCID: PMC10887105 DOI: 10.3390/biomedicines12020432] [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: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Hypercholesterolemia plays a crucial role in the formation of lipid plaques, particularly with elevated low-density lipoprotein (LDL-C) levels, which are linked to increased risks of cardiovascular disease, cerebrovascular disease, and peripheral arterial disease. Controlling blood cholesterol values, specifically reducing LDL-C, is widely recognized as a key modifiable risk factor for decreasing the morbidity and mortality associated with cardiovascular diseases. Historically, statins, by inhibiting the enzyme β-hydroxy β-methylglutaryl-coenzyme A (HMG)-CoA reductase, have been among the most effective drugs. However, newer non-statin agents have since been introduced into hypercholesterolemia therapy, providing a viable alternative with a favorable cost-benefit ratio. This paper aims to delve into the latest therapies, shedding light on their mechanisms of action and therapeutic benefits.
Collapse
Affiliation(s)
- Andrea Mormone
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Giovanni Tortorella
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Francesca Esposito
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
- Department of Experimental Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy
| | - Aldo Marrone
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Domenico Cozzolino
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy
| |
Collapse
|
80
|
Bhatia HS, Trainor P, Carlisle S, Tsai MY, Criqui MH, DeFilippis A, Tsimikas S. Aspirin and Cardiovascular Risk in Individuals With Elevated Lipoprotein(a): The Multi-Ethnic Study of Atherosclerosis. J Am Heart Assoc 2024; 13:e033562. [PMID: 38293935 PMCID: PMC11056170 DOI: 10.1161/jaha.123.033562] [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: 11/15/2023] [Accepted: 12/15/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND Effective therapies for reducing cardiovascular disease (CVD) risk in people with elevated lipoprotein(a) are lacking, especially for primary prevention. Because of the potential association of lipoprotein(a) with thrombosis, we evaluated the relationship between aspirin use and CVD events in people with elevated lipoprotein(a). METHODS AND RESULTS We used data from the MESA (Multi-Ethnic Study of Atherosclerosis), a prospective cohort study of individuals free of baseline cardiovascular disease. Due to potential confounding by indication, we matched aspirin users to nonusers using a propensity score based on CVD risk factors. We then evaluated the association between aspirin use and coronary heart disease (CHD) events (CHD death, nonfatal myocardial infarction) stratified by baseline lipoprotein(a) level (threshold of 50 mg/dL) using Cox proportional hazards models with adjustment for CVD risk factors. After propensity matching, the study cohort included 2183 participants, including 1234 (57%) with baseline aspirin use and 423 (19%) with lipoprotein(a) >50 mg/dL. Participants with lipoprotein(a) >50 mg/dL had a higher burden of CVD risk factors, more frequent aspirin use (61.7% versus 55.3%, P=0.02), and higher rate of incident CHD events (13.7% versus 8.9%, P<0.01). Aspirin was associated with a significant reduction in CHD events among those with elevated lipoprotein(a) (hazard ratio, 0.54 [95% CI, 0.32-0.94]; P=0.03). Those with lipoprotein(a) >50 mg/dL and aspirin use had similar CHD risk as those with lipoprotein(a) ≤50 mg/dL regardless of aspirin use. CONCLUSIONS Aspirin use was associated with a significantly lower risk for CHD events in participants with lipoprotein(a) >50 mg/dL without baseline CVD. The results of this observational propensity-matched study require confirmation in studies with randomization of aspirin use.
Collapse
Affiliation(s)
- Harpreet S. Bhatia
- Division of Cardiovascular Medicine, Department of MedicineUniversity of California, San DiegoLa JollaCA
| | - Patrick Trainor
- Department of Chemistry and BiochemistryNew Mexico State UniversityLas CrucesNM
| | - Samantha Carlisle
- Department of Chemistry and BiochemistryNew Mexico State UniversityLas CrucesNM
| | - Michael Y. Tsai
- Department of Laboratory Medicine and PathologyUniversity of MinnesotaMinneapolisMN
| | - Michael H. Criqui
- Division of Cardiovascular Medicine, Department of MedicineUniversity of California, San DiegoLa JollaCA
- Division of Preventive Medicine, Department of Family MedicineUniversity of California, San DiegoLa JollaCA
| | - Andrew DeFilippis
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTN
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Department of MedicineUniversity of California, San DiegoLa JollaCA
| |
Collapse
|
81
|
Welsh P, Al Zabiby A, Byrne H, Benbow HR, Itani T, Farries G, Costa-Scharplatz M, Ferber P, Martin L, Brown R, Fonseca AF, Sattar N. Elevated lipoprotein(a) increases risk of subsequent major adverse cardiovascular events (MACE) and coronary revascularisation in incident ASCVD patients: A cohort study from the UK Biobank. Atherosclerosis 2024; 389:117437. [PMID: 38219651 DOI: 10.1016/j.atherosclerosis.2023.117437] [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: 07/06/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND AND AIMS Elevated lipoprotein(a) [Lp(a)] is a genetic driver for atherosclerotic cardiovascular disease (ASCVD). We aimed to provide novel insights into the associated risk of elevated versus normal Lp(a) levels on major adverse cardiovascular events (MACE) in an incident ASCVD cohort. METHODS This was an observational cohort study of incident ASCVD patients. MACE counts and incidence rates (IRs) per 100-person-years were reported for patients with normal (<65 nmol/L) and elevated (>150 nmol/L) Lp(a) within the first year after incident ASCVD diagnosis and overall follow-up. Cox proportional hazard models quantified the risk of MACE associated with a 100 nmol/L increase in Lp(a). RESULTS The study cohort included 32,537 incident ASCVD patients; 5204 with elevated and 22,257 with normal Lp(a). Of those with elevated Lp(a), 41.2% had a subsequent MACE, versus 35.61% with normal Lp(a). Within the first year of follow-up, the IRs of composite MACE and coronary revascularisation were significantly higher (p < 0.001) in patients with elevated versus normal Lp(a) (IR difference 6.79 and 4.66). This trend was also observed in the overall follow-up (median 4.7 years). Using time to first subsequent MACE, a 100 nmol/L increase in Lp(a) was associated with an 8.0% increased risk of composite MACE, and 18.6% increased risk of coronary revascularisation during the overall follow-up period. CONCLUSIONS The association of elevated Lp(a) with increased risk of subsequent MACE and coronary revascularisation highlights a population who may benefit from earlier and more targeted intervention for cardiovascular risk including Lp(a), particularly within the first year after ASCVD diagnosis. Proactive Lp(a) testing as part of routine clinical practice can help identify and better manage these higher-risk individuals.
Collapse
Affiliation(s)
- Paul Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom.
| | | | | | | | | | | | | | | | | | - Rosemary Brown
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | | | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
82
|
Ciffone N, McNeal CJ, McGowan MP, Ferdinand KC. Lipoprotein(a): An important piece of the ASCVD risk factor puzzle across diverse populations. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 38:100350. [PMID: 38510747 PMCID: PMC10945898 DOI: 10.1016/j.ahjo.2023.100350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/21/2023] [Indexed: 03/22/2024]
Abstract
Elevated lipoprotein(a) (Lp[a]) is an independent, genetic risk factor for atherosclerotic cardiovascular disease (ASCVD) that impacts ~1.4 billion people globally. Generally, Lp(a) levels remain stable over time; thus, most individuals need only undergo Lp(a) testing through a non-fasting blood draw once in their lifetime, unless elevated Lp(a) is identified. Despite the convenience of the test for clinicians and patients, routine Lp(a) testing has not been widely adopted. This review provides a guide to the benefits of Lp(a) testing and solutions for overcoming common barriers in practice, including access to testing and lack of awareness. Lp(a) testing provides the opportunity to reclassify ASCVD risk and drive intensive cardiovascular risk factor management in individuals with elevated Lp(a), and to identify patients potentially less likely to respond to statins. Moreover, cascade screening can help to identify elevated Lp(a) in relatives of individuals with a personal or family history of premature ASCVD. Overall, given the profound impact of elevated Lp(a) on cardiovascular risk, Lp(a) testing should be an essential component of risk assessment by primary and specialty care providers.
Collapse
Affiliation(s)
- Nicole Ciffone
- Arizona Center for Advanced Lipidology, 3925 E Fort Lowell Rd, Tucson, AZ 85712, USA
| | | | - Mary P. McGowan
- The Family Heart Foundation, 680 E. Colorado Blvd, Suite 180, Pasadena, CA 91101, USA
- Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, 1 Rope Ferry Rd, Hanover, NH 03755, USA
| | - Keith C. Ferdinand
- John W. Deming Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| |
Collapse
|
83
|
Mulvagh SL, Colella TJ, Gulati M, Crosier R, Allana S, Randhawa VK, Bruneau J, Pacheco C, Jaffer S, Cotie L, Mensour E, Clavel MA, Hill B, Kirkham AA, Foulds H, Liblik K, Van Damme A, Grace SL, Bouchard K, Tulloch H, Robert H, Pike A, Benham JL, Tegg N, Parast N, Adreak N, Boivin-Proulx LA, Parry M, Gomes Z, Sarfi H, Iwegim C, Van Spall HG, Nerenberg KA, Wright SP, Limbachia JA, Mullen KA, Norris CM. The Canadian Women's Heart Health Alliance ATLAS on the Epidemiology, Diagnosis, and Management of Cardiovascular Disease in Women - Chapter 9: Summary of Current Status, Challenges, Opportunities, and Recommendations. CJC Open 2024; 6:258-278. [PMID: 38487064 PMCID: PMC10935707 DOI: 10.1016/j.cjco.2023.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/03/2023] [Indexed: 03/17/2024] Open
Abstract
This final chapter of the Canadian Women's Heart Health Alliance "ATLAS on the Epidemiology, Diagnosis, and Management of Cardiovascular Disease in Women" presents ATLAS highlights from the perspective of current status, challenges, and opportunities in cardiovascular care for women. We conclude with 12 specific recommendations for actionable next steps to further the existing progress that has been made in addressing these knowledge gaps by tackling the remaining outstanding disparities in women's cardiovascular care, with the goal to improve outcomes for women in Canada.
Collapse
Affiliation(s)
- Sharon L. Mulvagh
- Division of Cardiology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Tracey J.F. Colella
- KITE-UHN-Toronto Rehabilitation, Toronto, Ontario, Canada
- Lawrence S. Bloomberg Faculty of Nursing, Rehabilitation Sciences Institute, University of Toronto, Toronto, Ontario, Canada
| | - Martha Gulati
- Barbra Streisand Women’s Heart Center, Cedars Sinai Heart Institute, Los Angeles, California, USA
| | - Rebecca Crosier
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | | | | | - Jill Bruneau
- Memorial University, St John's, Newfoundland and Labrador, Canada
| | - Christine Pacheco
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Shahin Jaffer
- Department of Medicine, Division of Community Internal Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lisa Cotie
- KITE-UHN-Toronto Rehabilitation, Toronto, Ontario, Canada
| | - Emma Mensour
- University of Western Ontario, London, Ontario, Canada
| | | | - Braeden Hill
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Amy A. Kirkham
- KITE-UHN-Toronto Rehabilitation, Toronto, Ontario, Canada
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Heather Foulds
- College of Kinesiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Kiera Liblik
- Department of Medicine, Queen’s University, Kingston, Ontario, Canada
| | - Andrea Van Damme
- University of Alberta Faculty of Graduate & Postdoctoral Studies, Edmonton, Alberta, Canada
| | - Sherry L. Grace
- York University and University Health Network, Toronto, Ontario, Canada
| | - Karen Bouchard
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Heather Tulloch
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Helen Robert
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - April Pike
- Memorial University, St John's, Newfoundland and Labrador, Canada
| | - Jamie L. Benham
- Departments of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicole Tegg
- Faculties of Nursing, Medicine, and School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| | - Nazli Parast
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Najah Adreak
- University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Monica Parry
- Lawrence S. Bloomberg Faculty of Nursing, Rehabilitation Sciences Institute, University of Toronto, Toronto, Ontario, Canada
| | - Zoya Gomes
- Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hope Sarfi
- Canadian Women’s Heart Health Alliance, Ottawa, Ontario, Canada
| | - Chinelo Iwegim
- Fraser Health Authority, Surrey, British Columbia, Canada
| | - Harriette G.C. Van Spall
- Departments of Medicine and Health Research Methods, Evidence, and Impact, Research Institute of St Joe’s, Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Kara A. Nerenberg
- Departments of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | - Colleen M. Norris
- Faculties of Nursing, Medicine, and School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
84
|
Tasdighi E, Adhikari R, Almaadawy O, Leucker TM, Blaha MJ. LP(a): Structure, Genetics, Associated Cardiovascular Risk, and Emerging Therapeutics. Annu Rev Pharmacol Toxicol 2024; 64:135-157. [PMID: 37506332 DOI: 10.1146/annurev-pharmtox-031023-100609] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Lipoprotein(a) [Lp(a)] is a molecule bound to apolipoprotein(a) with some similarity to low-density lipoprotein cholesterol (LDL-C), which has been found to be a risk factor for cardiovascular disease (CVD). Lp(a) appears to induce inflammation, atherogenesis, and thrombosis. Approximately 20% of the world's population has increased Lp(a) levels, determined predominantly by genetics. Current clinical practices for the management of dyslipidemia are ineffective in lowering Lp(a) levels. Evolving RNA-based therapeutics, such as the antisense oligonucleotide pelacarsen and small interfering RNA olpasiran, have shown promising results in reducing Lp(a) levels. Phase III pivotal cardiovascular outcome trials [Lp(a)HORIZON and OCEAN(a)] are ongoing to evaluate their efficacy in secondary prevention of major cardiovascular events in patients with elevated Lp(a). The future of cardiovascular residual risk reduction may transition to a personalized approach where further lowering of either LDL-C, triglycerides, or Lp(a) is selected after high-intensity statin therapy based on the individual risk profile and preferences of each patient.
Collapse
Affiliation(s)
- Erfan Tasdighi
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rishav Adhikari
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Omar Almaadawy
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore, Maryland, USA
| | - Thorsten M Leucker
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael J Blaha
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
85
|
Chen X, Song C, Ma X, Tao J, Hu L, Xu Y, Yi Y, Yang X, Jiang L. High lipoprotein(a) concentration is associated with moyamoya disease. Lipids Health Dis 2024; 23:21. [PMID: 38254149 PMCID: PMC10802057 DOI: 10.1186/s12944-024-02015-1] [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: 10/05/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Moyamoya disease (MMD) has attracted the attention of scholars because of its rarity and unknown etiology. METHODS Data for this study were sourced from the Second Affiliated Hospital of Nanchang University. Regression analyses were conducted to examine the association in Lipoprotein [Lp(a)] and MMD. R and IBM SPSS were conducted. RESULTS A cohort comprising 1012 MMD patients and 2024 controls was established through the propensity score matching method. Compared with controls, MMD patients showed higher median Lp(a) concentrations [18.5 (9.6-37.8) mg/dL vs. 14.9 (7.8-30.5) mg/dL, P < 0.001]. The odds ratios and 95% confidence intervals for Lp(a) were calculated in three models: unadjusted model, model 1 (adjusted for body mass index and systolic blood pressure), and model 2 (adjusted for model 1 plus triglyceride, C-reactive protein, homocysteine, and low-density lipoprotein cholesterol). Results were [1.613 (1.299-2.002), P < 0.001], [1.598 (1.286-1.986), P < 0.001], and [1.661 (1.330-2.074), P < 0.001], respectively. Furthermore, age, sex, or hypertension status had nothing to do with this relationship. CONCLUSIONS Positive relationship exists between Lp(a) and MMD.
Collapse
Affiliation(s)
- Xinyue Chen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, 330006, China
| | - Chenxin Song
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, 330006, China
| | - Xianrun Ma
- School of Basic Medical Sciences, Nanchang University, Jiangxi, Nanchang, 330006, China
| | - Junjie Tao
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, 330006, China
| | - Lijuan Hu
- Department of Nursing, Nanchang Medical College, Nanchang, Jiangxi, China
| | - Yuan Xu
- Department of Medical Big Data Center, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yingping Yi
- Department of Medical Big Data Center, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xinlei Yang
- Biobank center, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Long Jiang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
86
|
Arnold N, Blaum C, Goßling A, Brunner FJ, Bay B, Ferrario MM, Brambilla P, Cesana G, Leoni V, Palmieri L, Donfrancesco C, Padró T, Andersson J, Jousilahti P, Ojeda F, Zeller T, Linneberg A, Söderberg S, Iacoviello L, Gianfagna F, Sans S, Veronesi G, Thorand B, Peters A, Tunstall-Pedoe H, Kee F, Salomaa V, Schnabel RB, Kuulasmaa K, Blankenberg S, Koenig W, Waldeyer C. C-reactive protein modifies lipoprotein(a)-related risk for coronary heart disease: the BiomarCaRE project. Eur Heart J 2024:ehad867. [PMID: 38240386 DOI: 10.1093/eurheartj/ehad867] [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: 05/31/2023] [Revised: 11/20/2023] [Accepted: 12/06/2023] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND AND AIMS Recent investigations have suggested an interdependence of lipoprotein(a) [Lp(a)]-related risk for cardiovascular disease with background inflammatory burden. The aim the present analysis was to investigate whether high-sensitive C-reactive protein (hsCRP) modulates the association between Lp(a) and coronary heart disease (CHD) in the general population. METHODS Data from 71 678 participants from 8 European prospective population-based cohort studies were used (65 661 without/6017 with established CHD at baseline; median follow-up 9.8/13.8 years, respectively). Fine and Gray competing risk-adjusted models were calculated according to accompanying hsCRP concentration (<2 and ≥2 mg/L). RESULTS Among CHD-free individuals, increased Lp(a) levels were associated with incident CHD irrespective of hsCRP concentration: fully adjusted sub-distribution hazard ratios [sHRs (95% confidence interval)] for the highest vs. lowest fifth of Lp(a) distribution were 1.45 (1.23-1.72) and 1.48 (1.23-1.78) for a hsCRP group of <2 and ≥2 mg/L, respectively, with no interaction found between these two biomarkers on CHD risk (Pinteraction = 0.82). In those with established CHD, similar associations were seen only among individuals with hsCRP ≥ 2 mg/L [1.34 (1.03-1.76)], whereas among participants with a hsCRP concentration <2 mg/L, there was no clear association between Lp(a) and future CHD events [1.29 (0.98-1.71)] (highest vs. lowest fifth, fully adjusted models; Pinteraction = 0.024). CONCLUSIONS While among CHD-free individuals Lp(a) was significantly associated with incident CHD regardless of hsCRP, in participants with CHD at baseline, Lp(a) was related to recurrent CHD events only in those with residual inflammatory risk. These findings might guide adequate selection of high-risk patients for forthcoming Lp(a)-targeting compounds.
Collapse
Affiliation(s)
- Natalie Arnold
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christopher Blaum
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alina Goßling
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian J Brunner
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Bay
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marco M Ferrario
- Research Center in Epidemiology and Preventive Medicine-EPIMED, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Paolo Brambilla
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Giancarlo Cesana
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Valerio Leoni
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST Brianza, School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Luigi Palmieri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità-ISS, Rome, Italy
| | - Chiara Donfrancesco
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità-ISS, Rome, Italy
| | - Teresa Padró
- Cardiovascular-Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Jonas Andersson
- Department of Public Health and Clinical Medicine, Skellefteå Research Unit, Umeå University, Skellefteå, Sweden
| | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Francisco Ojeda
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Zeller
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- University Center of Cardiovascular Science, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stefan Söderberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Licia Iacoviello
- Research Center in Epidemiology and Preventive Medicine-EPIMED, Department of Medicine and Surgery, University of Insubria, Varese, Italy
- Department of Epidemiology and Prevention, IRCCS Neuromed, Pozzilli, Italy
| | - Francesco Gianfagna
- Research Center in Epidemiology and Preventive Medicine-EPIMED, Department of Medicine and Surgery, University of Insubria, Varese, Italy
- Mediterranea Cardiocentro, Naples, Italy
| | - Susana Sans
- Catalan Department of Health, Barcelona, Spain
| | - Giovanni Veronesi
- Research Center in Epidemiology and Preventive Medicine-EPIMED, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology-IBE, Ludwig-Maximilians University of Munich, Munich, Germany
- German Center for Cardiovascular Disease Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Hugh Tunstall-Pedoe
- Cardiovascular Epidemiology Unit, Institute of Cardiovascular Research, University of Dundee, Dundee, Scotland
| | - Frank Kee
- Centre for Public Health, Queens University of Belfast, Belfast, Northern Ireland, UK
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Renate B Schnabel
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kari Kuulasmaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Stefan Blankenberg
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wolfgang Koenig
- German Center for Cardiovascular Disease Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- German Heart Center, Munich, Technical University of Munich, Lazarettstr. 36, Munich 80636, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Christoph Waldeyer
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
87
|
Wang X, Wang Z, He J. Similarities and Differences of Vascular Calcification in Diabetes and Chronic Kidney Disease. Diabetes Metab Syndr Obes 2024; 17:165-192. [PMID: 38222032 PMCID: PMC10788067 DOI: 10.2147/dmso.s438618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024] Open
Abstract
Presently, the mechanism of occurrence and development of vascular calcification (VC) is not fully understood; a range of evidence suggests a positive association between diabetes mellitus (DM) and VC. Furthermore, the increasing burden of central vascular disease in patients with chronic kidney disease (CKD) may be due, at least in part, to VC. In this review, we will review recent advances in the mechanisms of VC in the context of CKD and diabetes. The study further unveiled that VC is induced through the stimulation of pro-inflammatory factors, which in turn impairs endothelial function and triggers similar mechanisms in both disease contexts. Notably, hyperglycemia was identified as the distinctive mechanism driving calcification in DM. Conversely, in CKD, calcification is facilitated by mechanisms including mineral metabolism imbalance and the presence of uremic toxins. Additionally, we underscore the significance of investigating vascular alterations and newly identified molecular pathways as potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Xiabo Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Jianqiang He
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| |
Collapse
|
88
|
Chen J, Li W, Cao J, Lu Y, Wang C, Lu J. Risk factors for carotid plaque formation in type 2 diabetes mellitus. J Transl Med 2024; 22:18. [PMID: 38178198 PMCID: PMC10768372 DOI: 10.1186/s12967-023-04836-7] [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: 09/05/2023] [Accepted: 12/26/2023] [Indexed: 01/06/2024] Open
Abstract
OBJECT Patients with type 2 diabetes mellitus (T2DM) are at higher risk of developing atherosclerosis. Previous studies have analyzed the factors associated with diabetic macrovascular disease, although whether these factors are applicable to T2DM patients with carotid atherosclerosis remains unclear. Therefore, the aim of this study was to investigate the risk factors for the formation of carotid atherosclerotic plaque in hospitalized T2DM patients and to provide a theoretical basis for early prevention and treatment of carotid atherosclerosis in these patients. METHODS A total of 949 patients with T2DM were included in the study. Carotid ultrasound identified 531 patients with carotid atherosclerotic plaque. The waist-to-hip ratio (WHR), blood glucose, liver and kidney function, blood lipid profile, islet function, and other indicators were measured at the same time to identify the risk factors and predictive significance of T2DM carotid plaque. RESULTS The proportions of men, diabetes nephropathy (DN) and hypertension in T2DM patients with carotid plaque are higher than those without carotid plaque(P < 0.05). Age, duration of diabetes, WHR, Postprandial glucose (PPG), lipoprotein (a) [Lip (a)], carcinoembryonic antigen(CEA) and estimated glomerular filtration rate (eGFR) in T2DM patients with carotid plaque were higher than those without plaque (P < 0.05). Age, WHR, duration of diabetes, hypertension, males, and Lip (a) were independent risk factors for T2DM patients with carotid plaque. Age, WHR, duration of diabetes, and Lip (a) had a higher AUC to predict T2DM with carotid artery plaque (AUC: 0.750, 0.640, 0.678, 0.552 respectively; P all < 0.001). After constructing the logit (P) value of the above risk factors, the area under the ROC curve was 0.816 (0.789-0.842, P < 0.001). CONCLUSION Age, WHR, duration of diabetes, hypertension, males, and Lip (a) levels are the main risk factors for the formation of carotid plaque in T2DM patients. Combining the above risk factors provides a better prediction of carotid plaque formation in T2DM.
Collapse
Affiliation(s)
- Jin Chen
- Department of Endocrinology and Metabolism, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Wenwen Li
- Department of Endocrinology and Metabolism, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jingzhu Cao
- Department of Endocrinology and Metabolism, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yuhan Lu
- Department of health, The affiliated hospital of Qingdao University, Qingdao, China
| | - Chaoqun Wang
- Department of Endocrinology and Metabolism, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Jin Lu
- Department of Endocrinology and Metabolism, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
89
|
Liang D, Liang D, Liu J, Zheng Y, Huang D, Li Z, Huang X, Chen J. Impact of lipoprotein(a) and fibrinogen on prognosis in patients with coronary artery disease: A retrospective cohort study. Technol Health Care 2024; 32:3317-3328. [PMID: 38848204 PMCID: PMC11492067 DOI: 10.3233/thc-240005] [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: 01/02/2024] [Accepted: 04/16/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Despite the considerable progress made in preventative methods, medication, and interventional therapies, it remains evident that cardiovascular events (CVEs) continue to be the primary cause of both death and morbidity among individuals diagnosed with coronary artery disease (CAD). OBJECTIVE To compare the connection between lipoprotein a (Lp[a]), fibrinogen (Fib), and both parameters combined with all-cause mortality to detect their value as prognostic biomarkers. METHODS This is a retrospective study. Patients diagnosed with CAD between January 2007 and December 2020 at the Guangdong Provincial People's Hospital (China) were involved in the study. 43,367 patients met the eligibility criteria. The Lp(a) and Fib levels were distributed into three tertile groups (low, medium, and high). All of the patients included in the study were followed up for all-cause mortality. Kaplan-Meier and Cox regression were performed to determine the relationship between Lp(a), Fib, and all-cause mortality. A concordance statistics model was developed to detect the impact of Fib and Lp(a) in terms of anticipating poor outcomes in patients with CAD. RESULTS Throughout a median follow-up of 67.0 months, 6,883 (15.9%) patients died. Participants with high Lp(a) (above 27.60 mg/dL) levels had a significantly higher risk for all-cause mortality than individuals with low Lp(a) levels (below 11.13 mg/dL; adjusted hazard ratio [aHR] 1.219, 95% confidence interval [CI]: 1.141-1.304, p< 0.001). Similarly, patients with high Fib levels (above 4.32 g/L) had a significantly greater risk of developing all-cause mortality compared with those with reduced Fib levels (below 3.41 g/L; aHR 1.415, 95% CI: 1.323-1.514, p< 0.001). Patients with raised Lp(a) and Fib levels had the maximum risk for all-cause mortality (aHR 1.702; 95% CI: 1.558-1.859, p< 0.001). When considered together, Lp(a) and Fib caused a significant elevation of the concordance statistic by 0.009 (p< 0.05), suggesting a higher value for predicting mortality when combining the two indicators. CONCLUSION High Lp(a) and Fib levels could be used as predictive biomarkers for all-cause mortality in individuals with CAD. The prediction accuracy for all-cause mortality improved after combining the two parameters.
Collapse
Affiliation(s)
- Dakai Liang
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of Cardiology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Dandan Liang
- Department of Cardiology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Jin Liu
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yiying Zheng
- Department of Cardiology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Dehua Huang
- Department of Cardiology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Zeliang Li
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiaoyu Huang
- Department of Cardiology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Jiyan Chen
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
90
|
Mukherjee D, Nissen SE. Lipoprotein (a) as a Biomarker for Cardiovascular Diseases and Potential New Therapies to Mitigate Risk. Curr Vasc Pharmacol 2024; 22:171-179. [PMID: 38141196 DOI: 10.2174/0115701611267835231210054909] [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: 08/04/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is a molecule that induces inflammation of the blood vessels, atherogenesis, valvular calcification, and thrombosis. METHODS We review the available evidence that suggests that high Lp(a) levels are associated with a persisting risk for atherosclerotic cardiovascular diseases despite optimization of established risk factors, including low-density lipoprotein cholesterol (LDL-C) levels. OBSERVATIONS Approximately a quarter of the world population have Lp(a) levels of >50 mg/dL (125 nmol/L), a level associated with elevated cardiovascular risk. Lifestyle modification, statins, and ezetimibe do not effectively lower Lp(a) levels, while proprotein convertase subtilisin/kexin type 9 (PCSK-9) inhibitors and niacin only lower Lp(a) levels modestly. We describe clinical studies suggesting that gene silencing therapeutics, such as small interfering RNA (siRNA) and antisense oligonucleotide targeting Lp(a), offer a targeted approach with the potential for safe and robust Lp(a)- lowering with only a few doses (3-4) per year. Prospective randomized phase 3 studies are ongoing to validate safety, effectiveness in improving hard clinical outcomes, and tolerability to assess these therapies. CONCLUSION Several emerging treatments with robust Lp(a)-lowering effects may significantly lower atherosclerotic cardiovascular risk.
Collapse
Affiliation(s)
- Debabrata Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center at El Paso, Texas, USA
| | - Steven E Nissen
- Heart, Vascular, and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
91
|
Wang Z, Xu L, Sun L, Jiang X, Wang J. The role of computed tomography angiography in assessing the correlation between properties of coronary atherosclerotic plaque and blood lipids. Technol Health Care 2024; 32:2265-2275. [PMID: 38393936 DOI: 10.3233/thc-231036] [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: 02/25/2024]
Abstract
BACKGROUND Coronary atherosclerotic heart disease (CAHD) is the leading cause of death in developed countries. OBJECTIVE This study aimed to explore the correlation between the properties of coronary atherosclerotic plaque and blood lipids using computed tomography angiography (CTA). METHODS A total of 83 patients with coronary heart disease were included in this study (males: 50; females: 33; average age: [59 ± 8] years old). They were classified into the stable angina group and unstable angina group. Atherosclerotic plaques were classified as fatty plaques (soft plaques), fibrous plaques, and calcified plaques based on the computed tomography (CT) values. SPSS 17.0 statistical software was used to analyze the correlation between the properties of angina and the CT values of atherosclerotic plaques, blood lipids, and plaque properties, and then compared between the stable and unstable angina groups. RESULTS There were statistically significant differences in plaque properties between the stable and unstable angina groups (P< 0.001). During CTA examination, we found statistically significant differences in the CT density values of atherosclerotic plaques between the stable and unstable angina groups (P< 0.001). There were statistically significant differences between the properties of angina and the level of blood lipids (P< 0.05). CONCLUSION Anginal properties negatively correlated with calcified plaques and positively correlated with non-calcified plaques. Calcified plaques negatively correlated with total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and triglycerides (TG), and positively correlated with high-density lipoprotein cholesterol (HDL-C). Non-calcified plaques negatively correlated with HDL-C and positively correlated with TC, LDL-C, and TG.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Cardiovascular Medicine, Affiliated Hospital of Beihua University, Jilin, China
| | - Lei Xu
- Department of Cardiovascular Medicine, Affiliated Hospital of Beihua University, Jilin, China
| | - Lin Sun
- Department of Cardiovascular Medicine, Affiliated Hospital of Beihua University, Jilin, China
| | - Xin Jiang
- Department of Cardiovascular Medicine, Affiliated Hospital of Beihua University, Jilin, China
| | - Juan Wang
- Department of Gynecology, Jilin City Central Hospital, Jilin, China
| |
Collapse
|
92
|
Zhang K, Li J, Li Y, Yan K, Zhu P, Tang X, Yuan D, Yang Y, Gao R, Yuan J, Zhao X. Elevated High-Sensitivity C-Reactive Protein Level Enhances the Impact of Lipoprotein(a) on Platelet Reactivity in PCI Patients Treated with Clopidogrel. Clin Appl Thromb Hemost 2024; 30:10760296241280711. [PMID: 39246223 PMCID: PMC11388299 DOI: 10.1177/10760296241280711] [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: 09/10/2024] Open
Abstract
BACKGROUND Recently, the effect of Lipoprotein(a) [Lp(a)] on thrombogenesis has aroused great interest, while inflammation has been reported to modify the Lp(a)-associated risks through an unidentified mechanism. PURPOSE This study aimed to evaluate the association between platelet reactivity with Lp(a) and high-sensitivity C-reactive protein (hs-CRP) levels in percutaneous intervention (PCI) patients treated with clopidogrel. METHODS Data were collected from 10,724 consecutive PCI patients throughout the year 2013 in Fuwai Hospital. High on-treatment platelet reactivity (HTPR) and low on-treatment platelet reactivity (LTPR) were defined as thrombelastography (TEG) maximum amplitude of adenosine diphosphate-induced platelet (MAADP) > 47 mm and < 31 mm, respectively. RESULTS 6615 patients with TEG results were finally enrolled. The mean age was 58.24 ± 10.28 years and 5131 (77.6%) were male. Multivariable logistic regression showed that taking Lp(a) < 30 mg/dL and hs-CRP < 2 mg/L as the reference, isolated Lp(a) elevation [Lp(a) ≥ 30 mg/dL and hs-CRP < 2 mg/L] was not significantly associated with HTPR (P = 0.153) or LTPR (P = 0.312). However, the joint elevation of Lp(a) and hs-CRP [Lp(a) ≥ 30 mg/dL and hs-CRP ≥ 2 mg/L] exhibited enhanced association with both HTPR (OR:1.976, 95% CI 1.677-2.329) and LTPR (OR:0.533, 95% CI 0.454-0.627). CONCLUSIONS The isolated elevation of Lp(a) level was not an independent indicator for platelet reactivity, yet the concomitant elevation of Lp(a) and hs-CRP levels was significantly associated with increased platelet reactivity. Whether intensified antiplatelet therapy or anti-inflammatory strategies could mitigate the risks in patients presenting combined Lp(a) and hs-CRP elevation requires future investigation.
Collapse
Affiliation(s)
- Kexin Zhang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jiawen Li
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yulong Li
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Kailun Yan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Pei Zhu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xiaofang Tang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Deshan Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yuejin Yang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Runlin Gao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jinqing Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xueyan Zhao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| |
Collapse
|
93
|
Jasti M, Islam S, Steele N, Ivy K, Maimo W, Isiadinso I. Lp(a) and risk of cardiovascular disease - A review of existing evidence and emerging concepts. J Natl Med Assoc 2023:S0027-9684(23)00141-4. [PMID: 38143155 DOI: 10.1016/j.jnma.2023.11.009] [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: 10/15/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death among adults in the United States. There has been significant advancement in the diagnosis and treatment of atherosclerotic cardiovascular disease (ASCVD) and its underlying risk factors. In certain populations, there remains a significant residual risk despite adequate lowering of low-density lipoprotein cholesterol (LDL-C) and control of traditional risk factors. This has led to an interest in research to identify additional risk factors that contribute to atherosclerotic cardiovascular disease. Elevated lipoprotein (a) [Lp(a)] has been identified as an independent risk factor contributing to an increased risk for CVD. There are also ethnic and racial disparities in Lp(a) inheritance that need to be understood. This paper reviews the current literature on lipoprotein a, proposed mechanisms of actions for cardiovascular disease, recommendations for testing, and the current and emerging therapies for lowering Lp(a).
Collapse
Affiliation(s)
- Manasa Jasti
- Division of Cardiology, University of Tennessee Health Science Center/Ascension Saint Thomas, Nashville, TN, United States
| | - Sabrina Islam
- Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Nathan Steele
- Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Kendra Ivy
- Department of Internal Medicine, Morehouse School of Medicine, Atlanta, GA, United States
| | - Willibroad Maimo
- Division of Cardiology, Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Ijeoma Isiadinso
- Division of Cardiology, Department of Medicine, Center for Heart Disease Prevention, Emory University School of Medicine, Atlanta, GA, United States.
| |
Collapse
|
94
|
Chehab O, Abdollahi A, Whelton SP, Wu CO, Ambale-Venkatesh B, Post WS, Bluemke DA, Tsai MY, Lima JAC. Association of Lipoprotein(a) Levels With Myocardial Fibrosis in the Multi-Ethnic Study of Atherosclerosis. J Am Coll Cardiol 2023; 82:2280-2291. [PMID: 38057070 DOI: 10.1016/j.jacc.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/21/2023] [Accepted: 10/04/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Lipoprotein(a) (Lp[a]) has been identified as an emerging risk factor for adverse cardiovascular (CV) outcomes, including heart failure. However, the connections among Lp(a), myocardial fibrosis (interstitial and replacement), and cardiac remodeling as pathways to CV diseases remains unclear. OBJECTIVES This study investigated the relationship between Lp(a) levels and myocardial fibrosis by cardiac magnetic resonance (CMR) T1 mapping and late gadolinium enhancement, as well as cardiac remodeling by cine CMR, in the MESA (Multi-Ethnic Study of Atherosclerosis) cohort. METHODS The study included 2,040 participants with baseline Lp(a) measurements and T1 mapping for interstitial myocardial fibrosis (IMF) evaluation in 2010. Lp(a) was analyzed as a continuous variable (per log unit) and using clinical cutoff values of 30 and 50 mg/dL. Multivariate linear and logistic regression were used to assess the associations of Lp(a) with CMR measures of extracellular volume (ECV fraction [ECV%]), native T1 time, and myocardial scar, as well as parameters of cardiac remodeling, in 2,826 participants. RESULTS Higher Lp(a) levels were associated with increased ECV% (per log-unit Lp[a]; β = 0.2%; P = 0.007) and native T1 time (per log-unit Lp[a]; β = 4%; P < 0.001). Similar relationships were observed between elevated Lp(a) levels and a higher risk of clinically significant IMF defined by prognostic thresholds per log-unit Lp(a) of ECV% (OR: 1.20; 95% CI: 1.04-1.43) and native T1 (OR: 1.2; 95% CI: 1.1-1.4) equal to 30% and 955 ms, respectively. Clinically used Lp(a) cutoffs (30 and 50 mg/dL) were associated with greater prevalence of myocardial scar (OR: 1.85; 95% CI: 1.1-3.2 and OR: 1.9; 95% CI: 1.1-3.4, respectively). Finally, higher Lp(a) levels were associated with left atrial enlargement and dysfunction. CONCLUSIONS Elevated Lp(a) levels are linked to greater subclinical IMF, increased myocardial scar prevalence, and left atrial remodeling.
Collapse
Affiliation(s)
- Omar Chehab
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ashkan Abdollahi
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Seamus P Whelton
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Colin O Wu
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - David A Bluemke
- Department of Radiology, University of Wisconsin School of Medicine and Public Heath, Madison, Wisconsin, USA
| | - Michael Y Tsai
- Department of Pathology, University of Minnesota, Saint Paul-Minneapolis, Minneapolis, Minnesota, USA
| | - João A C Lima
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA; Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
95
|
Nissen SE, Linnebjerg H, Shen X, Wolski K, Ma X, Lim S, Michael LF, Ruotolo G, Gribble G, Navar AM, Nicholls SJ. Lepodisiran, an Extended-Duration Short Interfering RNA Targeting Lipoprotein(a): A Randomized Dose-Ascending Clinical Trial. JAMA 2023; 330:2075-2083. [PMID: 37952254 PMCID: PMC10641766 DOI: 10.1001/jama.2023.21835] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/04/2023] [Indexed: 11/14/2023]
Abstract
Importance Epidemiological and genetic data have implicated lipoprotein(a) as a potentially modifiable risk factor for atherosclerotic disease and aortic stenosis, but there are no approved pharmacological treatments. Objectives To assess the safety, tolerability, pharmacokinetics, and effects of lepodisiran on lipoprotein(a) concentrations after single doses of the drug; lepodisiran is a short interfering RNA directed at hepatic synthesis of apolipoprotein(a), an essential component necessary for assembly of lipoprotein(a) particles. Design, Setting, and Participants A single ascending-dose trial conducted at 5 clinical research sites in the US and Singapore that enrolled 48 adults without cardiovascular disease and with lipoprotein(a) serum concentrations of 75 nmol/L or greater (or ≥30 mg/dL) between November 18, 2020, and December 7, 2021; the last follow-up visit occurred on November 9, 2022. Interventions Participants were randomized to receive placebo or a single dose of lepodisiran (4 mg, 12 mg, 32 mg, 96 mg, 304 mg, or 608 mg) administered subcutaneously. Main Outcomes and Measures The primary outcome was the safety and tolerability of the single ascending doses of lepodisiran. The secondary outcomes included plasma levels of lepodisiran for 168 days after dose administration and changes in fasting lipoprotein(a) serum concentrations through a maximum follow-up of 336 days (48 weeks). Results Of the 48 participants enrolled (mean age, 46.8 [SD, 11.6] years; 35% were women), 1 serious adverse event occurred. The plasma concentrations of lepodisiran reached peak levels within 10.5 hours and were undetectable by 48 hours. The median baseline lipoprotein(a) concentration was 111 nmol/L (IQR, 78 to 134 nmol/L) in the placebo group, 78 nmol/L (IQR, 50 to 152 nmol/L) in the 4 mg of lepodisiran group, 97 nmol/L (IQR, 86 to 107 nmol/L) in the 12-mg dose group, 120 nmol/L (IQR, 110 to 188 nmol/L) in the 32-mg dose group, 167 nmol/L (IQR, 124 to 189 nmol/L) in the 96-mg dose group, 96 nmol/L (IQR, 72 to 132 nmol/L) in the 304-mg dose group, and 130 nmol/L (IQR, 87 to 151 nmol/L) in the 608-mg dose group. The maximal median change in lipoprotein(a) concentration was -5% (IQR, -16% to 11%) in the placebo group, -41% (IQR, -47% to -20%) in the 4 mg of lepodisiran group, -59% (IQR, -66% to -53%) in the 12-mg dose group, -76% (IQR, -76% to -75%) in the 32-mg dose group, -90% (IQR, -94% to -85%) in the 96-mg dose group, -96% (IQR, -98% to -95%) in the 304-mg dose group, and -97% (IQR, -98% to -96%) in the 608-mg dose group. At day 337, the median change in lipoprotein(a) concentration was -94% (IQR, -94% to -85%) in the 608 mg of lepodisiran group. Conclusions and Relevance In this phase 1 study of 48 participants with elevated lipoprotein(a) levels, lepodisiran was well tolerated and produced dose-dependent, long-duration reductions in serum lipoprotein(a) concentrations. The findings support further study of lepodisiran. Trial Registration ClinicalTrials.gov Identifier: NCT04914546.
Collapse
Affiliation(s)
| | | | - Xi Shen
- Eli Lilly and Company, Indianapolis, Indiana
| | - Kathy Wolski
- Cleveland Clinic Center for Clinical Research, Cleveland, Ohio
| | - Xiaosu Ma
- Eli Lilly and Company, Indianapolis, Indiana
| | - Shufen Lim
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | - Grace Gribble
- Cleveland Clinic Center for Clinical Research, Cleveland, Ohio
| | - Ann Marie Navar
- Department of Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas
| | | |
Collapse
|
96
|
Tan JS, Xu W, Wang J, Li T, Wang L, Zhu J, Yang Y. Aspirin for prevention of cardiovascular disease in patients with high levels of lipoprotein(a): Evidence from two-sample Mendelian randomization analysis. J Evid Based Med 2023; 16:432-434. [PMID: 38047660 DOI: 10.1111/jebm.12562] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 11/19/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023]
Affiliation(s)
- Jiang-Shan Tan
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Xu
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingyang Wang
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianqi Li
- Key Laboratory of Pulmonary Vascular Medicine, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lulu Wang
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Zhu
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanmin Yang
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
97
|
González-Quijano M, Grande-Trillo A, Esteve-Ruiz I, Aranda-Dios A, Sobrino-Márquez JM, Rangel-Sousa D. Elevated Lipoprotein A Levels and Development of Moderate or Severe Cardiac Allograft Vasculopathy in Patients with Heart Transplants. Transplant Proc 2023; 55:2295-2298. [PMID: 37914618 DOI: 10.1016/j.transproceed.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 09/23/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Cardiac allograft vasculopathy (CAV) is a high-incident complication of heart transplant (HT) and is the leading cause of death beyond the first post-HT year. Traditional risk factors have been related to CAV development. Elevated lipoprotein (a) (Lp[a]) is an independent, genetic, and causal risk factor for cardiovascular disease; nonetheless, its association with the development or worsening of CAV in HT has not been firmly established. METHODS An observational nested case-control study including HT recipients under follow-up in a tertiary center. Lipoprotein (a) levels were determined at the time of inclusion. We considered elevated Lp(a) ≥30 mg/dL. We evaluated the association between Lp(a) levels and the presence and severity of CAV (The International Society For Heart And Lung Transplantation [ISHLT] Cardiac Allograft Vasculopathy Grading Scheme), dividing the sample between No or Mild CAV (0-1) and Moderate-Severe CAV (2-3). Routine coronary angiographies were performed the first year after the transplant and were subsequently symptom-driven. RESULTS One hundred fifty patients with HTs were included, with a mean follow-up of 110 ± 77 months. Patients with CAV 2 to 3 presented higher median Lp(a) levels (17 vs 86 mg/dL, P = 0.001). Elevated Lp(a) level was an independent risk factor for developing CAV 2 to 3 (odds ratio 8.57 [95% CI 2.82-26.04]; P < .001). Patients with Lp(a) ≥30 mg also showed an earlier onset compared with those with Lp(a) <30 mg/dL. CONCLUSIONS Our study suggests that Lp(a) may play a role in the development of CAV. Lipoprotein (a) ≥30 mg/dL defines a subgroup of high-risk patients with HTs as portends to earlier onset and more severe CAV. Lipoprotein (a) determination should be a standard-of-care test in patients with HTs.
Collapse
Affiliation(s)
- Marta González-Quijano
- Heart Failure and Heart Transplant Unit, Cardiology Department, Virgen del Rocío University Hospital, Seville, Spain.
| | - Antonio Grande-Trillo
- Heart Failure and Heart Transplant Unit, Cardiology Department, Virgen del Rocío University Hospital, Seville, Spain
| | - Iris Esteve-Ruiz
- Heart Failure and Heart Transplant Unit, Cardiology Department, Virgen del Rocío University Hospital, Seville, Spain
| | - Antonio Aranda-Dios
- Heart Failure and Heart Transplant Unit, Cardiology Department, Virgen del Rocío University Hospital, Seville, Spain
| | - José Manuel Sobrino-Márquez
- Heart Failure and Heart Transplant Unit, Cardiology Department, Virgen del Rocío University Hospital, Seville, Spain
| | - Diego Rangel-Sousa
- Heart Failure and Heart Transplant Unit, Cardiology Department, Virgen del Rocío University Hospital, Seville, Spain
| |
Collapse
|
98
|
Makris A, Barkas F, Sfikakis PP, Liberopoulos E, Filippatos TD, Ray KK, Agouridis AP. Lipoprotein(a), Interleukin-6 inhibitors, and atherosclerotic cardiovascular disease: Is there an association? ATHEROSCLEROSIS PLUS 2023; 54:1-6. [PMID: 37720252 PMCID: PMC10500445 DOI: 10.1016/j.athplu.2023.09.001] [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: 06/20/2023] [Revised: 08/09/2023] [Accepted: 09/06/2023] [Indexed: 09/19/2023]
Abstract
Background and aims Lipoprotein(a) [Lp(a)] and interleuking-6 (IL-6), an inflammation biomarker, have been established as distinct targets of the residual atherosclerotic cardiovascular disease (ASCVD) risk. We aimed to investigate the association between them, and the potential clinical implications in ASCVD prevention. Methods A literature search was conducted in PubMed until December 31st, 2022, using relevant keywords. Results Elevated lipoprotein(a) [Lp(a)] levels constitute the most common inherited lipid disorder associated with ASCVD. Although Lp(a) levels are mostly determined genetically by the LPA gene locus, they may be altered by acute conditions of stress and chronic inflammatory diseases. Considering its resemblance with low-density lipoproteins, Lp(a) is involved in atherosclerosis, but it also exerts oxidative, thrombotic, antifibrinolytic and inflammatory properties. The cardiovascular efficacy of therapies lowering Lp(a) by >90% is currently investigated. On the other hand, interleukin (IL)-1b/IL-6 pathway also plays a pivotal role in atherosclerosis and residual ASCVD risk. IL-6 receptor inhibitors [IL-6(R)i] lower Lp(a) by 16-41%, whereas ongoing trials are investigating their potential anti-atherosclerotic effect. The Lp(a)-lowering effect of IL-6(R)i might be attributed to the inhibition of the IL-6 response elements in the promoter region of the LPA gene. Conclusions Although the effect of IL-6(R)i on Lp(a) levels is inferior to that of available Lp(a)-lowering therapies, the dual effect of the former on both inflammation and apolipoprotein (a) synthesis may prove of equal or even greater significance when it comes ASCVD outcomes. More trials are required to establish IL-6(R)i in ASCVD prevention and elucidate their interplay with Lp(a) as well as its clinical significance.
Collapse
Affiliation(s)
- Anastasios Makris
- School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Fotios Barkas
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Petros P. Sfikakis
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Evangelos Liberopoulos
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | | | - Kausik K. Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Aris P. Agouridis
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Department of Internal Medicine, German Oncology Center, Limassol, Cyprus
| |
Collapse
|
99
|
Jing S, Zhu H. Relationship between lipoprotein(a) and whole blood reducing viscosity: A cross-sectional study. Medicine (Baltimore) 2023; 102:e36236. [PMID: 38050213 PMCID: PMC10695618 DOI: 10.1097/md.0000000000036236] [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: 08/14/2023] [Accepted: 10/31/2023] [Indexed: 12/06/2023] Open
Abstract
Lipoprotein(a) [Lp(a)] has been confirmed as a causal risk factor of atherosclerotic cardiovascular disease, but its role on circulation is not completely clear and is still being explored. Therefore, this study attempts to explore the relationship between Lp(a) and whole blood reducing viscosity (WBRV), to better understand the role of Lp(a) in circulatory and cardiovascular diseases. We retrospectively analyzed the data of consecutive subjects in the physical examination center of the Affiliated Hospital of Ningbo University Medical College from January 2022 to May 2022. Pearson or spearman correlation analysis was used to test the statistical relationship between 2 continuous variables according to whether they are normal; 131 participants were retrospectively enrolled in this study. The low-density lipoprotein concentration was associated with whole blood viscosity at low-shear (R = 0.220, P = .012), middle-shear (R = 0.226, P = .01), and high-shear viscosity (R = 0.212, P = .015), as well as plasma viscosity (RS = 0.207, P = .018). Lp(a) was not associated with whole blood viscosity at low, middle, and high shear rates, but was associated with WBRV at low shear (RS = 0.204, P = .019) and middle shear rates (RS = 0.197, P = .024). Lp(a) is associated with high WBRV, which may impart more insights into the role of Lp(a) in cardiovascular disease.
Collapse
Affiliation(s)
- Sheng Jing
- The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Haibo Zhu
- The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
100
|
Yan X, Gong J, Wang Z, Wang F, Qi C. Association of lipoprotein(a) with left ventricular hypertrophy assessed by electrocardiogram in adults: a large cross-sectional study. Front Endocrinol (Lausanne) 2023; 14:1260050. [PMID: 38098866 PMCID: PMC10720892 DOI: 10.3389/fendo.2023.1260050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/27/2023] [Indexed: 12/17/2023] Open
Abstract
Background and aims Increasing evidence supports a causal relationship between lipoprotein(a) [Lp(a)] and atherosclerotic cardiovascular disease, yet its association with left ventricular hypertrophy (LVH) assessed by electrocardiogram (ECG) remains unknown. The aim of this study was to explore the relationship between Lp(a) and LVH assessed by ECG in general population. Methods and results In this cross-sectional study, we screened 4,052 adults from the participants of the third National Health and Nutrition Examination Survey for analysis. Lp(a) was regarded as an exposure variable. LVH defined by the left ventricular mass index estimated from ECG was considered as an outcome variable. Multivariate logistic regression and restricted cubic spline (RCS) were used to assess the relationship between Lp(a) and LVH. Individuals with LVH had higher Lp(a) compared to individuals without LVH (P< 0.001). In the fully adjusted model, Lp(a) was strongly associated with LVH when as a continuous variable (per 1-unit increment, OR: 1.366, 95% CI: 1.043-1.789, P = 0.024), and higher Lp(a) remained independently associated with a higher risk of LVH when participants were divided into four groups according to quartiles of Lp(a) (Q4 vs Q1, OR: 1.508, 95% CI: 1.185-1.918, P = 0.001). And in subgroup analysis, this association remained significant among participants< 60 years, ≥ 60 years, male, with body mass index< 30 kg/m2, with hypertension and without diabetes (P< 0.05). In addition, we did not observe a nonlinear and threshold effect of Lp(a) with LVH in the RCS analysis (P for nonlinearity = 0.113). Conclusion Lp(a) was closely associated with LVH assessed by ECG in general population.
Collapse
Affiliation(s)
- Xuejiao Yan
- Department of Cardiology, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Jing Gong
- Department of Geriatrics, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangfang Wang
- Department of Cardiology, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Chunjian Qi
- Medical Research Center, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|