1
|
Zendjebil S, Steg PG. PCSK9 Monoclonal Antibodies Have Come a Long Way. Curr Atheroscler Rep 2024; 26:721-732. [PMID: 39384735 DOI: 10.1007/s11883-024-01243-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 10/11/2024]
Abstract
PURPOSE OF THE REVIEW This review examines the pivotal role of monoclonal antibodies against PCSK9 in lipid-lowering therapy, emphasizing their biological and clinical impact. RECENT FINDINGS Randomized controlled trials have validated that PCSK9 monoclonal antibodies (Mabs) effectively reduce LDL-c levels by approximately 50%, even when added to maximal statin therapy. They moreover produce a notable 15-20% relative decrease in major cardiovascular events, with a greater reduction among high-risk patients and no evidence for serious adverse effects, assuaging previous concerns. This review highlights the benefits of PCSK9 Mabs in high cardiovascular risk patients. Despite their efficacy and safety, these therapies are hindered by limited access, and require broader integration into clinical practice to optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Sandra Zendjebil
- Université Paris-Cité, Paris, France
- French Alliance for Cardiovascular Trials (FACT), INSERM U_1148/LVTS, AP-HP, Hôpital Bichat, Paris, France
- Département de Cardiologie, Hôpital Bichat, AP-HP 46 Rue Henri Huchard, 75018, Paris, France
| | - Philippe Gabriel Steg
- Université Paris-Cité, Paris, France.
- French Alliance for Cardiovascular Trials (FACT), INSERM U_1148/LVTS, AP-HP, Hôpital Bichat, Paris, France.
- Département de Cardiologie, Hôpital Bichat, AP-HP 46 Rue Henri Huchard, 75018, Paris, France.
| |
Collapse
|
2
|
Fichtner I, Macchi C, Rizzuto AS, Carugo S, Corsini A, Ruscica M. Lipoprotein(a) and the atherosclerotic burden - Should we wait for clinical trial evidence before taking action? ATHEROSCLEROSIS PLUS 2024; 58:16-23. [PMID: 39435317 PMCID: PMC11492331 DOI: 10.1016/j.athplu.2024.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/23/2024]
Abstract
The fact that lipoprotein(a) levels should be regarded as a causal residual risk factor in the atherosclerotic cardiovascular diseases (ASCVD) is now a no-brainer. This review article aims to summarize the latest evidence supporting the causal role of lipoprotein(a) in ASCVD and the potential strategies to reduce the lipoprotein(a) burden until clinical trial results are available. Epidemiological and genetic data demonstrate the causal link between lipoprotein(a) and increased ASCVD risk. That being said, a specific question comes to mind: "must we wait for outcome trials in order to take action?". Given that lipoprotein(a) levels predict incident ASCVD in both primary and secondary prevention contexts, with a linear risk gradient across its distribution, measuring lipoprotein(a) can unequivocally help identify patients who may later benefit from specific lipoprotein(a)-lowering therapies. This understanding has led various National Societies to recommend dosing lipoprotein(a) in high-risk individuals and to support the recommendation of measuring lipoprotein(a) levels at least once in every adult for risk stratification.
Collapse
Affiliation(s)
- Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | | | - Stefano Carugo
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
3
|
Shah NP, Mulder H, Lydon E, Chiswell K, Hu X, Lampron Z, Cohen L, Patel MR, Taubes S, Song W, Mulukutla SR, Saeed A, Morin DP, Bradley SM, Hernandez AF, Pagidipati NJ. Lipoprotein (a) Testing in Patients With Atherosclerotic Cardiovascular Disease in 5 Large US Health Systems. J Am Heart Assoc 2024; 13:e035610. [PMID: 39494552 DOI: 10.1161/jaha.124.035610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/12/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Lipoprotein (a) is an independent risk factor for atherosclerotic cardiovascular disease. However, lipoprotein (a) testing remains variable and it is unclear what factors influence testing and if testing changes clinical management. METHODS AND RESULTS A retrospective study using electronic medical record data from 5 health systems identified an atherosclerotic cardiovascular disease cohort divided into those with and without a lipoprotein (a) test between 2019 and 2021. Baseline characteristics and lipid-lowering therapy patterns were assessed. Multivariable regression modeling was used to determine factors associated with lipoprotein (a) testing. Among 595 684 patients with atherosclerotic cardiovascular disease, only 2587 (0.4%) were tested for lipoprotein (a). Those who were older or Black individuals were less likely to have lipoprotein (a) testing, while those with familial hypercholesterolemia, ischemic stroke/transient ischemic attack, peripheral artery disease, prior lipid-lowering therapy, or low-density lipoprotein cholesterol ≥130 mg/dL were more likely to be tested. Those with a lipoprotein (a) test, regardless of the lipoprotein (a) value, were more frequently initiated on any statin therapy (30.3% versus 10.6%, P < 0.001), ezetimibe (7.65% versus 0.8%, P < 0.001), or proprotein convertase substilisin/kexin type 9 inhibitor (6.7% versus 0.3%, P < 0.001) compared with those without a test. Those with an elevated lipoprotein (a) level more frequently initiated ezetimibe (11.5% versus 5.9%, P < 0.001) or proprotein convertase substilisin/kexin type 9 inhibitor (10.9% versus 4.8%, P < 0.001). CONCLUSIONS Lipoprotein (a) testing in patients with atherosclerotic cardiovascular disease is infrequent, with evidence of disparities among older or Black individuals. Testing for lipoprotein (a), regardless of level, is associated with greater initiation of any lipid-lowering therapy, while elevated lipoprotein (a) is associated with greater initiation of nonstatin lipid-lowering therapy. There is a critical need for multidisciplinary and inclusive approaches to raise awareness for lipoprotein (a) testing, and its implications on management.
Collapse
Affiliation(s)
- Nishant P Shah
- Division of Cardiology Duke University Hospital Durham NC USA
- Duke Clinical Research Institute Durham NC USA
| | | | | | | | - Xingdi Hu
- Novartis Pharmaceuticals Corporation East Hanover NJ USA
| | | | | | - Manesh R Patel
- Division of Cardiology Duke University Hospital Durham NC USA
- Duke Clinical Research Institute Durham NC USA
| | - Susan Taubes
- Novartis Pharmaceuticals Corporation East Hanover NJ USA
| | - Wenliang Song
- Division of Cardiology Vanderbilt University Nashville TN USA
| | | | - Anum Saeed
- Division of Cardiology University of Pittsburgh Pittsburgh PA USA
| | - Daniel P Morin
- Division of Cardiology Ochsner Medical Center New Orleans LA USA
| | | | - Adrian F Hernandez
- Division of Cardiology Duke University Hospital Durham NC USA
- Duke Clinical Research Institute Durham NC USA
| | - Neha J Pagidipati
- Division of Cardiology Duke University Hospital Durham NC USA
- Duke Clinical Research Institute Durham NC USA
| |
Collapse
|
4
|
Shi W, Feng D, Hu X, Wang C, Niu G, Zhao Z, Zhang H, Wang M, Wu Y. Lipoprotein(a) and High-Sensitivity C-Reactive Protein Compound the Risk of Hypoattenuating Leaflet Thickening After Transcatheter Aortic Valve Replacement. J Am Heart Assoc 2024; 13:e035597. [PMID: 39424417 DOI: 10.1161/jaha.124.035597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/11/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The mechanism for hypoattenuating leaflet thickening (HALT) after transcatheter aortic valve replacement is still not well elucidated, and the role of Lp(a) (lipoprotein[a]) and hs-CRP (high-sensitivity C-reactive protein) has rarely been studied. This study sought to test the hypothesis that the risk of HALT is associated with an elevated level of Lp(a) or hs-CRP. METHODS AND RESULTS A total of 307 consecutive individuals who underwent a transcatheter aortic valve replacement procedure were included. All patients received their first postoperative computed tomography scans within 12 months, and raw data were analyzed on 3mensio software. HALT was defined as visually identified increased leaflet thickness with typical meniscal appearance and at least 2 different multiplanar reformation projections. Associations of Lp(a) or hs-CRP with the risk of HALT were evaluated using multivariable logistic regression analysis. The incidence of HALT within 12 months after transcatheter aortic valve replacement in this study was 36.2%, and the risk of HALT was associated with higher baseline Lp(a) (the multivariable adjusted odds ratio [OR] for every 10 mg/dL change was 1.18 [95% CI, 1.09-1.29]) and hs-CRP level (the multivariable adjusted OR for every 1 mg/L change was 1.08 [95% CI, 1.00-1.27]). Compared with individuals out of the top 25th percentile for both Lp(a) and hs-CRP, the multivariable adjusted OR for HALT was 4.74 (95% CI, 1.65-14.37) for the top 25th percentile. This result remained consistent after excluding patients receiving anticoagulant therapy. CONCLUSIONS The top 25th percentile of Lp(a) level (≥40 mg/dL) combined with the top 25th percentile of hs-CRP level (≥3.5 mg/L) conferred a 4.74-fold risk of HALT.
Collapse
Affiliation(s)
- Wence Shi
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease Chinese Academy of Medical Science and Peking Union Medical College Beijing China
| | - Dejing Feng
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease Chinese Academy of Medical Science and Peking Union Medical College Beijing China
| | - Xiangming Hu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease Chinese Academy of Medical Science and Peking Union Medical College Beijing China
| | - Can Wang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease Chinese Academy of Medical Science and Peking Union Medical College Beijing China
| | - Guannan Niu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease Chinese Academy of Medical Science and Peking Union Medical College Beijing China
| | - Zhenyan Zhao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease Chinese Academy of Medical Science and Peking Union Medical College Beijing China
| | - Hongliang Zhang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease Chinese Academy of Medical Science and Peking Union Medical College Beijing China
| | - Moyang Wang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease Chinese Academy of Medical Science and Peking Union Medical College Beijing China
| | - Yongjian Wu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease Chinese Academy of Medical Science and Peking Union Medical College Beijing China
| |
Collapse
|
5
|
Chong T, Lan NSR, Courtney W, He A, Strange G, Playford D, Dwivedi G, Hillis GS, Ihdayhid AR. Medical Therapy to Prevent or Slow Progression of Aortic Stenosis: Current Evidence and Future Directions. Cardiol Rev 2024; 32:473-482. [PMID: 36961371 DOI: 10.1097/crd.0000000000000528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
Degenerative aortic stenosis is a growing clinical problem owing to the high incidence in an aging population and its significant morbidity and mortality. Currently, aortic valve replacement remains the only treatment. Despite promising observational data, pharmacological management to slow or halt progression of aortic stenosis has remained elusive. Nevertheless, with a greater understanding of the mechanisms which underpin aortic stenosis, research has begun to explore novel treatment strategies. This review will explore the historical agents used to manage aortic stenosis and the emerging agents that are currently under investigation.
Collapse
Affiliation(s)
- Travis Chong
- From the Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, Perth, Australia
| | - Nick S R Lan
- From the Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, Perth, Australia
- Internal Medicine, Medical School, The University of Western Australia, Perth, Australia
| | - William Courtney
- Internal Medicine, Medical School, The University of Western Australia, Perth, Australia
- Department of Cardiology, Royal Perth Hospital, Perth, Australia
| | - Albert He
- From the Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, Perth, Australia
| | - Geoff Strange
- School of Medicine, University of Notre Dame, Fremantle, Australia
| | - David Playford
- School of Medicine, University of Notre Dame, Fremantle, Australia
| | - Girish Dwivedi
- From the Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, Perth, Australia
- Internal Medicine, Medical School, The University of Western Australia, Perth, Australia
| | - Graham S Hillis
- Internal Medicine, Medical School, The University of Western Australia, Perth, Australia
- Department of Cardiology, Royal Perth Hospital, Perth, Australia
| | - Abdul Rahman Ihdayhid
- From the Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, Perth, Australia
- Curtin Medical School, Curtin University, Perth, Australia
| |
Collapse
|
6
|
Lin YC, Lai TS, Chen YT, Chou YH, Chen YM, Hung KY, Tu YK. Comparative efficacy and choice of lipid-lowering drugs for cardiovascular and kidney outcomes in patients with chronic kidney disease: A systematic review and network meta-analysis. J Formos Med Assoc 2024:S0929-6646(24)00474-1. [PMID: 39389802 DOI: 10.1016/j.jfma.2024.09.037] [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: 08/08/2024] [Revised: 09/07/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND The effect of exact classes of lipid-lowering drugs (LLDs) on preventing major adverse cardiovascular events (MACEs) and poor renal outcomes is not well characterized in the chronic kidney disease (CKD) population. METHODS We performed a frequentist random-effects network meta-analysis of randomized controlled trials (RCTs) to evaluate the protective effect of the LLDs in non-dialysis CKD patients. The PubMed, Embase, Web of Science, and Cochrane Library databases were systematically searched for relevant trials published before March 31, 2024. The primary outcome was the incidence of MACEs. The secondary outcomes comprised all-cause mortality, end-stage kidney disease, changes in estimated glomerular filtration rate (eGFR) and proteinuria, and safety. RESULTS Forty-nine eligible RCTs with 77,826 participants with non-dialysis CKD were included. With moderate confidence in the evidence, rosuvastatin and atorvastatin showed statistically significantly more efficacy in reducing the risk of MACE, with a pooled risk ratio of 0.55 (95% CI 0.33-0.91) for rosuvastatin and 0.67 (0.49-0.90) for atorvastatin, respectively, compared with the control group. For the change in the eGFR, atorvastatin (mean difference [MD], 1.40; 95% CI, 0.61 to 2.18), rosuvastatin (MD, 1.73; 95% CI, 0.63 to 2.83), and statin plus ezetimibe (MD, 2.35; 95% CI, 0.44 to 4.26) showed statistically significant increases in the mean eGFR. CONCLUSION In patients with non-dialysis CKD, there is sufficient evidence to show that rosuvastatin and atorvastatin were statistically significantly more effective and preferable in reducing the risk of MACE and increasing the mean eGFR compared with the control group.
Collapse
Affiliation(s)
- Yi-Chih Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Medicine, National Taiwan University Hospital Jinshan Branch, New Taipei City, Taiwan
| | - Tai-Shuan Lai
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Yi-Ting Chen
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Hsiang Chou
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yung-Ming Chen
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuan-Yu Hung
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Kang Tu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan; Health Data Research Center, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
7
|
Fujii E, Ako J, Takahashi Y, Toda M, Iekushi K, Yamashita S. Serum Lipoprotein(a) Levels and Their Association with Atherosclerotic Cardiovascular Disease in Japan. J Atheroscler Thromb 2024:64953. [PMID: 39370270 DOI: 10.5551/jat.64953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024] Open
Abstract
AIMS To investigate the distribution of lipoprotein(a) (Lp(a)) and its association with atherosclerotic cardiovascular disease (ASCVD) in Japanese patients at high risk for ASCVD using a health insurance database. METHODS Between July 2013 and June 2021, patients eligible for ASCVD prevention according to the 2017 Japan Atherosclerosis Society (JAS) guidelines with documented Lp(a) test results were extracted from the Medical Data Vision claims database and divided into three groups: primary prevention high-risk (Group I), secondary prevention (Group II) and secondary prevention high-risk (Group III). Data on lipid levels, cardiovascular morbidity risk factors and lipid-lowering treatments were extracted. RESULTS Of 700,580 patients with documented low-density lipoprotein cholesterol (LDL-C), 2,967 (0.42%) were tested for Lp(a). In 2,170 eligible patients, the median [interquartile range] serum concentration of Lp(a) was 13.9 [7.5-24.6] mg/dL, with 151 patients (7.0%) above the recommended risk threshold of ≥ 50 mg/dL. Lp(a) levels increased with risk across all prevention groups. Being in the highest Lp(a) quintile (Q5) was associated with an increased frequency of ASCVD (28.9% versus 18.9% in the lowest quintile (Q1) for unstable angina; 18.7% versus 10.1% for myocardial infarction; 27.9% versus 17.0% for ischemic stroke). In the secondary prevention groups, the proportion of patients meeting an LDL-C target of <70 mg/dL decreased from 30.2% in Q1 to 19.0% in Q5 for Group II and from 32.9% to 16.3% for Group III. CONCLUSIONS Despite a high prevalence of Lp(a) ≥ 50mg/dL in Japanese patients at high risk for ASCVD, it found that the Lp(a) testing rate was very low.
Collapse
Affiliation(s)
- Emi Fujii
- Medical Affairs Division, Novartis Pharma K.K
| | - Junya Ako
- Department of Cardiovascular Medicine, Kitasato University School of Medicine
| | | | | | | | | |
Collapse
|
8
|
Lian PA, Zhu WQ, Zhao WX, Huang PP, Ran JL, Tang YX, Huang XS, Li R. Lipoprotein(a) in atherosclerotic cardiovascular disease and proprotein convertase subtilisin/kexin-type 9 inhibitors. Clin Chim Acta 2024; 565:119982. [PMID: 39366516 DOI: 10.1016/j.cca.2024.119982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
High plasma lipoprotein(a) (Lp(a)) levels increase the cardiovascular risk in populations with atherosclerotic cardiovascular disease (ASCVD). Apolipoprotein (a) [apo(a)], a unique protein component of Lp(a), plays an important role in the pathogenesis of atherosclerosis. Statins, the primary medication in managing ASCVD, lower low-density lipoprotein cholesterol (LDL-C) but concurrently elevate plasma Lp(a) levels, contributing to an increased residual cardiovascular risk. In turn, proprotein convertase subtilisin/kexin-type 9 (PCSK9) inhibitors, a novel class of LDL-C lowering drugs, effectively reduce plasma Lp(a) levels, which is believed to decrease residual cardiovascular risk. However, the mechanism by which PCSK9 inhibitors reduce Lp(a) levels remains unknown. In addition, there are some clinical limitations of PCSK9 inhibitors. Here, we systematically review the past, present, and prospects of studies pertaining to Lp(a), PCSK9 inhibitors, and ASCVD.
Collapse
Affiliation(s)
- Ping-An Lian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen-Qiang Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei-Xin Zhao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Piao-Piao Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan-Li Ran
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya-Xin Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xian-Sheng Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cardiovascular Medicine, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China
| | - Rong Li
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Stomatology, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China.
| |
Collapse
|
9
|
Ellberg CC, Bhatia HS. Strategies for management of patients with elevated lipoprotein(a). Curr Opin Lipidol 2024; 35:234-240. [PMID: 39145610 PMCID: PMC11387124 DOI: 10.1097/mol.0000000000000950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
PURPOSE OF REVIEW There is growing literature that supports the testing of Lp(a). However, few patients are tested, including those with a personal or family history of cardiovascular disease (CVD). One often noted barrier to more widespread testing is uncertainty regarding what to do with an elevated Lp(a) level. Although guidelines vary, there is agreement on the use of Lp(a) as a risk enhancer to guide medical care and shared decision-making. This review will discuss a clinical approach with supporting evidence for management of patients with elevated Lp(a). RECENT FINDINGS At the minimum, elevated Lp(a) increases cardiovascular risk and can be incorporated into existing risk stratification paradigms. The cornerstone of management is aggressive management of traditional cardiovascular risk factors, including LDL-cholesterol (LDL-C). More recent studies have highlighted the potential role for proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), aspirin in primary prevention, and prolonged dual antiplatelet therapy in secondary prevention. SUMMARY Although there is optimism for Lp(a)-targeted therapies in the near future, an elevated Lp(a) level is actionable today, and uncertainty regarding the management of patients with elevated Lp(a) should not be a barrier to more widespread testing.
Collapse
Affiliation(s)
| | - Harpreet S. Bhatia
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA
| |
Collapse
|
10
|
Cao Zhang AM, Ziogos E, Harb T, Gerstenblith G, Leucker TM. Emerging clinical role of proprotein convertase subtilisin/kexin type 9 inhibition-Part two: Current and emerging concepts in the clinical use of PCSK9 inhibition. Eur J Clin Invest 2024; 54:e14272. [PMID: 38924090 DOI: 10.1111/eci.14272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors have emerged as a novel class of drugs with cardioprotective effects through their lipid-lowering effects. OBJECTIVE This review aims to discuss existing and novel strategies of PCSK9 inhibition, providing an overview of established randomized controlled trials and ongoing outcome trials that assess the efficacy and long-term safety of PCSK9 inhibitors. It also explores the evolving role of PCSK9 beyond lipid metabolism and outlines the pleiotropic actions of PCSK9 inhibition in various disorders and future directions including novel strategies to target PCSK9. CONCLUSION PCSK9 inhibition shows promise not only in lipid metabolism but also in other disease processes, including atherosclerotic plaque remodeling, acute coronary syndrome, stroke, inflammation, and immune response.
Collapse
Affiliation(s)
- Alexander M Cao Zhang
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Efthymios Ziogos
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tarek Harb
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gary Gerstenblith
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thorsten M Leucker
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Rogozik J, Rokicki JK, Grabowski M, Główczyńska R. Gene Mutation in Patients with Familial Hypercholesterolemia and Response to Alirocumab Treatment-A Single-Centre Analysis. J Clin Med 2024; 13:5615. [PMID: 39337102 PMCID: PMC11433266 DOI: 10.3390/jcm13185615] [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: 08/11/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Familial hypercholesterolemia (FH) is an autosomal dominant genetic disorder characterized by significantly elevated levels of low-density lipoprotein (LDL) cholesterol, which plays a major role in the progression of atherosclerosis and leads to a heightened risk of premature atherosclerotic cardiovascular disease. Methods: We have carried out an observational study on a group of 17 patients treated at the Outpatient Lipid Clinic from 2019 to 2024. Result: The most frequent mutation observed was found in the LDL receptor (LDLR) gene, which was identified in ten patients (58.8%). Five patients were identified to have a mutation in the apolipoprotein B (APOB) gene, whereas two patients had two points mutations, one in the LDLR, and the other in the APOB gene. The average age of patients with LDLR mutation was 54.8 (12.3); for APOB mutation it was 61.4 (9.3) and for patients with two points mutation it was 61.5 (14.8). The study results showed that at Week 12, individuals with LDLR-defective heterozygotes who were given alirocumab 150 mg every two weeks experienced a 63.0% reduction in LDL cholesterol levels. On the other hand, individuals with APOB heterozygotes experienced a 59% reduction in LDL cholesterol levels. However, in patients with double heterozygous for mutations in LDLR and APOB genes, there was a hyporesponsiveness to alirocumab, and the reduction in LDL-C was only by 23% in two individuals. Conclusions: In patients with a single mutation, there was a greater response to treatment with alirocumab in contrast to patients with double heterozygous mutation, who did not respond to treatment with PCSK9 inhibitors.
Collapse
Affiliation(s)
- Joanna Rogozik
- 1st Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Jakub Kosma Rokicki
- 1st Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of Medical Informatics and Telemedicine, Medical University of Warsaw, 00-581 Warsaw, Poland
| | - Marcin Grabowski
- 1st Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Renata Główczyńska
- 1st Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
12
|
Sinha M, Maged R, Tarar P, Bandi VV, Koneru HM, Sarwar H. Efficacy of Traditional Anti-lipidemic Drugs in Lowering Lipoprotein(a) Levels: A Systematic Review. Cureus 2024; 16:e69824. [PMID: 39435209 PMCID: PMC11491766 DOI: 10.7759/cureus.69824] [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: 08/18/2024] [Accepted: 09/20/2024] [Indexed: 10/23/2024] Open
Abstract
Lipoprotein(a), or Lp(a), was identified in the early 1960. Its role as an independent risk factor for atherosclerotic cardiovascular disease (ASCVD) became widely recognized by the late 20th century, regardless of other traditional risk markers such as low-density lipoproteins and high-density lipoproteins. This study aimed to systematically review available literature and compare the efficacy of different lipid-lowering drugs, both approved for clinical use and currently undergoing trials, in lowering Lp(a) levels. A comprehensive search of medical databases including PubMed, PubMed Central (PMC), Medline, ScienceDirect, Cochrane Library, and Google Scholar was conducted to identify relevant studies. A total of 29 research papers met the inclusion criteria, focusing on the impact of various lipid-lowering drugs on Lp(a) concentration in patients with significantly elevated baseline Lp(a) levels. Plasma Lp(a) levels exceeding 30 mg/dL are associated with a higher risk of ASCVD, including myocardial infarction, stroke, aortic valve stenosis, heart failure, peripheral arterial disease, and increased all-cause mortality. Most commonly used lipid-lowering agents, such as statins, fibrates, ezetimibe, and nutraceuticals like coenzyme Q10 (CoQ10), showed no significant effect on Lp(a) plasma levels. However, Lp(a) apheresis and proprotein convertase subtilisin/kexin type 9 (PCSK-9) inhibitors were found to effectively reduce plasma Lp(a) concentrations. Emerging therapies targeting apolipoprotein(a) RNA, including anti-sense oligonucleotides (ASO) and small interfering RNA (siRNA), significantly reduced Lp(a) levels in Phase 2 trials. While several lipid-lowering agents have minimal impact on Lp(a) levels, therapies like Lp(a) apheresis, PCSK-9 inhibitors, and novel RNA-targeting drugs show promise in effectively reducing Lp(a) concentrations. However, whether these reductions translate into decreased cardiovascular events remains to be determined.
Collapse
Affiliation(s)
- Mohit Sinha
- Internal Medicine, Jawaharlal Nehru Medical College, Belgaum, IND
| | - Rafik Maged
- Internal Medicine, Ain Shams University, Cairo, EGY
| | - Pakeeza Tarar
- Internal Medicine, Allama Iqbal Medical College, Lahore, PAK
| | | | - Hema Manvi Koneru
- Internal Medicine, Rajiv Gandhi Institute of Medical Sciences, Adilabad, IND
| | - Hooria Sarwar
- Psychiatry, Inside Out CURE Psychiatry LLC, Princeton, USA
| |
Collapse
|
13
|
Siemens R, Pryjma M, Buchkowsky S, Barry AR. Real-world effectiveness of monoclonal antibody inhibitors of PCSK9 in patients with heterozygous familial hypercholesterolemia: A retrospective cohort study. Pharmacotherapy 2024; 44:730-737. [PMID: 39239754 DOI: 10.1002/phar.4609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Heterozygous familial hypercholesterolemia (HeFH) is a genetic condition that is associated with a high risk of atherosclerotic cardiovascular disease (ASCVD) due to elevated lipid levels. Proprotein convertase subtilisin/kexin type 9 (PCSK9) monoclonal antibody inhibitors have been shown to reduce low-density lipoprotein cholesterol (LDL-C) substantially. This study aimed to assess the real-world effectiveness of PCSK9 inhibitor therapy among patients with HeFH. METHODS Retrospective cohort study of patients with probable or definite HeFH on a PCSK9 inhibitor at a specialized lipid clinic between 2015 and 2022. The primary objective was the proportion of patients who attained a ≥50% reduction in LDL-C after 12 months of treatment. RESULTS In total, 141 patients were screened and 95 were included. Mean age was 63 years, 51% were female, and mean baseline LDL-C level was 4.0 mmol/L (155 mg/dL). A majority of patients (60%) had statin intolerance, and 73% were on ezetimibe. The most common PCSK9 inhibitor was evolocumab (94%). Overall, 74% of patients achieved a ≥50% reduction in LDL-C after 12 months of therapy. Mean LDL-C concentration decreased to 1.7 mmol/L (66 mg/dL) (approximately 59% reduction from baseline) after 12 months of follow-up but increased to 1.9 mmol/L (73 mg/dL) after ≥24 months of follow-up. Similar trends were observed in non-high-density lipoprotein cholesterol and apolipoprotein B. Lipoprotein(a) was significantly reduced by 45% over 12 months. Twelve percent of patients permanently discontinued therapy. Barriers to PCSK9i use were mostly related to cost. CONCLUSIONS In a real-world cohort of HeFH patients, most of which were intolerant to statins, a high majority were able to achieve a ≥50% reduction in LDL-C after 12 months of PCSK9 inhibitor therapy (mean reduction of approximately 59%), which is similar to clinical trial data of patients with ASCVD. A significant reduction in non-high-density lipoprotein cholesterol, apolipoprotein B, and lipoprotein(a) were also observed.
Collapse
Affiliation(s)
- Rebecca Siemens
- Surrey Memorial Hospital, Fraser Health (Lower Mainland Pharmacy Services), Surrey, British Columbia, Canada
| | - Mark Pryjma
- Jim Pattison Outpatient Care and Surgery Centre, Fraser Health (Lower Mainland Pharmacy Services), Surrey, British Columbia, Canada
| | - Susan Buchkowsky
- Richmond Hospital, Vancouver Coastal Health (Lower Mainland Pharmacy Services), Richmond, British Columbia, Canada
| | - Arden R Barry
- Jim Pattison Outpatient Care and Surgery Centre, Fraser Health (Lower Mainland Pharmacy Services), Surrey, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
14
|
Adrichem R, van den Dorpel MMP, Hirsch A, Geleijnse ML, Budde RPJ, Van Mieghem NM. Moderate Aortic Stenosis-Advanced Imaging, Risk Assessment, and Treatment Strategies. STRUCTURAL HEART : THE JOURNAL OF THE HEART TEAM 2024; 8:100279. [PMID: 39290682 PMCID: PMC11403096 DOI: 10.1016/j.shj.2023.100279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/02/2023] [Accepted: 12/21/2023] [Indexed: 09/19/2024]
Abstract
Moderate aortic stenosis is increasingly recognized as a disease entity with poor prognosis. Diagnosis of moderate aortic stenosis may be complemented by laboratory tests and advanced imaging techniques focused at detecting signs of cardiac damage such as increase of cardiac enzymes (N-terminal pro-B-type Natriuretic Peptide, troponin), left ventricular remodeling (hypertrophy, reduced left ventricular ejection fraction), or myocardial fibrosis. Therapy should include guideline-directed optimal medical therapy for heart failure. Patients with signs of cardiac damage may benefit from early intervention, which is the focus of several ongoing randomized controlled trials. As yet, no evidence-based therapy exists to halt the progression of aortic valve calcification.
Collapse
Affiliation(s)
- Rik Adrichem
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mark M P van den Dorpel
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Alexander Hirsch
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marcel L Geleijnse
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ricardo P J Budde
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicolas M Van Mieghem
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
15
|
Safarova M, Bimal T, Soffer DE, Hirsh B, Shapiro MD, Mintz G, Cha A, Gianos E. Advances in targeting LDL cholesterol: PCSK9 inhibitors and beyond. Am J Prev Cardiol 2024; 19:100701. [PMID: 39070027 PMCID: PMC11278114 DOI: 10.1016/j.ajpc.2024.100701] [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: 01/17/2024] [Revised: 06/07/2024] [Accepted: 06/22/2024] [Indexed: 07/30/2024] Open
Abstract
There is a direct relationship between the duration and level of exposure to low density lipoprotein cholesterol (LDL-C) levels over one's lifespan and cardiovascular events. Early treatment to lower elevated LDL-C is crucial for better outcomes with multiple therapies currently available to reduce atherogenic lipoproteins. Statins remain the foundation of LDL-C lowering therapy as one of the most cost-effective drugs to reduce atherosclerotic events (ASCVD) and mortality. Nonetheless, LDL-driven goal attainment remains suboptimal globally, highlighting a considerable need for non-statin therapies to address residual risk related to statin intolerance, non-adherence, and inherited lipoprotein disorders. LDL-C lowering interventions beyond statins include ezetimibe, PCSK9 monoclonal antibodies, inclisiran and bempedoic acid with specific guideline recommendations as to when to consider each. For patients with homozygous familial hypercholesterolemia requiring more advanced therapy, lomitapide and evinacumab are available, providing mechanisms that are not LDL receptor dependent. Lipoprotein apheresis remains an effective option for clinical familial hypercholesterolemia as well as elevated lipoprotein (a). There are investigational therapies being explored to add to our current armamentarium including CETP inhibitors, a third-generation PCSK9 inhibitor (small recombinant fusion protein oral PCSK9 inhibitor) and gene editing which aims to directly restore or disrupt genes of interest at the DNA level. This article is a brief review of the pharmacotherapy options beyond statins for lowering LDL-C and their impact on ASCVD risk reduction. Our primary aim is to guide physicians on the role these therapies play in achieving appropriate LDL-C goals, with an algorithm of when to consider each based on efficacy, safety and outcomes.
Collapse
Affiliation(s)
- Maya Safarova
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI USA
| | - Tia Bimal
- Northwell, New Hyde Park, NY, Cardiovascular Institute, Lenox Hill Hospital, USA
| | - Daniel E. Soffer
- Department of Internal Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Benjamin Hirsh
- Department of Cardiology, Donald and Barbara Zucker School of Medicine at Hofstra/ Northwell, Hempstead, NY, USA
- Northwell, New Hyde Park, NY, Cardiovascular Institute, Sandra Atlas Bass Heart Hospital, USA
| | - Michael D. Shapiro
- Center for the Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Guy Mintz
- Department of Cardiology, Donald and Barbara Zucker School of Medicine at Hofstra/ Northwell, Hempstead, NY, USA
- Northwell, New Hyde Park, NY, Cardiovascular Institute, Sandra Atlas Bass Heart Hospital, USA
| | - Agnes Cha
- Northwell/Vivo Health Pharmacy, Ambulatory Pharmacy Services, Lake Success, NY, USA
| | - Eugenia Gianos
- Northwell, New Hyde Park, NY, Cardiovascular Institute, Lenox Hill Hospital, USA
- Department of Cardiology, Donald and Barbara Zucker School of Medicine at Hofstra/ Northwell, Hempstead, NY, USA
| |
Collapse
|
16
|
Zhang Y, Pei Z, Chen B, Qu Y, Dong X, Yu B, Wang G, Xu F, Lu D, He Z, Chen B, Ma L, Wang M, Li B, Xia M, Zheng B, Huo Y. Ebronucimab in Chinese patients with hypercholesterolemia---A randomized double-blind placebo-controlled phase 3 trial to evaluate the efficacy and safety of ebronucimab. Pharmacol Res 2024; 207:107340. [PMID: 39111557 DOI: 10.1016/j.phrs.2024.107340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/25/2024]
Abstract
Randomized clinical trials (RCTs) of PCSK9 monoclonal antibody(mAb) specifically for Chinese patients have been limited. This multi-center RCT is to clarify the efficacy and safety of a novel mAb, Ebronucimab, in Chinese patients. Patients diagnosed with primary hypercholesterolemia, including Heterozygous Familial Hypercholesterolemia, or mixed dyslipidemia, were categorized by ASCVD risk and randomly assigned at a ratio of 2:1:2:1 to receive Ebronucimab 450 mg or matching placebo every 4 weeks (Q4W), or Ebronucimab 150 mg or matching placebo every 2 weeks (Q2W). The primary outcome was the percentage change of LDL-C from baseline to week 12 for all groups. The least squares mean reduction difference (95 %CI) in LDL-C from baseline to week 12 of Ebronucimab 450 mg Q4W and Ebronucimab 150 mg Q2W groups versus the placebo group was -59.13 (-64.103, -54.153) (Adjusted p<0.0001) and -60.43 (-65.450, -55.416) (Adjusted p<0.0001), respectively. Meanwhile, the Ebronucimab group exhibited notably high rates in reaching LDL-C goals of each cardiovascular risk stratification. In addition, Ebronucimab effectively improved other lipid panel. During the double-blind treatment period, relatively frequently reported adverse events (AEs) were injection site reactions (ISR), urinary tract infection, and hyperuricemia (Incidence rate are 6.9 %, 4.8 % and 3.5 %). Among treatment-associated AEs, only injection site reactions (ISR) occurred more in the dose groups. In conclusion, Ebronucimab, with either 450 mg Q4W or 150 mg Q2W doses, demonstrated significant efficacy in lowering serum LDL-C level with a favorable safety and immunogenicity profile among hypercholesterolemic patients.
Collapse
Affiliation(s)
- Yanyan Zhang
- Peking University First Hospital, Department of Cardiology, Beijing 100034, China
| | - Zhaohui Pei
- The Third Hospital of Nanchang, The Second Department of Cardiology, Nanchang 200072, China
| | - Beijian Chen
- Heze Municipal Hospital, Department of Cardiology, Heze 274099, China
| | - Yanling Qu
- Yuncheng Central Hospital, Department of Cardiology, Yuncheng 044099, China
| | - Xiaolin Dong
- Jinan Central Hospital, Affiliated to Shandong University, No. 105, Jinan 250013, China
| | - Binge Yu
- Akeso Biopharma, Inc., Zhongshan, China
| | | | - Fang Xu
- Akeso Biopharma, Inc., Zhongshan, China
| | | | - Zhimei He
- Akeso Biopharma, Inc., Zhongshan, China
| | | | - Lei Ma
- Akeso Biopharma, Inc., Zhongshan, China
| | - Max Wang
- Akeso Biopharma, Inc., Zhongshan, China
| | | | | | - Bo Zheng
- Peking University First Hospital, Department of Cardiology, Beijing 100034, China.
| | - Yong Huo
- Peking University First Hospital, Department of Cardiology, Beijing 100034, China
| |
Collapse
|
17
|
Eidensohn Y, Bhatla A, Ding J, Blumenthal RS, Martin SS, Marvel FA. Testing practices and clinical management of lipoprotein(a) levels: A 5-year retrospective analysis from the Johns Hopkins Hospital. Am J Prev Cardiol 2024; 19:100686. [PMID: 39070024 PMCID: PMC11278112 DOI: 10.1016/j.ajpc.2024.100686] [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: 01/04/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Objective Elevated lipoprotein(a) [Lp(a)] is an independent, genetically determined risk factor for atherosclerotic cardiovascular disease (ASCVD). We evaluated the frequency of testing for elevated Lp(a) and subsequent management at the Johns Hopkins Hospital, a large academic medical center, over a 5-year period. Methods The Johns Hopkins Hospital (JHH) electronic medical record was queried to identify patients with an encounter between 2017 and 2021, either with established ASCVD or at increased risk, defined as being on any lipid lowering medication or having LDL-C ≥ 190 mg/dL. The frequency of Lp(a) testing and of elevated levels were identified for each year. Results Among 111,350 unique adult patients, 2,785 (2.5 %) had at least one Lp(a) test. Patients with Lp(a) testing, compared to those without testing, were younger (mean age 56 years vs. 66 years), more often female (49 % vs. 44 %), Black (24.7 % vs. 24.6 %) or "other" race/ethnicity (12 % vs 10 %), and had higher LDL-C levels (median 118 vs. 91 mg/dL; p < 0.001). The number and frequency of Lp(a) testing increased from 167 (0.57 %) in 2017 to 1155 (5.67 %) in 2021. Lp(a) levels were abnormal in 43.4 % of patients (moderate [75-125 nmol/L]: 10.3 %, high [126-600 nmol/L]: 32.2 %, severe [>600 nmol/L]: 0.9 %). Among 920 patients with high or severe Lp(a) levels, 200 (22 %) had a subsequent referral to cardiology or lipid specialist, and 180 (20 %) had a new lipid-lowering medication prescribed in the subsequent 18 months. Conclusion Based on a single-center experience, the frequency of incident Lp(a) testing among increased-risk patients was low but increased significantly over 5-years, likely due to Lipid Clinic referrals with reflex Lp(a) testing and greater awareness about this risk factor. Future work should target appropriate population based Lp(a) testing strategies and clinical decision-making regarding risk management once Lp(a) elevation is diagnosed.
Collapse
Affiliation(s)
- Yehuda Eidensohn
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Anjali Bhatla
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Jie Ding
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Roger S. Blumenthal
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Seth S. Martin
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Francoise A. Marvel
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
18
|
Yang J, Zhang R, Han B, Li H, Wang J, Xiao Y, Yu X, Guan S, Dai C, Yan H, Jiang T, Cui H, Yang S, Zheng Z, Dong Y, Wang A, Su G, Wang Y. Atherogenic lipid profile in patients with statin treatment after acute coronary syndrome: a real-world analysis from Chinese cardiovascular association database. Lipids Health Dis 2024; 23:271. [PMID: 39198852 PMCID: PMC11351259 DOI: 10.1186/s12944-024-02244-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Adverse atherogenic lipid profile is associated with an increased risk of major adverse cardiac events in patients after acute coronary syndrome (ACS). Knowledge regarding the impact of statins on lipid profile remains limited. METHODS We retrospectively analysed multicenter, real-world data from the Chinese Cardiovascular Association Database-iHeart Project. Patients with a primary diagnosis of ACS from 2014 to 2021 during index hospitalisation and having at least one lipid panel record after discharge within 12 months were enrolled. We analysed target achievement of atherogenic lipid profile, including apolipoprotein B (< 80 mg/dL), low-density lipoprotein cholesterol (LDL-C) (< 1.8 mmol/L), lipoprotein(a) [Lp(a)] (< 30 mg/dL), triglycerides (< 1.7 mmol/L), remnant cholesterol (RC) (< 0.78 mmol/L), non-high-density lipoprotein cholesterol (< 2.6 mmol/L) at baseline and follow-up. Multivariate Cox regression models were employed to investigate the association between patient characteristics and target achievement. RESULTS Among 4861 patients, the mean age was 64.9 years. Only 7.8% of patients had all atherogenic lipids within the target range at follow-up. The proportion of target achievement was for LDL-C 42.7%, Lp(a) 73.3%, and RC 78.5%. Patients with female sex, younger age, myocardial infarction, hypertension, and hypercholesteremia were less likely to control LDL-C, Lp(a), and RC. An increase in the burden of comorbidities was negatively associated with LDL-C and Lp(a) achievements but not with RC. CONCLUSIONS A substantial gap exists between lipid control and the targets recommended by contemporary guidelines. Novel therapeutics targeting the whole atherogenic lipid profile will be warranted to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Jing Yang
- Department of Cardiology, Zhongshan-Xuhui Hospital, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, 200237, China.
| | - Rui Zhang
- China Heart House, Suzhou, 215124, China
| | - Bing Han
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical School of Southeast University, Xuzhou, Jiangsu, 221009, China
| | - Hui Li
- Department of Cardiology, the second affiliated hospital of Soochow University, Suzhou, 215004, China
| | - Jingfeng Wang
- Department of Cardiovascular Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yihui Xiao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, China
| | - Xiaofan Yu
- Department of Cardiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230001, China
| | - Shaofeng Guan
- Department of Cardiology, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Cuilian Dai
- School of Medicine, Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361000, China
| | - Hua Yan
- Department of Cardiology, Wuhan Asia Heart Hospital, Wuhan, 430022, China
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Hanbin Cui
- Cardiology Center, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Shuang Yang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Zeqi Zheng
- Department of Cardiology, the first affiliated hospital of Nanchang University, Nanchang, 330006, China
| | - Yugang Dong
- Department of Cardiology, the first affiliated hospital of Sun Yat-sen University, Guangzhou, 510080, China
- NHC key Laboratory of assisted Circulation, Sun Yat-sen University, Guangzhou, 510080, China
| | - Annai Wang
- China Heart House, Suzhou, 215124, China
| | - Guohai Su
- Department of Cardiology, Jinan Central Hospital, Shandong First Medical University, Shandong, 250013, China.
| | - Yan Wang
- Department of Cardiology, Xiamen Cardiovascular Hospital Xiamen University, Xiamen, 200080, China.
| |
Collapse
|
19
|
Szarek M, Reijnders E, Steg PG, Jukema JW, Schwertfeger M, Bhatt DL, Bittner VA, Diaz R, Fazio S, Garon G, Goodman SG, Harrington RA, White HD, Zeiher AM, Cobbaert C, Schwartz GG. Comparison of change in lipoprotein(a) mass and molar concentrations by alirocumab and risk of subsequent cardiovascular events in ODYSSEY OUTCOMES. Eur J Prev Cardiol 2024; 31:e75-e78. [PMID: 38501249 DOI: 10.1093/eurjpc/zwae110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/19/2024] [Accepted: 03/11/2024] [Indexed: 03/20/2024]
Affiliation(s)
- Michael Szarek
- Division of Cardiology, University of Colorado School of Medicine, Box B130, Aurora, CO 80045, USA
- CPC Clinical Research, 2115 N. Scranton Street, Suite 2040, Aurora, CO 80045, USA
- Department of Epidemiology and Biostatistics, State University of New York, Downstate Health Sciences University School of Public Health, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Esther Reijnders
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Centre, LUMC Main Building, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Ph Gabriel Steg
- Université Paris-Cité, 85 boulevard Saint-Germain, 75006 Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, FACT (French Alliance for Cardiovascular Trials), INSERM U1148, 46 rue Henri Huchard, 75018 Paris, France
- Institut Universitaire de France, 1 rue Descartes, 75005 Paris, France
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
- Netherlands Heart Institute, Moreelsepark 1, 3511 EP Utrecht, The Netherlands
| | - Markus Schwertfeger
- RCardiometabolism, Neuroscience, Endocrinology, PHCS, PoC, RDS Clinical Development and Medical Affairs (CDMA), Roche Diagnostics International Ltd., Building 5/Floor 10, Forrenstrasse 2, 6343 Rotkreuz, Switzerland
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Health System, 1 Gustave Levy Place, Box 1030, New York, NY 10029, USA
| | - Vera A Bittner
- Division of Cardiovascular Disease, University of Alabama at Birmingham, 701 19th Street South - LHRB 310, Birmingham, AL 35294, USA
| | - Rafael Diaz
- Estudios Cardiológicos Latino América, Instituto Cardiovascular de Rosario, Paraguay 160, Rosario, Santa Fe, Rosario 2000, Argentina
| | - Sergio Fazio
- Scientific Council -Cardiometabolic Global Development, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Genevieve Garon
- Global Medical Department - General Medicines GBU, Sanofi, Ontario M2R 3T4, Canada
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, 87 Ave NW, Edmonton, Alberta T6G 2E1, Canada
- Division of Cardiology, St. Michael's Hospital, Room 6-034 Donnelly Wing, Toronto, Ontario M5B 1W8, Canada
| | - Robert A Harrington
- Stephen and Suzanne Weiss Dean, Weill Cornell Medicine, 1300 York Avenue, F-113, New York, NY 10065, USA
| | - Harvey D White
- Green Lane Cardiovascular Services, Auckland City Hospital, 5 Park Road, Grafton, Auckland 1142, New Zealand
| | - Andreas M Zeiher
- Department of Medicine III, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany
| | - Christa Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Centre, LUMC Main Building, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Gregory G Schwartz
- Division of Cardiology, University of Colorado School of Medicine, Box B130, Aurora, CO 80045, USA
| |
Collapse
|
20
|
Renkens MPL, Coerkamp CF, Witte LS, Sivanesan S, Nurmohamed NS, Westerterp M, Serruys P, Onuma Y, Grundeken MJ, Kalkman DN, Beijk M, Vis MM, Henriques JPS, Delewi R, Stroes E, Wykrzykowska JJ, de Winter RJ, Claessen BEPM. Lipoprotein(a) in interventional cardiology: identifying patients at highest risk of recurrent cardiovascular events through early recognition - a case based review. Expert Rev Cardiovasc Ther 2024; 22:353-366. [PMID: 39258965 DOI: 10.1080/14779072.2024.2387678] [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: 06/26/2024] [Accepted: 07/30/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Lipoprotein(a) [Lp(a)] is linked to higher risks of atherosclerotic cardiovascular disease (ASCVD). Current guideline recommendations are quite liberal on measuring Lp(a) (Class IIa, Level C), and may lead to underuse among (interventional) cardiologists. AREAS COVERED This case-based narrative review outlines four clinical cases of patients with elevated Lp(a) to illustrate its pathophysiological impact on coronary artery disease (CAD). The expert consensus statements from the American Heart Association (AHA) and European Atherosclerosis Society (EAS) served as the basis of this review. More recent publications, from 2023 to 2024, were accessed through the MEDLINE online library. EXPERT OPINION We highlighted the importance of routine Lp(a) measurement in identifying patients at high risk for atherosclerosis, necessitating potent risk mitigation. Measuring Lp(a) helps clinicians identify which patients are at highest residual risk, who require potent pharmacological treatment and special attention during catheter interventions. As noninvasive and advanced intravascular imaging modalities evolve, future catheterization laboratories will integrate advanced imaging, diagnostics, and treatment, facilitating tailored patient care. Knowing Lp(a) levels is crucial in this context. While Lp(a)-lowering drugs are currently investigated in clinical trials, it is of paramount importance to know Lp(a) levels and strive toward aggressive management of other modifiable risk factors in patients with elevated Lp(a) and established symptomatic CAD being diagnosed or treated in catheterization laboratories.
Collapse
Affiliation(s)
- Mick P L Renkens
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Casper F Coerkamp
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Lars S Witte
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Shabiga Sivanesan
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Nick S Nurmohamed
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Marit Westerterp
- Department of Pediatrics, University Medical Centre Groningen, Groningen, The Netherlands
| | - Patrick Serruys
- CORRIB Research Centre for Advanced Imaging and Core Laboratory, University of Galway, Galway, Ireland
| | - Yoshinobu Onuma
- CORRIB Research Centre for Advanced Imaging and Core Laboratory, University of Galway, Galway, Ireland
| | - Maik J Grundeken
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Deborah N Kalkman
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Marcel Beijk
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Marije M Vis
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - José P S Henriques
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Ronak Delewi
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Robbert J de Winter
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Bimmer E P M Claessen
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Afzal Z, Cao H, Chaudhary M, Chigurupati HD, Neppala S, Alruwaili W, Awad M, Sandesara D, Siddique M, Farman A, Zafrullah F, Gonuguntla K, Sattar Y. Elevated lipoprotein(a) levels: A crucial determinant of cardiovascular disease risk and target for emerging therapies. Curr Probl Cardiol 2024; 49:102586. [PMID: 38653440 DOI: 10.1016/j.cpcardiol.2024.102586] [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: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 04/25/2024]
Abstract
Cardiovascular disease (CVD) remains a significant global health challenge despite advancements in prevention and treatment. Elevated Lipoprotein(a) [Lp(a)] levels have emerged as a crucial risk factor for CVD and aortic stenosis, affecting approximately 20 of the global population. Research over the last decade has established Lp(a) as an independent genetic contributor to CVD and aortic stenosis, beginning with Kare Berg's discovery in 1963. This has led to extensive exploration of its molecular structure and pathogenic roles. Despite the unknown physiological function of Lp(a), studies have shed light on its metabolism, genetics, and involvement in atherosclerosis, inflammation, and thrombosis. Epidemiological evidence highlights the link between high Lp(a) levels and increased cardiovascular morbidity and mortality. Newly emerging therapies, including pelacarsen, zerlasiran, olpasiran, muvalaplin, and lepodisiran, show promise in significantly lowering Lp(a) levels, potentially transforming the management of cardiovascular disease. However, further research is essential to assess these novel therapies' long-term efficacy and safety, heralding a new era in cardiovascular disease prevention and treatment and providing hope for at-risk patients.
Collapse
Affiliation(s)
- Zeeshan Afzal
- Department of Medicine, Shanxi Medical University, China
| | - Huili Cao
- Department of Cardiology, Second Hospital of Shanxi Medical University, China
| | | | - Himaja Dutt Chigurupati
- Department of Internal Medicine, New York Medical College at Saint Michael's Medical Center, NJ, USA
| | - Sivaram Neppala
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Waleed Alruwaili
- Department of Internal Medicine, West Virginia University, Morgantown, WV, USA
| | - Maan Awad
- Department of Internal Medicine, West Virginia University, Morgantown, WV, USA
| | | | | | - Ali Farman
- Department of Medicine, Corewell Health Dearborn Hospital, Dearborn, MI, USA
| | - Fnu Zafrullah
- Department of Cardiology, Ascension Borgess Hospital, MI, USA
| | | | - Yasar Sattar
- Department of Cardiology, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
22
|
Terasaka K, Gohbara M, Abe T, Yoshii T, Hanajima Y, Kirigaya J, Horii M, Kikuchi S, Nakahashi H, Matsushita K, Minamimoto Y, Okada K, Matsuzawa Y, Iwahashi N, Kosuge M, Sugano T, Ebina T, Hibi K. Association between evolocumab use and slow progression of aortic valve stenosis. Heart Vessels 2024; 39:725-734. [PMID: 38499696 DOI: 10.1007/s00380-024-02386-6] [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: 10/31/2023] [Accepted: 02/28/2024] [Indexed: 03/20/2024]
Abstract
No medications have been reported to inhibit the progression of aortic valve stenosis (AS). The present study aimed to investigate whether evolocumab use is related to the slow progression of AS evaluated by serial echocardiography. This was a retrospective observational study from 2017 to 2022 at Yokohama City University Medical Center. Patients aged ≥ 18 with moderate AS were included. Exclusion criteria were (1) mild AS; (2) severe AS defined by maximum aortic valve (AV) velocity ≥ 4.0 m/s; and/or (3) no data of annual follow-up echocardiography. The primary endpoint was the association between evolocumab use and annual changes in the maximum AV-velocity or peak AV-pressure gradient (PG). A total of 57 patients were enrolled: 9 patients treated with evolocumab (evolocumab group), and the other 48 patients assigned to a control group. During a median follow-up of 33 months, the cumulative incidence of AS events (a composite of all-cause death, AV intervention, or unplanned hospitalization for heart failure) was 11% in the evolocumab group and 58% in the control group (P = 0.012). Annual change of maximum AV-velocity or peak AV-PG from the baseline to the next year was 0.02 (- 0.18 to 0.22) m/s per year or 0.60 (- 4.20 to 6.44) mmHg per year in the evolocumab group, whereas it was 0.29 (0.04-0.59) m/s per year or 7.61 (1.46-16.48) mmHg per year in the control group (both P < 0.05). Evolocumab use was associated with slow progression of AS and a low incidence of AS events in patients with moderate AS.
Collapse
Affiliation(s)
- Kengo Terasaka
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Advanced Critical Care and Emergency Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Masaomi Gohbara
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
| | - Takeru Abe
- Advanced Critical Care and Emergency Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Tomohiro Yoshii
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Yohei Hanajima
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Jin Kirigaya
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Advanced Critical Care and Emergency Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Mutsuo Horii
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Department of Laboratory Medicine and Clinical Investigation, Yokohama City University Medical Center, Yokohama, Japan
| | - Shinnosuke Kikuchi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Hidefumi Nakahashi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Kensuke Matsushita
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Yugo Minamimoto
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Advanced Critical Care and Emergency Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Kozo Okada
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Yasushi Matsuzawa
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Noriaki Iwahashi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Department of Cardiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masami Kosuge
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Teruyasu Sugano
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Toshiaki Ebina
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Department of Laboratory Medicine and Clinical Investigation, Yokohama City University Medical Center, Yokohama, Japan
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Department of Cardiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
23
|
Haskiah F, Abdelhai K, Hilu R, Khaskia A. Sex Differences in Low-Density Lipoprotein Cholesterol Treatment Among Young Israeli Patients Following Premature Acute Coronary Syndrome. Metab Syndr Relat Disord 2024; 22:439-446. [PMID: 38546443 DOI: 10.1089/met.2023.0310] [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: 08/02/2024] Open
Abstract
Introduction: Effective management of dyslipidemias is crucial for reducing morbidity and mortality among patients after acute coronary syndrome (ACS). Sex differences in dyslipidemia management after premature ACS in Israeli patients have not been extensively studied. This study aimed to investigate potential disparities between men and women in managing dyslipidemia, considering current guidelines. Methods: This retrospective cohort study examined patients who were 55 years old or younger and admitted to Meir Medical Center for ACS from January 2018 to February 2019. The study aimed to evaluate the use of lipid-lowering therapy (LLT), measure the achievement of target low-density lipoprotein cholesterol (LDL-C) levels, and analyze the occurrence of major adverse cardiovascular and cerebrovascular events (MACCE) in both male and female patients. Results: The study included a total of 687 participants, of which 23.3% were identified as females. Upon discharge, ∼80% of the patients were prescribed high-intensity statins. After 1 year, it was observed that females had higher levels of LDL-C and lower rates of achieving target LDL-C levels (<70 and 55 mg/dL) as compared with males (45% vs. 54.6% and 30% vs. 42.2%, respectively). The use of non-statin LLT at the 1-year mark was minimal in both groups. Finally, it was found that the occurrence of MACCE was similar between males and females. Conclusion: Sex disparities in dyslipidemia management after a premature ACS were apparent, with females having higher LDL-C levels and lower rates of target achievement. Intervention is necessary to address these disparities and encourage greater use of non-statin LLT.
Collapse
Affiliation(s)
- Feras Haskiah
- Department of Internal Medicine D, Meir Medical Center, Kfar Saba, Israel
- Leviev Heart Center, Chaim Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Karam Abdelhai
- Department of Internal Medicine D, Meir Medical Center, Kfar Saba, Israel
| | - Ranin Hilu
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Cardiology, Meir Medical Center, Kfar Saba, Israel
| | - Abid Khaskia
- Department of Cardiology, Meir Medical Center, Kfar Saba, Israel
| |
Collapse
|
24
|
Goodman SG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Harrington RA, Jukema JW, White HD, Zeiher AM, Manvelian G, Pordy R, Poulouin Y, Stipek W, Garon G, Schwartz GG. Safety of the PCSK9 inhibitor alirocumab: insights from 47 296 patient-years of observation. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2024; 10:342-352. [PMID: 38658193 PMCID: PMC11249957 DOI: 10.1093/ehjcvp/pvae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/25/2024] [Accepted: 04/23/2024] [Indexed: 04/26/2024]
Abstract
The ODYSSEY OUTCOMES trial, comprising over 47 000 patient-years of placebo-controlled observation, demonstrated important reductions in the risk of recurrent ischaemic cardiovascular events with the monoclonal antibody to proprotein convertase subtilisin/kexin type 9 alirocumab, as well as lower all-cause death. These benefits were observed in the context of substantial and persistent lowering of low-density lipoprotein cholesterol with alirocumab compared with that achieved with placebo. The safety profile of alirocumab was indistinguishable from matching placebo except for a ∼1.7% absolute increase in local injection site reactions. Further, the safety of alirocumab compared with placebo was evident in vulnerable groups identified before randomization, such as the elderly and those with diabetes mellitus, previous ischaemic stroke, or chronic kidney disease. The frequency of adverse events and laboratory-based abnormalities was generally similar to that in placebo-treated patients. Thus, alirocumab appears to be a safe and effective lipid-modifying treatment over a duration of at least 5 years.
Collapse
Affiliation(s)
- Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
- St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Philippe Gabriel Steg
- Université Paris-Cité, Institut Universitaire de France, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, FACT (French Alliance for Cardiovascular Trials), and INSERM U1148, F-75018 Paris, France
| | - Michael Szarek
- CPC Clinical Research and Division of Cardiology, University of Colorado School of Medicine, Aurora, 80045 CO, USA
- State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vera A Bittner
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Rafael Diaz
- Estudios Cardiológicos Latinoamérica, Instituto Cardiovascular de Rosario, S2000 Rosario, Argentina
| | | | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Netherlands Heart Institute, 3511 EP Utrecht, The Netherlands
| | - Harvey D White
- Green Lane Cardiovascular Research Unit, Te Whatu Ora—Health New Zealand, Te Toka Tumai, and University of Auckland, Auckland 1030, New Zealand
| | - Andreas M Zeiher
- Department of Medicine III, Goethe University, 60596 Frankfurt am Main, Germany
| | | | - Robert Pordy
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | | | - Wanda Stipek
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | | | | |
Collapse
|
25
|
Delgado-Lista J, Mostaza JM, Arrobas-Velilla T, Blanco-Vaca F, Masana L, Pedro-Botet J, Perez-Martinez P, Civeira F, Cuende-Melero JI, Gomez-Barrado JJ, Lahoz C, Pintó X, Suarez-Tembra M, Lopez-Miranda J, Guijarro C. Consensus on lipoprotein(a) of the Spanish Society of Arteriosclerosis. Literature review and recommendations for clinical practice. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:243-266. [PMID: 38599943 DOI: 10.1016/j.arteri.2024.03.002] [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: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
The irruption of lipoprotein(a) (Lp(a)) in the study of cardiovascular risk factors is perhaps, together with the discovery and use of proprotein convertase subtilisin/kexin type 9 (iPCSK9) inhibitor drugs, the greatest novelty in the field for decades. Lp(a) concentration (especially very high levels) has an undeniable association with certain cardiovascular complications, such as atherosclerotic vascular disease (AVD) and aortic stenosis. However, there are several current limitations to both establishing epidemiological associations and specific pharmacological treatment. Firstly, the measurement of Lp(a) is highly dependent on the test used, mainly because of the characteristics of the molecule. Secondly, Lp(a) concentration is more than 80% genetically determined, so that, unlike other cardiovascular risk factors, it cannot be regulated by lifestyle changes. Finally, although there are many promising clinical trials with specific drugs to reduce Lp(a), currently only iPCSK9 (limited for use because of its cost) significantly reduces Lp(a). However, and in line with other scientific societies, the SEA considers that, with the aim of increasing knowledge about the contribution of Lp(a) to cardiovascular risk, it is relevant to produce a document containing the current status of the subject, recommendations for the control of global cardiovascular risk in people with elevated Lp(a) and recommendations on the therapeutic approach to patients with elevated Lp(a).
Collapse
Affiliation(s)
- Javier Delgado-Lista
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España.
| | - Jose M Mostaza
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario La Paz, Madrid, España
| | - Teresa Arrobas-Velilla
- Sociedad Española de Medicina de Laboratorio (SEQCML), Laboratorio de Bioquímica Clínica, Hospital Universitario Virgen Macarena, Sevilla, España
| | - Francisco Blanco-Vaca
- Departamento de Bioquímica Clínica, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB Sant Pau), Barcelona; Departamento de Bioquímica y Biología Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, España
| | - Luis Masana
- Unidad de Medicina Vascular y Metabolismo, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Tarragona, España
| | - Juan Pedro-Botet
- Unidad de Lípidos y Riesgo Vascular, Servicio de Endocrinología y Nutrición, Hospital del Mar, Barcelona; Departamento de Medicina, Universidad Autónoma de Barcelona, Barcelona, España
| | - Pablo Perez-Martinez
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España
| | - Fernando Civeira
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Servicio de Medicina Interna, Hospital Universitario Miguel Servet, IIS Aragón, Universidad de Zaragoza, Zaragoza; CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, España
| | - Jose I Cuende-Melero
- Consulta de Riesgo Vascular, Servicio de Medicina Interna, Complejo Asistencial Universitario de Palencia, Palencia; Departamento de Medicina, Dermatología y Toxicología, Facultad de Medicina, Universidad de Valladolid, Valladolid, España
| | - Jose J Gomez-Barrado
- Unidad de Cuidados Cardiológicos Agudos y Riesgo Cardiovascular, Servicio de Cardiología, Hospital Universitario San Pedro de Alcántara, Cáceres, España
| | - Carlos Lahoz
- Unidad de Lípidos y Arteriosclerosis, Servicio de Medicina Interna, Hospital La Paz-Carlos III, Madrid, España
| | - Xavier Pintó
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell-Universidad de Barcelona-CiberObn, España
| | - Manuel Suarez-Tembra
- Unidad de Lípidos y RCV, Servicio de Medicina Interna, Hospital San Rafael, A Coruña, España
| | - Jose Lopez-Miranda
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España.
| | - Carlos Guijarro
- Unidad de Medicina Interna, Hospital Universitario Fundación Alcorcón, Universidad Rey Juan Carlos, Alcorcón, Madrid, España
| |
Collapse
|
26
|
Sekhar A, Kuttan A, Lange RA. Recent updates on therapeutic targeting of lipoprotein(a) with RNA interference. Curr Opin Cardiol 2024; 39:292-299. [PMID: 38547148 DOI: 10.1097/hco.0000000000001144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
PURPOSE OF REVIEW RNA interference (RNAi)-based therapies that target specific gene products have impacted clinical medicine with 16 FDA approved drugs. RNAi therapy focused on reducing plasma lipoprotein(a) [Lp(a)] levels are under evaluation. RECENT FINDINGS RNAi-based therapies have made significant progress over the past 2 decades and currently consist of antisense oligonucleotides (ASO) and small interfering RNA (siRNA). Chemical modification of the RNA backbone and conjugation of siRNA enables efficient gene silencing in hepatocytes allowing development of effective cholesterol lowering therapies. Multiple lines of evidence suggest a causative role for Lp(a) in atherosclerotic cardiovascular disease, and recent analyses indicate that Lp(a) is more atherogenic than low density lipoprotein- cholesterol (LDL-C). These findings have led to the 'Lp(a) hypothesis' that lowering Lp(a) may significantly improve cardiovascular outcomes. Four RNAi-based drugs have completed early phase clinical trials demonstrating >80% reduction in plasma Lp(a) levels. Phase 3 clinical trials examining clinical outcomes with these agents are currently underway. SUMMARY Currently, four RNAi-based drugs have been shown to be effective in significantly lowering plasma Lp(a) levels. Clinical outcome data from phase 3 trials will evaluate the Lp(a) hypothesis.
Collapse
Affiliation(s)
- Aravind Sekhar
- Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | | | - Richard A Lange
- Texas Tech University Health Sciences Center, El Paso, Texas, USA
| |
Collapse
|
27
|
Singh R, Chandi SK, Sran S, Aulakh SK, Nijjar GS, Singh K, Singh S, Tanvir F, Kaur Y, Sandhu APS. Emerging Therapeutic Strategies in Cardiovascular Diseases. Cureus 2024; 16:e64388. [PMID: 39131016 PMCID: PMC11317025 DOI: 10.7759/cureus.64388] [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: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Cardiovascular diseases (CVDs), including ischemic heart disease and stroke, are the leading cause of mortality worldwide, causing nearly 20 million deaths annually. Traditional therapies, while effective, have not curbed the rising prevalence of CVDs driven by aging populations and lifestyle factors. This review highlights innovative therapeutic strategies that show promise in improving patient outcomes and transforming cardiovascular care. Emerging pharmacological treatments, such as proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors and sodium-glucose co-transporter 2 (SGLT2) inhibitors, introduce novel mechanisms to complement existing therapies, significantly reducing cardiovascular events and mortality. These advancements emphasize the necessity of ongoing clinical trials and research to discover new therapeutic targets. Advanced biological therapies, including gene therapy, stem cell therapy, and RNA-based treatments, offer groundbreaking potential for repairing and regenerating damaged cardiovascular tissues. Despite being in various stages of clinical validation, early results are promising, suggesting these therapies could fundamentally change the CVD treatment landscape. Innovative medical devices and technologies, such as implantable devices, minimally invasive procedures, and wearable technology, are revolutionizing CVD management. These advancements facilitate early diagnosis, continuous monitoring, and effective treatment, driving care out of hospitals and into homes, improving patient outcomes and reducing healthcare costs. Personalized medicine, driven by genetic profiling and biomarker identification, allows for tailored therapies that enhance treatment efficacy and minimize adverse effects. However, the adoption of these emerging therapies faces significant challenges, including regulatory hurdles, cost and accessibility issues, and ethical considerations. Addressing these barriers and fostering interdisciplinary collaboration are crucial for accelerating the development and implementation of innovative treatments. Integrating emerging therapeutic strategies in cardiovascular care holds immense potential to transform CVD management. By prioritizing future research and overcoming existing challenges, a new era of personalized, effective, and accessible cardiovascular care can be achieved.
Collapse
Affiliation(s)
- Rajinderpal Singh
- Internal Medicine, Government Medical College Amritsar, Amritsar, IND
| | | | - Seerat Sran
- Internal Medicine, Sri Guru Ram Das University of Health Sciences and Research, Amritsar, IND
| | - Smriti K Aulakh
- Internal Medicine, Sri Guru Ram Das University of Health Sciences and Research, Amritsar, IND
| | | | | | - Sumerjit Singh
- Medicine, Government Medical College Amritsar, Amritsar, IND
| | - Fnu Tanvir
- Medicine, Government Medical College Amritsar, Amritsar, IND
| | - Yasmeen Kaur
- Medicine, Government Medical College Amritsar, Amritsar, IND
| | - Ajay Pal Singh Sandhu
- Medicine, Sri Guru Ram Das University of Health Sciences and Research, Amritsar, IND
| |
Collapse
|
28
|
Bittner VA, Schwartz GG, Bhatt DL, Chua T, De Silva HA, Diaz R, Goodman SG, Harrington RA, Jukema JW, McGinniss J, Pordy R, Garon G, Scemama M, White HD, Steg PG, Szarek M. Alirocumab and cardiovascular outcomes according to sex and lipoprotein(a) after acute coronary syndrome: a report from the ODYSSEY OUTCOMES study. J Clin Lipidol 2024; 18:e548-e561. [PMID: 38960812 DOI: 10.1016/j.jacl.2024.04.122] [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: 11/23/2023] [Revised: 03/28/2024] [Accepted: 04/06/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND The ODYSSEY OUTCOMES trial (NCT01663402) compared the effects of the proprotein convertase subtilisin/kexin type 9 inhibitor alirocumab with placebo on major adverse cardiovascular events (MACE) in patients with recent acute coronary syndrome (ACS). OBJECTIVE We assessed efficacy and safety of alirocumab versus placebo according to sex and lipoprotein(a) level. METHODS This prespecified analysis compared the effects of alirocumab versus placebo on lipoproteins, MACE (coronary heart disease death, non-fatal myocardial infarction, fatal/non-fatal ischemic stroke, unstable angina requiring hospitalization), death, total cardiovascular events, and adverse events in 4762 women and 14,162 men followed for a median of 2.8 years. In post-hoc analysis, we evaluated total cardiovascular events according to sex, baseline lipoprotein(a), and treatment. RESULTS Women were older, had higher baseline low-density lipoprotein cholesterol (LDL-C) levels (89.6 vs 85.3 mg/dL) and lipoprotein(a) (28.0 vs 19.3 mg/dL) and had more co-morbidities than men. At 4 months, alirocumab lowered LDL-C by 49.4 mg/dL in women and 54.0 mg/dL in men and lipoprotein(a) by 9.7 and 8.1 mg/dL, respectively (both p < 0.0001). Alirocumab reduced MACE, death, and total cardiovascular events similarly in both sexes. In the placebo group, lipoprotein(a) was a risk factor for total cardiovascular events in women and men. In both sexes, reduction of total cardiovascular events was greater at higher baseline lipoprotein(a), but this effect was more evident in women than men (pinteraction=0.08). Medication adherence and adverse event rates were similar in both sexes. CONCLUSIONS Alirocumab improves cardiovascular outcomes after ACS irrespective of sex. Reduction of total cardiovascular events was greater at higher baseline lipoprotein(a).
Collapse
Affiliation(s)
- Vera A Bittner
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, UK (Dr Bittner).
| | - Gregory G Schwartz
- Division of Cardiology, University of Colorado School of Medicine, Aurora, CO (Dr Schwartz)
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Health System, New York, NY, USA (Dr Bhatt)
| | | | - H Asita De Silva
- Clinical Trials Unit, Department of Pharmacology, Faculty of Medicine, University of Kelaniya, Sri Lanka (Dr De Silva)
| | - Rafael Diaz
- Estudios Cardiológicos Latino América, Instituto Cardiovascular de Rosario, Rosario, Argentina (Dr Diaz)
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada (Dr Goodman); St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada (Dr Goodman)
| | - Robert A Harrington
- Stanford Center for Clinical Research, Department of Medicine, Stanford University, Stanford, CA (Dr Harrington)
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands (Dr Jukema); Netherlands Heart Institute, Utrecht, the Netherlands (Dr Jukema)
| | - Jennifer McGinniss
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, USA (Dr McGinniss, Pordy)
| | - Robert Pordy
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, USA (Dr McGinniss, Pordy)
| | | | - Michel Scemama
- Sanofi Research and Development, Paris, France (Dr Scemama)
| | - Harvey D White
- Green Lane Cardiovascular Research Unit, Te Whatu Ora - Health New Zealand, Te Toka Tumai, and University of Auckland, Auckland, New Zealand (Dr White)
| | - Ph Gabriel Steg
- Université Paris-Cité, Institut Universitaire de France, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, FACT (French Alliance for Cardiovascular Trials), and INSERM U1148, Paris, France (Dr Steg)
| | - Michael Szarek
- CPC Clinical Research and Division of Cardiology, University of Colorado School of Medicine, Aurora, CO (Dr Szarek); State University of New York, Downstate Health Sciences University, Brooklyn, NY (Dr Szarek)
| |
Collapse
|
29
|
Yang C, Zhu CG, Sui YG, Guo YL, Wu NQ, Dong Q, Xu RX, Qian J, Li JJ. Synergetic impact of lipoprotein(a) and fibrinogen on stroke in coronary artery disease patients. Eur J Clin Invest 2024; 54:e14179. [PMID: 38363025 DOI: 10.1111/eci.14179] [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: 11/16/2023] [Revised: 01/21/2024] [Accepted: 02/03/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Emerging data suggested that lipoprotein(a) [Lp(a)] is an independent risk factor for atherosclerotic cardiovascular disease. Previous studies indicated fibrinogen (Fib) had synergetic effect on Lp(a)-induced events. However, combined impact of Fib and Lp(a) on ischemic stroke has not been elucidated. METHODS In this prospective study, we consecutively enrolled 8263 patients with stable coronary artery diseases (CAD) from 2011 to 2017. Patients were categorized into three groups according to tertiles of Lp(a) levels [Lp(a)-low, Lp(a)-medium, and Lp(a)-high] and further divided into nine groups by Lp(a) and Fib levels. All subjects were followed up for the occurrence of ischemic stroke. RESULTS During a median follow-up of 37.7 months, 157 (1.9%) ischemic strokes occurred. Stroke incidence increased by Lp(a) (1.1 vs. 2.1 vs. 2.5%, Cochran-Armitage p < .001) and Fib (1.1 vs. 2.0 vs. 2.6%, Cochran-Armitage p < .001) categories. When further classified into nine groups by Lp(a) and Fib levels, the incidence of ischemic stroke in group 9 [Lp(a)-high and Fib-high] was significantly higher than that in group 1 [Lp(a)-low and Fib-low] (3.1 vs. 6%, p < .001). The group 9 was associated with a highest risk for ischemic stroke (adjusted HR 4.907, 95% CI: 2.154-11.18, p < .001), compared with individuals in the Lp(a)-high (adjusted HR 2.290, 95% CI: 1.483-3.537, p < .001) or Fib-high (adjusted HR 1.184, 95% CI: 1.399-3.410, p = .001). Furthermore, combining Lp(a) with Fib increased C-statistics by .045 (p = .004). CONCLUSIONS Current study first demonstrated that elevated Lp(a) combining with Fib evaluation enhanced the risk of ischemic stroke in patients with CAD beyond Lp(a) or Fib alone.
Collapse
Affiliation(s)
- Cheng Yang
- Cardiometabolic Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng-Gang Zhu
- Cardiometabolic Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong-Gang Sui
- Cardiometabolic Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan-Lin Guo
- Cardiometabolic Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na-Qiong Wu
- Cardiometabolic Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Dong
- Cardiometabolic Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui-Xia Xu
- Cardiometabolic Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Qian
- Cardiometabolic Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Jun Li
- Cardiometabolic Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Kaur G, Abdelrahman K, Berman AN, Biery DW, Shiyovich A, Huck D, Garshick M, Blankstein R, Weber B. Lipoprotein(a): Emerging insights and therapeutics. Am J Prev Cardiol 2024; 18:100641. [PMID: 38646022 PMCID: PMC11033089 DOI: 10.1016/j.ajpc.2024.100641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/08/2024] [Accepted: 02/24/2024] [Indexed: 04/23/2024] Open
Abstract
The strong association between lipoprotein (a) [Lp(a)] and atherosclerotic cardiovascular disease has led to considerations of Lp(a) being a potential target for mitigating residual cardiovascular risk. While approximately 20 % of the population has an Lp(a) level greater than 50 mg/dL, there are no currently available pharmacological lipid-lowering therapies that have demonstrated substantial reduction in Lp(a). Novel therapies to lower Lp(a) include antisense oligonucleotides and small-interfering ribonucleic acid molecules and have shown promising results in phase 2 trials. Phase 3 trials are currently underway and will test the causal relationship between Lp(a) and ASCVD and whether lowering Lp(a) reduces cardiovascular outcomes. In this review, we summarize emerging insights related to Lp(a)'s role as a risk-enhancing factor for ASCVD, association with calcific aortic stenosis, effects of existing therapies on Lp(a) levels, and variations amongst patient populations. The evolving therapeutic landscape of emerging therapeutics is further discussed.
Collapse
Affiliation(s)
- Gurleen Kaur
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Adam N. Berman
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - David W. Biery
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Albert Einstein College of Medicine, New York, NY, USA
| | - Arthur Shiyovich
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel Huck
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Ron Blankstein
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Brittany Weber
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
Wang Z, Tang J, Shi Q, Fang L, Liu N, Zhang J. Synergistic effect of lipoprotein(a) and high-sensitivity C-reactive protein on the risk of all-cause and cardiovascular death in patients with acute myocardial infarction: a large prospective cohort study. Front Endocrinol (Lausanne) 2024; 15:1392859. [PMID: 38812817 PMCID: PMC11133541 DOI: 10.3389/fendo.2024.1392859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
Objective Although lipoprotein(a) [Lp(a)] and high-sensitivity C-reactive protein (Hs-CRP) are closely associated with the mortality of acute myocardial infarction (AMI), their synergistic effect on the risk of death remains unknown. Therefore, this study aimed to explore the combined effect of Lp(a) and Hs-CRP on the incidence of all-cause and cardiovascular death in AMI patients. Methods A comprehensive cohort study enrolled 912 AMI patients, categorizing them into four groups based on Lp(a) and Hs-CRP levels: Group 1 [Lp(a) < 30 mg/dL & Hs-CRP < 2 mg/L], Group 2 [Lp(a) < 30 mg/dL & Hs-CRP ≥ 2 mg/L], Group 3 [Lp(a) ≥ 30 mg/dL & Hs-CRP < 2 mg/L], and Group 4 [Lp(a) ≥ 30 mg/dL & Hs-CRP ≥ 2 mg/L]. Cox regression analysis, Kaplan-Meier survival analysis and sensitivity analysis were employed to determine the combined effects of Lp(a) and Hs-CRP on the risk of all-cause and cardiovascular death. Results Over a median observation period of 38.98 months, 217 patients passed away, with 137 deaths attributed to cardiovascular causes. The multivariate Cox regression analysis revealed that in the comprehensively adjusted Model 3, only Lp(a) and the combination of Lp(a) and Hs-CRP exhibited a strong association with cardiovascular death risk. Specifically, for Lp(a) levels ≥ 30 mg/dL compared to < 30 mg/dL, the hazard ratio (HR) was 2.434 with a 95% confidence interval (CI) of 1.653-3.583 (P < 0.001); for log10(Lp(a)), the HR was 2.630 with a 95% CI of 1.530-4.523 (P < 0.001); for Group 4 versus Group 1, the HR was 2.346 with a 95% CI of 1.054-5.220 (P = 0.037); and for Group 4 versus Groups 1 + 2 + 3, the HR was 1.878 with a 95% CI of 1.284-2.748 (P = 0.001). Sensitivity analysis indicated that the synergy between Lp(a) and Hs-CRP continued to be independently associated with the risk of cardiovascular death. For Group 3 versus Group 1, the HR was 3.353 with a 95% CI of 1.133-9.917 (P = 0.029); for Group 4 versus Group 1, the HR was 3.710 with a 95% CI of 1.466-9.392 (P = 0.006); and for Group 4 versus Groups 1 + 2 + 3, the HR was 2.433 with a 95% CI of 1.620-3.656 (P < 0.001). Conclusions Compared to elevated levels of either Lp(a) or Hs-CRP alone, the concurrent high levels of both significantly increased the risk of cardiovascular death in patients with AMI, underscoring the importance of considering their combined effects in the prognostic management of AMI patients.
Collapse
Affiliation(s)
- Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Qian Shi
- Neonatal Intensive Care Unit, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijuan Fang
- Department of Cardiology, The First Hospital of Hohhot, Hohhot, China
| | - Naifeng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| |
Collapse
|
34
|
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
|
35
|
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
|
36
|
Koschinsky ML, Bajaj A, Boffa MB, Dixon DL, Ferdinand KC, Gidding SS, Gill EA, Jacobson TA, Michos ED, Safarova MS, Soffer DE, Taub PR, Wilkinson MJ, Wilson DP, Ballantyne CM. A focused update to the 2019 NLA scientific statement on use of lipoprotein(a) in clinical practice. J Clin Lipidol 2024; 18:e308-e319. [PMID: 38565461 DOI: 10.1016/j.jacl.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
Since the 2019 National Lipid Association (NLA) Scientific Statement on Use of Lipoprotein(a) in Clinical Practice was issued, accumulating epidemiological data have clarified the relationship between lipoprotein(a) [Lp(a)] level and cardiovascular disease risk and risk reduction. Therefore, the NLA developed this focused update to guide clinicians in applying this emerging evidence in clinical practice. We now have sufficient evidence to support the recommendation to measure Lp(a) levels at least once in every adult for risk stratification. Individuals with Lp(a) levels <75 nmol/L (30 mg/dL) are considered low risk, individuals with Lp(a) levels ≥125 nmol/L (50 mg/dL) are considered high risk, and individuals with Lp(a) levels between 75 and 125 nmol/L (30-50 mg/dL) are at intermediate risk. Cascade screening of first-degree relatives of patients with elevated Lp(a) can identify additional individuals at risk who require intervention. Patients with elevated Lp(a) should receive early, more-intensive risk factor management, including lifestyle modification and lipid-lowering drug therapy in high-risk individuals, primarily to reduce low-density lipoprotein cholesterol (LDL-C) levels. The U.S. Food and Drug Administration approved an indication for lipoprotein apheresis (which reduces both Lp(a) and LDL-C) in high-risk patients with familial hypercholesterolemia and documented coronary or peripheral artery disease whose Lp(a) level remains ≥60 mg/dL [∼150 nmol/L)] and LDL-C ≥ 100 mg/dL on maximally tolerated lipid-lowering therapy. Although Lp(a) is an established independent causal risk factor for cardiovascular disease, and despite the high prevalence of Lp(a) elevation (∼1 of 5 individuals), measurement rates are low, warranting improved screening strategies for cardiovascular disease prevention.
Collapse
Affiliation(s)
- Marlys L Koschinsky
- Department of Physiology & Pharmacology and Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada (Drs Koschinsky, Boffa)
| | - Archna Bajaj
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA (Drs Bajaj, Soffer)
| | - Michael B Boffa
- Department of Physiology & Pharmacology and Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada (Drs Koschinsky, Boffa)
| | - Dave L Dixon
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, VA, USA (Dr Dixon)
| | - Keith C Ferdinand
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA (Dr. Ferdinand)
| | - Samuel S Gidding
- Department of Genomic Health, Geisinger. Danville, PA, USA (Dr Gidding)
| | - Edward A Gill
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA (Dr Gill)
| | - Terry A Jacobson
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA (Dr Jacobson)
| | - Erin D Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA (Dr Michos)
| | - Maya S Safarova
- Division of Cardiovascular Medicine, Department of Internal Medicine, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, USA (Dr Safarova)
| | - Daniel E Soffer
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA (Drs Bajaj, Soffer)
| | - Pam R Taub
- Department of Medicine, University of California San Diego, La Jolla, CA, USA (Drs Taub, Wilkinson)
| | - Michael J Wilkinson
- Department of Medicine, University of California San Diego, La Jolla, CA, USA (Drs Taub, Wilkinson)
| | - Don P Wilson
- Department of Pediatric Endocrinology and Diabetes, Cook Children's Medical Center, Fort Worth, TX, USA (Dr Wilson)
| | - Christie M Ballantyne
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA (Dr Ballantyne).
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
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
|
40
|
Nicholls SJ, Nelson AJ. New targets and mechanisms of action for lipid-lowering and anti-inflammatory therapies in atherosclerosis: where does the field stand? Expert Opin Ther Targets 2024; 28:375-384. [PMID: 38815057 DOI: 10.1080/14728222.2024.2362644] [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: 12/20/2023] [Accepted: 05/29/2024] [Indexed: 06/01/2024]
Abstract
INTRODUCTION Atherosclerotic cardiovascular disease remains a leading cause of morbidity and mortality worldwide, despite widespread use of statins. There is a need to develop additional therapeutic strategies that will complement statins to achieve more effective reductions in cardiovascular risk. AREAS COVERED This review provides a comprehensive summary of current areas of therapeutic development targeting both lipid and inflammatory factors implicated in the pathogenesis of atherosclerosis. In addition to develop of novel approaches that will produce more effective lowering of low-density lipoprotein cholesterol, clinical trials are currently evaluating the potential to target other atherogenic lipid parameters such as triglyceride-rich lipoproteins and Lp(a), in addition to promoting the biological properties of high-density lipoproteins. Targeting inflammation within the vascular wall has emerged as a new frontier in cardiovascular prevention, with early evidence that use of anti-inflammatory agents have the potential to reduce cardiovascular risk. EXPERT OPINION Clinical practice has an increasing array of therapeutic tools to achieve more effective lowering of low-density lipoprotein cholesterol for high-risk patients. In addition, clinical trials have the potential to deliver a range of additional agents to the clinic, that target alternative lipid and inflammatory mediators. This will permit the potential to personalize cardiovascular prevention.
Collapse
Affiliation(s)
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, Melbourne, Australia
| |
Collapse
|
41
|
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
|
42
|
Blaha MJ, Bhatia HS. Lipoprotein(a), Residual Cardiovascular Risk, and the Search for Targeted Therapy. J Am Coll Cardiol 2024; 83:1540-1542. [PMID: 38537912 PMCID: PMC11216951 DOI: 10.1016/j.jacc.2024.03.370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/19/2024]
Affiliation(s)
- Michael J Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Atherosclerotic Cardiovascular Disease, Baltimore, Maryland, USA.
| | - Harpreet S Bhatia
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
43
|
Wulff AB, Nordestgaard BG, Langsted A. Novel Therapies for Lipoprotein(a): Update in Cardiovascular Risk Estimation and Treatment. Curr Atheroscler Rep 2024; 26:111-118. [PMID: 38311667 DOI: 10.1007/s11883-024-01192-9] [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] [Accepted: 01/19/2024] [Indexed: 02/06/2024]
Abstract
PURPOSE OF REVIEW Lipoprotein(a) is an important causal risk factor for cardiovascular disease but currently no available medication effectively reduces lipoprotein(a). This review discusses recent findings regarding lipoprotein(a) as a causal risk factor and therapeutic target in cardiovascular disease, it reviews current clinical recommendations, and summarizes new lipoprotein(a) lowering drugs. RECENT FINDINGS Epidemiological and genetic studies have established lipoprotein(a) as a causal risk factor for cardiovascular disease and mortality. Guidelines worldwide now recommend lipoprotein(a) to be measured once in a lifetime, to offer patients with high lipoprotein(a) lifestyle advise and initiate other cardiovascular medications. Clinical trials including antisense oligonucleotides, small interfering RNAs, and an oral lipoprotein(a) inhibitor have shown great effect on lowering lipoprotein(a) with reductions up to 106%, without any major adverse effects. Recent clinical phase 1 and 2 trials show encouraging results and ongoing phase 3 trials will hopefully result in the introduction of specific lipoprotein(a) lowering drugs to lower the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Anders Berg Wulff
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Frederiksberg, Denmark
| | - Anne Langsted
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen Ø, Denmark.
| |
Collapse
|
44
|
Ranjan A, Agarwal R, Mudgal SK, Bhattacharya S, Kumar B. Young hearts at risk: Unveiling novel factors in myocardial infarction susceptibility and prevention. J Family Med Prim Care 2024; 13:1200-1205. [PMID: 38827682 PMCID: PMC11141995 DOI: 10.4103/jfmpc.jfmpc_1639_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/14/2023] [Accepted: 01/05/2024] [Indexed: 06/04/2024] Open
Abstract
The increasing incidence of acute myocardial infarction (AMI) among the young population represents a significant and emerging health concern, contributing substantially to both mortality and morbidity. Unlike myocardial infarctions occurring in older individuals, traditional risk factors such as diabetes and hypertension exhibit a weaker association in the younger demographic. Consequently, there is a pressing need for a deeper understanding of novel risk factors that contribute to AMI in young patients. In this review, we explore distinct risk factor profiles associated with young-onset AMI in comparison to older patients. Special attention is given to novel risk factors, examining their susceptibility factors and exploring preventive measures. The comprehensive risk profile of extremely young South Asians who develop early coronary arterial disease is not yet fully understood. There are many novel evolving risk factors associated with young AMI which need intervention to reduce morbidity and mortality. It has been seen that established inflammatory markers like lipoprotein (a), dyslipidaemia, long COVID, and new emerging risk factors like air pollution (micro- and nanoplastics), periodontitis, acute stress, energy drinks, misuse of recreational drugs may increase risk and influence treatment, and outcomes of AMI in this young population. Screening of emerging novel risk markers and their optimization is important in preventing young patients with AMI. The role of conventional risk factors should not be overlooked and should be treated aggressively. Sex and geographic-specific base approaches are required to reduce risk factors and prevent AMI in young. More prospective studies are needed to evaluate the increasing incidence of young AMI and its associated novel risk factors.
Collapse
Affiliation(s)
- Ashis Ranjan
- Department of Cardiology, AIIMS Deoghar, Jharkhand, India
| | - Rajat Agarwal
- Department of Cardiothoracic Surgery, AIIMS Deoghar, Jharkhand, India
| | | | - Sudip Bhattacharya
- Department of Community and Family Medicine, AIIMS Deoghar, Jharkhand, India
| | - Barun Kumar
- Department of Cardiology, AIIMS Rishikesh, Uttarakhand, India
| |
Collapse
|
45
|
Laffin LJ, Nissen SE. Lp(a) - an overlooked risk factor. Trends Cardiovasc Med 2024; 34:193-199. [PMID: 36681362 DOI: 10.1016/j.tcm.2023.01.003] [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: 10/05/2022] [Revised: 12/09/2022] [Accepted: 01/14/2023] [Indexed: 01/20/2023]
Abstract
Lipoprotein(a) (Lp(a)) is an increasingly discussed and studied risk factor for atherosclerotic cardiovascular disease and aortic valve stenosis. Many genetic and epidemiological studies support the important causal role that Lp(a) plays in the incidence of cardiovascular disease. Although dependent upon the threshold and unit of measurement of Lp(a), most estimates suggest between 20 and 30% of the world's population have elevated serum levels of Lp(a). Lp(a) levels are predominantly mediated by genetics and are not significantly modified by lifestyle interventions. Efforts are ongoing to develop effective pharmacotherapies to lower Lp(a) and to determine if lowering Lp(a) with these medications ultimately decreases the incidence of adverse cardiovascular events. In this review, the genetics and pathophysiological properties of Lp(a) will be discussed as well as the epidemiological data demonstrating its impact on the incidence of cardiovascular disease. Recommendations for screening and how to currently approach patients with elevated Lp(a) are also noted. Finally, the spectrum of pharmacotherapies under development for Lp(a) lowering is detailed.
Collapse
|
46
|
Hilleman DE, Vacek JL, Backes JM. Elevated Lp(a): Guidance for Identifying and Managing Patients. South Med J 2024; 117:208-213. [PMID: 38569611 DOI: 10.14423/smj.0000000000001675] [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: 04/05/2024]
Abstract
Lipoprotein(a) (Lp(a)) is a unique low-density lipoprotein-like lipoprotein that is considered an independent and causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and calcific aortic valve stenosis. The Lp(a) molecule also contains apolipoprotein A and apolipoprotein B, which collectively promote atherosclerosis, thrombosis, and inflammation. Lp(a) is highly genetic and minimally responsive to nonpharmacological measures. Lp(a) serum levels ≥125 nmol/L are associated with increased ASCVD risk, but this threshold has not been accepted universally. Elevated Lp(a) is the most common genetic dyslipidemia affecting approximately 20% of the general population. Certain currently available lipid-lowering drugs, including the proprotein convertase subtilisin/kexin type 9 therapies, produce moderate reductions in Lp(a); however, none are indicated for the treatment of elevated Lp(a). There are currently four investigational RNA-based therapeutic agents that reduce Lp(a) by 70% to 100%. Two of these agents are being evaluated for ASCVD risk reduction in adequately powered outcomes trials, with results expected in 2 to 3 years. Until such therapies become available and demonstrate favorable clinical outcomes, strategies for elevated Lp(a) primarily involve early and intensive ASCVD risk factor management.
Collapse
Affiliation(s)
- Daniel E Hilleman
- From the Creighton University School of Pharmacy and Health Professions, Omaha, Nebraska
| | - James L Vacek
- Department of Cardiovascular Medicine, University of Kansas Health System, Kansas City
| | - James M Backes
- the Atherosclerosis and LDL-Apheresis Center, University of Kansas Medical Center, KU School of Pharmacy, Lawrence
| |
Collapse
|
47
|
Wilkinson MJ, Bajaj A, Brousseau ME, Taub PR. Harnessing RNA Interference for Cholesterol Lowering: The Bench-to-Bedside Story of Inclisiran. J Am Heart Assoc 2024; 13:e032031. [PMID: 38456415 PMCID: PMC11010004 DOI: 10.1161/jaha.123.032031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/06/2023] [Indexed: 03/09/2024]
Abstract
Lowering low-density lipoprotein cholesterol (LDL-C) is a cornerstone of reducing risk for atherosclerotic cardiovascular disease. Despite the approval of nonstatin therapies for LDL-C lowering over the past 2 decades, these medications are underused, and most patients are still not at guideline-recommended LDL-C goals. Barriers include poor adherence, clinical inertia, concern for side effects, cost, and complex prior authorization processes. With atherosclerotic cardiovascular disease-related mortality increasing globally, there remains a need for additional therapeutic options for lowering LDL-C as part of an atherosclerotic cardiovascular disease prevention strategy. Following the identification of PCSK9 (proprotein convertase subtilisin/kexin type 9) as a promising therapeutic target, inclisiran was developed using the natural process of RNA interference for robust, sustained prevention of hepatic PCSK9 synthesis. Twice-yearly maintenance subcutaneous inclisiran (following initial loading doses at Day 1 and Day 90) reduces circulating LDL-C levels by ≈50% versus placebo when added to maximally tolerated statins. Long-term safety and tolerability of inclisiran have been assessed, with studies underway to evaluate the effects of inclisiran on cardiovascular outcomes and to provide additional safety and effectiveness data. In 2021, <20 years after the discovery of PCSK9, inclisiran became the first RNA interference therapeutic approved in the United States for LDL-C lowering in patients with established atherosclerotic cardiovascular disease or familial hypercholesterolemia and has since been approved for use in patients with primary hyperlipidemia. This article reviews the journey of inclisiran from bench to bedside, including early development, the clinical trial program, key characteristics of inclisiran, and practical points for its use in the clinic.
Collapse
Affiliation(s)
- Michael J. Wilkinson
- Division of Cardiovascular Medicine, Department of MedicineCardiovascular Institute, University of California San DiegoSan DiegoCAUSA
| | - Archna Bajaj
- Department of MedicinePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | - Margaret E. Brousseau
- Cardiovascular and Metabolism Disease Area, Novartis Institutes for BioMedical ResearchCambridgeMAUSA
| | - Pam R. Taub
- Division of Cardiovascular Medicine, Department of MedicineCardiovascular Institute, University of California San DiegoSan DiegoCAUSA
| |
Collapse
|
48
|
Yoon D, Choi B, Kim JE, Kim EY, Chung SH, Min HJ, Sung Y, Chang EJ, Song JK. Autotaxin inhibition attenuates the aortic valve calcification by suppressing inflammation-driven fibro-calcific remodeling of valvular interstitial cells. BMC Med 2024; 22:122. [PMID: 38486246 PMCID: PMC10941471 DOI: 10.1186/s12916-024-03342-x] [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: 06/25/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Patients with fibro-calcific aortic valve disease (FCAVD) have lipid depositions in their aortic valve that engender a proinflammatory impetus toward fibrosis and calcification and ultimately valve leaflet stenosis. Although the lipoprotein(a)-autotaxin (ATX)-lysophosphatidic acid axis has been suggested as a potential therapeutic target to prevent the development of FCAVD, supportive evidence using ATX inhibitors is lacking. We here evaluated the therapeutic potency of an ATX inhibitor to attenuate valvular calcification in the FCAVD animal models. METHODS ATX level and activity in healthy participants and patients with FCAVD were analyzed using a bioinformatics approach using the Gene Expression Omnibus datasets, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, and western blotting. To evaluate the efficacy of ATX inhibitor, interleukin-1 receptor antagonist-deficient (Il1rn-/-) mice and cholesterol-enriched diet-induced rabbits were used as the FCAVD models, and primary human valvular interstitial cells (VICs) from patients with calcification were employed. RESULTS The global gene expression profiles of the aortic valve tissue of patients with severe FCAVD demonstrated that ATX gene expression was significantly upregulated and correlated with lipid retention (r = 0.96) or fibro-calcific remodeling-related genes (r = 0.77) in comparison to age-matched non-FCAVD controls. Orally available ATX inhibitor, BBT-877, markedly ameliorated the osteogenic differentiation and further mineralization of primary human VICs in vitro. Additionally, ATX inhibition significantly attenuated fibrosis-related factors' production, with a detectable reduction of osteogenesis-related factors, in human VICs. Mechanistically, ATX inhibitor prohibited fibrotic changes in human VICs via both canonical and non-canonical TGF-β signaling, and subsequent induction of CTGF, a key factor in tissue fibrosis. In the in vivo FCAVD model system, ATX inhibitor exposure markedly reduced calcific lesion formation in interleukin-1 receptor antagonist-deficient mice (Il1rn-/-, P = 0.0210). This inhibition ameliorated the rate of change in the aortic valve area (P = 0.0287) and mean pressure gradient (P = 0.0249) in the FCAVD rabbit model. Moreover, transaortic maximal velocity (Vmax) was diminished with ATX inhibitor administration (mean Vmax = 1.082) compared to vehicle control (mean Vmax = 1.508, P = 0.0221). Importantly, ATX inhibitor administration suppressed the effects of a high-cholesterol diet and vitamin D2-driven fibrosis, in association with a reduction in macrophage infiltration and calcific deposition, in the aortic valves of this rabbit model. CONCLUSIONS ATX inhibition attenuates the development of FCAVD while protecting against fibrosis and calcification in VICs, suggesting the potential of using ATX inhibitors to treat FCAVD.
Collapse
Affiliation(s)
- Dohee Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Bongkun Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Ji-Eun Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Young Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Soo-Hyun Chung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hyo-Jin Min
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yoolim Sung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Jae-Kwan Song
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
49
|
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
|
50
|
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
|