1
|
Sherrill C, Ehrhardt-Humbert L, Salem D. The Role of Lipoprotein (a) in the Progression of Aortic Stenosis. Am J Med 2025:S0002-9343(25)00138-X. [PMID: 40049573 DOI: 10.1016/j.amjmed.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025]
Abstract
Calcific aortic stenosis is a complex process that involves several factors, including genetics, lipid infiltration, inflammation, and hemodynamic stress. This process begins at an early age and progresses throughout one's lifetime. Lipoprotein (a), or Lp(a), has been shown to drive progression of aortic stenosis in several studies. Additionally, analyses have demonstrated that a higher serum level of Lp(a) is associated with earlier onset of aortic stenosis and faster progression of mean pressure across the aortic valve. The current cornerstone medications to lower low-density lipoprotein do not have a meaningful impact on Lp(a) levels. In this article, we will review current therapeutics that are in development to lower serum Lp(a), including several that involve ribonucleic acid modification. Although these novel therapeutics are promising, it is to be determined whether lowering serum Lp(a) correlates to reduced calcific aortic stenosis and improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Clay Sherrill
- Department of Medicine, Tufts Medical Center, Boston, Mass
| | | | - Deeb Salem
- Center for Cardiovascular Disease, Department of Medicine, Tufts Medical Center, Boston, Mass
| |
Collapse
|
2
|
Greco A, Finocchiaro S, Spagnolo M, Faro DC, Mauro MS, Raffo C, Sangiorgio G, Imbesi A, Laudani C, Mazzone PM, Ammirabile N, Giacoppo D, Landolina D, Capodanno D. Lipoprotein(a) as a Pharmacological Target: Premises, Promises, and Prospects. Circulation 2025; 151:400-415. [PMID: 39928714 DOI: 10.1161/circulationaha.124.069210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 10/29/2024] [Indexed: 02/12/2025]
Abstract
Atherosclerotic cardiovascular disease is a major health concern worldwide and requires effective preventive measures. Lp(a) (lipoprotein [a]) has recently garnered attention as an independent risk factor for astherosclerotic cardiovascular disease, with proinflammatory and prothrombotic mechanisms contributing to its atherogenicity. On an equimolar basis, Lp(a) is ~5 to 6 times more atherogenic than particles that have been widely associated with adverse cardiovascular outcomes, such as LDL (low-density lipoprotein). Lp(a) can enter the vessel wall, leading to the accumulation of oxidized phospholipids in the arterial intima, which are crucial for initiating plaque inflammation and triggering vascular disease progression. In addition, Lp(a) may cause atherothrombosis through interactions between apoA (apolipoprotein A) and the platelet PAR-1 (protease-activated receptor 1) receptor, as well as competitive inhibition of plasminogen. Because Lp(a) is mostly determined on genetic bases, a 1-time assessment in a lifetime can suffice to identify patients with elevated levels. Mendelian randomization studies and post hoc analyses of randomized trials of LDL cholesterol-lowering drugs showed a causal link between Lp(a) concentrations and cardiovascular outcomes, with therapeutic reduction of Lp(a) expected to contribute to estimated cardiovascular risk mitigation. Many Lp(a)-lowering drugs, including monoclonal antibodies, small interfering ribonucleic acids, antisense oligonucleotides, small molecules, and gene editing compounds, are at different stages of clinical investigation and show promise for clinical use. In particular, increased Lp(a) testing and treatment are expected to have a substantial impact at the population level, enabling the identification of high-risk individuals and the subsequent prevention of a large number of cardiovascular events. Ongoing phase 3 trials will further elucidate the cardiovascular benefits of Lp(a) reduction over the long term, offering potential avenues for targeted interventions and improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Antonio Greco
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Simone Finocchiaro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Marco Spagnolo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Denise Cristiana Faro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Maria Sara Mauro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Carmelo Raffo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Giuseppe Sangiorgio
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Antonino Imbesi
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Claudio Laudani
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Placido Maria Mazzone
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Nicola Ammirabile
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Daniele Giacoppo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Davide Landolina
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| |
Collapse
|
3
|
Krzesińska A, Marlęga-Linert J, Chyła-Danił G, Marcinkowska M, Rogowska P, Stumska K, Fijałkowski M, Gruchała M, Jankowski M, Mickiewicz A, Kuchta A. Reduced Oxidative Susceptibility of Lp(a) and LDL Fractions as a Pleiotropic Effect of Lipoprotein Apheresis in Patients with Elevated Lp(a) and ASCVDs. Int J Mol Sci 2024; 25:13597. [PMID: 39769362 PMCID: PMC11676408 DOI: 10.3390/ijms252413597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Oxidative modifications of lipoproteins play a crucial role in the initiation of atherosclerotic cardiovascular diseases (ASCVDs). Nowadays, the one effective strategy for the treatment of patients with hyperlipoproteinemia(a) is lipoprotein apheresis (LA), which has a pleiotropic effect on reducing the risk of ASCVDs. The significance of oxidative susceptibility of the LDL fraction in ASCVDs has been extensively studied. Whether LA alters the susceptibility of lipoprotein(a) to oxidative modifications remains an unresolved issue. In this study, we isolated lipoprotein fractions by ultracentrifugation in patients with hyperlipoproteinemia(a) undergoing apheresis (LA group) at three time points and patients who were qualified for LA but did not consent to the procedure (non-LA group). We performed copper-mediated oxidation of Lp(a) and LDL fractions and determined autotaxin activity. After apheresis, we observed a lower susceptibility to oxidation of the Lp(a) and LDL fractions as expressed by the extended value of oxidation lag time, decreased slope of the oxidation curve, and decreased final concentration of conjugated dienes. No significant differences were found between these parameters before and 7 days after LA. Additionally, both patients undergoing and not undergoing LA had a significant correlation between autotaxin activity and all parameters characterizing susceptibility to oxidation in the Lp(a) fraction. Our results demonstrate that the pleiotropic effect of apheresis may be related to the reduced oxidative susceptibility of Lp(a) and LDL particles, which may influence the reduction in ASCVD risk in patients undergoing apheresis. The results of the rebound effect 7 days after LA will contribute to a better definition of apheresis frequency guidelines.
Collapse
Affiliation(s)
- Aleksandra Krzesińska
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (A.K.); (G.C.-D.); (P.R.); (K.S.); (M.J.)
| | - Joanna Marlęga-Linert
- 1st Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (J.M.-L.); (M.M.); (M.F.); (M.G.); (A.M.)
| | - Gabriela Chyła-Danił
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (A.K.); (G.C.-D.); (P.R.); (K.S.); (M.J.)
| | - Marta Marcinkowska
- 1st Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (J.M.-L.); (M.M.); (M.F.); (M.G.); (A.M.)
| | - Paulina Rogowska
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (A.K.); (G.C.-D.); (P.R.); (K.S.); (M.J.)
| | - Katarzyna Stumska
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (A.K.); (G.C.-D.); (P.R.); (K.S.); (M.J.)
| | - Marcin Fijałkowski
- 1st Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (J.M.-L.); (M.M.); (M.F.); (M.G.); (A.M.)
| | - Marcin Gruchała
- 1st Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (J.M.-L.); (M.M.); (M.F.); (M.G.); (A.M.)
| | - Maciej Jankowski
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (A.K.); (G.C.-D.); (P.R.); (K.S.); (M.J.)
| | - Agnieszka Mickiewicz
- 1st Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (J.M.-L.); (M.M.); (M.F.); (M.G.); (A.M.)
| | - Agnieszka Kuchta
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (A.K.); (G.C.-D.); (P.R.); (K.S.); (M.J.)
| |
Collapse
|
4
|
Gianos E, Duell PB, Toth PP, Moriarty PM, Thompson GR, Brinton EA, Hudgins LC, Nametka M, Byrne KH, Raghuveer G, Nedungadi P, Sperling LS. Lipoprotein Apheresis: Utility, Outcomes, and Implementation in Clinical Practice: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2024; 44:e304-e321. [PMID: 39370995 DOI: 10.1161/atv.0000000000000177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Despite the availability of multiple classes of lipoprotein-lowering medications, some high-risk patients have persistent hypercholesterolemia and may require nonpharmacologic therapy. Lipoprotein apheresis (LA) is a valuable but underused adjunctive therapeutic option for low-density lipoprotein cholesterol and lipoprotein(a) lowering, particularly in children and adults with familial hypercholesterolemia. In addition to lipid lowering, LA reduces serum levels of proinflammatory and prothrombotic factors, reduces blood viscosity, increases microvascular myocardial perfusion, and may provide beneficial effects on endothelial function. Multiple observational studies demonstrate strong evidence for improved cardiovascular outcomes with LA; however, use in the United States is limited to a fraction of its Food and Drug Administration-approved indications. In addition, there are limited data regarding LA benefit for refractory focal segmental glomerulosclerosis. In this scientific statement, we review the history of LA, mechanisms of action, cardiovascular and renal outcomes data, indications, and options for treatment.
Collapse
|
5
|
Khan MI, Zahir RS, Dominguez AC, Romeo FJ. Role of Lipoprotein (A) in aortic valve stenosis: Novel disease mechanisms and emerging pharmacotherapeutic approaches. IJC HEART & VASCULATURE 2024; 55:101543. [PMID: 39555492 PMCID: PMC11564994 DOI: 10.1016/j.ijcha.2024.101543] [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: 03/24/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024]
Abstract
Lipoprotein(a) (Lp(a)) has garnered increasing attention as a significant contributor to the pathogenesis of aortic stenosis (AS), prompting a focused investigation into innovative pharmacological strategies to target this lipoprotein and its associated risks. Despite its recognized role in AS progression, Lp(a) often remains overlooked in clinical assessments, mirroring the broader challenges observed in holistic disease management. This review delves into the mechanistic intricacies of Lp(a) involvement in AS pathophysiology and its potential as a therapeutic target. Drawing parallels with the imperative for healthcare providers to proactively engage with patients regarding treatment regimens, this review underscores the essential role of cardiologists and physicians in recognizing and addressing Lp(a) as a modifiable risk factor in AS management. Furthermore, it explores promising avenues of novel drug approaches, including emerging pharmacotherapies and targeted interventions, aimed at modulating Lp(a) levels and attenuating AS progression. By navigating the complexities of Lp(a) modulation and its implications for AS management, this review aims to bridge critical gaps in understanding and clinical practice, ultimately optimizing treatment strategies and improving patient outcomes in the realm of AS therapeutics.
Collapse
Affiliation(s)
- Mohammad Ishrak Khan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raisa Subaita Zahir
- College of Allopathic Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Abel Casso Dominguez
- Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francisco José Romeo
- Department of Cardiology, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, FL, USA
| |
Collapse
|
6
|
Schumann F, Kassner U, Spira D, Zimmermann FF, Bobbert T, Steinhagen-Thiessen E, Hollstein T. Long-term lipoprotein apheresis reduces cardiovascular events in high-risk patients with isolated lipoprotein(a) elevation. J Clin Lipidol 2024; 18:e738-e745. [PMID: 38908966 DOI: 10.1016/j.jacl.2024.04.134] [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: 07/04/2023] [Revised: 03/31/2024] [Accepted: 04/23/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Elevated lipoprotein(a) (Lp(a)) is an established risk factor for cardiovascular disease (CVD). To date, the only approved treatment to lower Lp(a) is lipoprotein apheresis (LA). Previous studies have demonstrated that LA is effective in reducing cardiovascular (CV) risk in patients with elevated low-density lipoprotein cholesterol (LDL-C) and/or Lp(a). Here we report our long-term experience with LA and its effectiveness in reducing CVD events in patients with elevated Lp(a). METHODS This retrospective open-label, single-center study included 25 individuals with Lp(a) elevation >60 mg/dL and LDL-C < 2.59 mmol/L who had indication for LA. The primary endpoint of this study was the incidence of any CV event (determined by medical records) after initiation of LA. RESULTS Mean LA treatment duration was 7.1 years (min-max: 1-19 years). Median Lp(a) was reduced from 95.0 to 31.1 mg/dL after LA (-67.3%, p < 0.0001). Mean LDL-C was reduced from 1.85 to 0.76 mmol/L after LA (-58.9%, p < 0.0001). Prior to LA, 81 CV events occurred in total (0.87 events/patient/year). During LA, 49 CV events occurred in total (0.24 events/patient/year; -0.63, p = 0.001). Yearly major adverse cardiac event (MACE) rate was reduced from 0.34 to 0.006 (-0.33, p = 0.0002). Similar results were obtained when considering only individuals with baseline LDL-C below 1.42 mmol/L. CONCLUSION In this observational study of a heterogeneous CV high-risk cohort with elevated Lp(a), LA reduced Lp(a) levels and was paralleled by a decrease in CV events and MACE. We recommend LA for patients with high Lp(a) who still have CV events despite optimal lipid-lowering medication and lifestyle changes.
Collapse
Affiliation(s)
- Friederike Schumann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology, Division of Lipid Disorders, Augustenburger Platz 1, 13353 Berlin, Germany (Drs Schumann, Kassner, Spira, Bobbert, Steinhagen-Thiessen and Hollstein)
| | - Ursula Kassner
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology, Division of Lipid Disorders, Augustenburger Platz 1, 13353 Berlin, Germany (Drs Schumann, Kassner, Spira, Bobbert, Steinhagen-Thiessen and Hollstein)
| | - Dominik Spira
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology, Division of Lipid Disorders, Augustenburger Platz 1, 13353 Berlin, Germany (Drs Schumann, Kassner, Spira, Bobbert, Steinhagen-Thiessen and Hollstein)
| | - Felix F Zimmermann
- Physikalisch-Technische Bundesanstalt (PTB), Berlin and Braunschweig, Germany (Dr Zimmermann)
| | - Thomas Bobbert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology, Division of Lipid Disorders, Augustenburger Platz 1, 13353 Berlin, Germany (Drs Schumann, Kassner, Spira, Bobbert, Steinhagen-Thiessen and Hollstein)
| | - Elisabeth Steinhagen-Thiessen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology, Division of Lipid Disorders, Augustenburger Platz 1, 13353 Berlin, Germany (Drs Schumann, Kassner, Spira, Bobbert, Steinhagen-Thiessen and Hollstein)
| | - Tim Hollstein
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology, Division of Lipid Disorders, Augustenburger Platz 1, 13353 Berlin, Germany (Drs Schumann, Kassner, Spira, Bobbert, Steinhagen-Thiessen and Hollstein); University of Kiel, Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, Arnold Heller Straße 3, Kiel 24105, Germany (Dr Hollstein).
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
Reijman MD, Kusters DM, Groothoff JW, Arbeiter K, Dann EJ, de Boer LM, de Ferranti SD, Gallo A, Greber-Platzer S, Hartz J, Hudgins LC, Ibarretxe D, Kayikcioglu M, Klingel R, Kolovou GD, Oh J, Planken RN, Stefanutti C, Taylan C, Wiegman A, Schmitt CP. Clinical practice recommendations on lipoprotein apheresis for children with homozygous familial hypercholesterolaemia: An expert consensus statement from ERKNet and ESPN. Atherosclerosis 2024; 392:117525. [PMID: 38598969 DOI: 10.1016/j.atherosclerosis.2024.117525] [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: 12/22/2023] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024]
Abstract
Homozygous familial hypercholesterolaemia is a life-threatening genetic condition, which causes extremely elevated LDL-C levels and atherosclerotic cardiovascular disease very early in life. It is vital to start effective lipid-lowering treatment from diagnosis onwards. Even with dietary and current multimodal pharmaceutical lipid-lowering therapies, LDL-C treatment goals cannot be achieved in many children. Lipoprotein apheresis is an extracorporeal lipid-lowering treatment, which is used for decades, lowering serum LDL-C levels by more than 70% directly after the treatment. Data on the use of lipoprotein apheresis in children with homozygous familial hypercholesterolaemia mainly consists of case-reports and case-series, precluding strong evidence-based guidelines. We present a consensus statement on lipoprotein apheresis in children based on the current available evidence and opinions from experts in lipoprotein apheresis from over the world. It comprises practical statements regarding the indication, methods, treatment goals and follow-up of lipoprotein apheresis in children with homozygous familial hypercholesterolaemia and on the role of lipoprotein(a) and liver transplantation.
Collapse
Affiliation(s)
- M Doortje Reijman
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - D Meeike Kusters
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jaap W Groothoff
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Klaus Arbeiter
- Division of Paediatric Nephrology and Gastroenterology, Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Eldad J Dann
- Blood Bank and Apheresis Unit Rambam Health Care Campus, Haifa, Israel
| | - Lotte M de Boer
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Sarah D de Ferranti
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Antonio Gallo
- Sorbonne Université, INSERM, UMR 1166, Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié-Salpêtrière, F-75013, Paris, France
| | - Susanne Greber-Platzer
- Clinical Division of Paediatric Pulmonology, Allergology and Endocrinology, Department of Paediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria
| | - Jacob Hartz
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Lisa C Hudgins
- The Rogosin Institute, Weill Cornell Medical College, New York, NY, USA
| | - Daiana Ibarretxe
- Vascular Medicine and Metabolism Unit (UVASMET), Hospital Universitari Sant Joan, Spain; Universitat Rovira i Virgili, Spain; Institut Investigació Sanitària Pere Virgili (IISPV)-CERCA, Spain; Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Meral Kayikcioglu
- Department of Cardiology, Medical Faculty, Ege University, 35100, Izmir, Turkey
| | - Reinhard Klingel
- Apheresis Research Institute, Stadtwaldguertel 77, 50935, Cologne, Germany(†)
| | - Genovefa D Kolovou
- Metropolitan Hospital, Department of Preventive Cardiology, 9, Ethn. Makariou & 1, El. Venizelou, N. Faliro, 185 47, Athens, Greece
| | - Jun Oh
- University Medical Center Hamburg/Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - R Nils Planken
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands
| | - Claudia Stefanutti
- Department of Molecular Medicine, Lipid Clinic and Atherosclerosis Prevention Centre, 'Umberto I' Hospital 'Sapienza' University of Rome, I-00161, Rome, Italy
| | - Christina Taylan
- Paediatric Nephrology, Children's and Adolescents' Hospital, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Albert Wiegman
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Claus Peter Schmitt
- Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University of Heidelberg, Germany
| |
Collapse
|
11
|
Pawlos A, Khoury E, Gaudet D. Emerging therapies for refractory hypercholesterolemia: a narrative review. Future Cardiol 2024; 20:317-334. [PMID: 38985520 PMCID: PMC11318688 DOI: 10.1080/14796678.2024.2367860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/11/2024] [Indexed: 07/12/2024] Open
Abstract
Refractory hypercholesterolemia (RH) is characterized by the failure of patients to achieve therapeutic targets for low-density lipoprotein-cholesterol (LDL-C) despite receiving maximal tolerable doses of standard lipid-lowering treatments. It predominantly impacts individuals with familial hypercholesterolemia (FH), thereby elevating the risk of cardiovascular complications. The prevalence of RH is now recognized to be substantially greater than previously thought. This review provides a comprehensive insight into current and emerging therapies for RH patients, including groundbreaking genetic-based therapeutic approaches. The review places emphasis on the dependency of therapies on low-density lipoprotein receptors (LDLRs) and highlights the critical role of considering LDLR activity in RH patients for individualization of the treatment.
Collapse
Affiliation(s)
- Agnieszka Pawlos
- Department of Internal Diseases & Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Kniaziewicza 1/5, 91-347, Lodz, Poland
| | - Etienne Khoury
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical Research Center, Chicoutimi, QC, Canada
| | - Daniel Gaudet
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical Research Center, Chicoutimi, QC, Canada
| |
Collapse
|
12
|
Gomez-Delgado F, Raya-Cruz M, Katsiki N, Delgado-Lista J, Perez-Martinez P. Residual cardiovascular risk: When should we treat it? Eur J Intern Med 2024; 120:17-24. [PMID: 37845117 DOI: 10.1016/j.ejim.2023.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/02/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023]
Abstract
Cardiovascular disease (CVD) still being the most common cause of death in worldwide. In spite of development of new lipid-lowering therapies which optimize low-density lipoprotein cholesterol (LDL-c) levels, recurrence of CVD events implies addressing factors related with residual cardiovascular (CV) risk. The key determinants of residual CV risk include triglyceride-rich lipoproteins (TRLs) and remnant cholesterol (RC), lipoprotein(a) [Lp(a)] and inflammation including its biochemical markers such as high sensitivity C reactive protein (hs-CRP). On the other hand, unhealthy lifestyle habits, environmental pollution, residual thrombotic risk and the residual metabolic risk determined by obesity and type 2 diabetes (T2D) have a specific weight in the residual CV risk. New pharmacologic therapies and pathways are being explored such as inhibition of apolipoprotein C-III (apoC-III) and angiopoietin-related protein 3 (ANGPTL3) in order to explore if a reduction in TRLs and RC reduce CVD events. Therapeutic target of inflammation plays an attractive way to reduce the atherosclerotic process and to date, approved therapies as colchicine plays a beneficial effect in chronic inflammation and residual CV risk. Lp(a) constitutes one of the most residual CV risk factor due to linkage with CVD and aortic valve stenosis. New and hopeful treatments including antisense oligonucleotides (ASO) and small-interfering ribonucleic acid (siRNA) which interfere in LP(a) codification have been developed to achieve an adequate control in Lp(a) levels. This review points out the paradigms of residual CV risk, discus how we should manage their features and summarize the different therapies targeting each residual CV risk factor.
Collapse
Affiliation(s)
- Francisco Gomez-Delgado
- Vascular Risk Unit, Internal Medicine Unit, Jaen University Hospital, Av. del Ejercito Español, 10, PC: 23007, Jaen, Spain; CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, PC: 28029, Madrid, Spain
| | - Manuel Raya-Cruz
- Vascular Risk Unit, Internal Medicine Unit, Jaen University Hospital, Av. del Ejercito Español, 10, PC: 23007, Jaen, Spain
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, 57400, Thessaloniki, Greece; School of Medicine, European University Cyprus, Nicosia, 2404, Cyprus
| | - Javier Delgado-Lista
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, PC: 28029, Madrid, Spain; Lipids and Atherosclerosis Unit, IMIBIC, Reina Sofía University Hospital, University of Cordoba, Av. Menendez Pidal, s/n, PC: 14004, Cordoba, Spain
| | - Pablo Perez-Martinez
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, PC: 28029, Madrid, Spain; Lipids and Atherosclerosis Unit, IMIBIC, Reina Sofía University Hospital, University of Cordoba, Av. Menendez Pidal, s/n, PC: 14004, Cordoba, Spain.
| |
Collapse
|
13
|
Thomas PE, Vedel-Krogh S, Nordestgaard BG. Measuring lipoprotein(a) for cardiovascular disease prevention - in whom and when? Curr Opin Cardiol 2024; 39:39-48. [PMID: 38078600 DOI: 10.1097/hco.0000000000001104] [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: 12/18/2023]
Abstract
PURPOSE OF REVIEW The aim of this study is to summarize major cardiovascular guideline recommendations on lipoprotein(a) and highlighting recent findings that emphasize how measuring lipoprotein(a) once in all adults is meaningful regardless of age, sex, comorbidities, or ethnicity. RECENT FINDINGS Many international guidelines now recommend once in a lifetime measurement of lipoprotein(a) in all adult individuals to facilitate accurate risk prediction. Lipoprotein(a)-lowering therapy to reduce cardiovascular disease is on the horizon, with results from the first phase 3 trial expected in 2025. SUMMARY Elevated lipoprotein(a) is an independent causal risk factor for atherosclerotic cardiovascular disease and aortic valve stenosis and measuring lipoprotein(a) once in all individuals regardless of age, sex, comorbidities, or ethnicity is meaningful to aid in risk stratification.
Collapse
Affiliation(s)
- Peter E Thomas
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Signe Vedel-Krogh
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Reijman MD, Kusters DM, Groothoff JW, Arbeiter K, Dann EJ, de Boer LM, de Ferranti SD, Gallo A, Greber-Platzer S, Hartz J, Hudgins LC, Ibarretxe D, Kayikcioglu M, Klingel R, Kolovou GD, Oh J, Planken RN, Stefanutti C, Taylan C, Wiegman A, Schmitt CP. Clinical practice recommendations on lipoprotein apheresis for children with homozygous familial hypercholesterolemia: an expert consensus statement from ERKNet and ESPN. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.14.23298547. [PMID: 38014132 PMCID: PMC10680892 DOI: 10.1101/2023.11.14.23298547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Homozygous familial hypercholesterolaemia is a life-threatening genetic condition, which causes extremely elevated LDL-C levels and atherosclerotic cardiovascular disease very early in life. It is vital to start effective lipid-lowering treatment from diagnosis onwards. Even with dietary and current multimodal pharmaceutical lipid-lowering therapies, LDL-C treatment goals cannot be achieved in many children. Lipoprotein apheresis is an extracorporeal lipid-lowering treatment, which is well established since three decades, lowering serum LDL-C levels by more than 70% per session. Data on the use of lipoprotein apheresis in children with homozygous familial hypercholesterolaemia mainly consists of case-reports and case-series, precluding strong evidence-based guidelines. We present a consensus statement on lipoprotein apheresis in children based on the current available evidence and opinions from experts in lipoprotein apheresis from over the world. It comprises practical statements regarding the indication, methods, treatment targets and follow-up of lipoprotein apheresis in children with homozygous familial hypercholesterolaemia and on the role of lipoprotein(a) and liver transplantation.
Collapse
Affiliation(s)
- M. Doortje Reijman
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - D. Meeike Kusters
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - Jaap W. Groothoff
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - Klaus Arbeiter
- Division of Paediatric Nephrology and Gastroenterology, Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Eldad J. Dann
- Blood Bank and apheresis unit Rambam Health care campus, Haifa, Israel
| | - Lotte M. de Boer
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - Sarah D. de Ferranti
- Department of Cardiology, Boston Children’s Hospital, Boston, MA, USA, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Antonio Gallo
- Sorbonne Université, INSERM, UMR 1166, Lipidology and cardiovascular prevention Unit, Department of Nutrition, APHP, Hôpital Pitié-Salpêtrière F-75013 Paris, France
| | - Susanne Greber-Platzer
- Clinical Division of Paediatric Pulmonology, Allergology and Endocrinology, Department of Paediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria
| | - Jacob Hartz
- Department of Cardiology, Boston Children’s Hospital, Boston, MA, USA, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Lisa C. Hudgins
- The Rogosin Institute, Weill Cornell Medical College, New York, New York, USA
| | - Daiana Ibarretxe
- Vascular Medicine and Metabolism Unit (UVASMET), Hospital Universitari Sant Joan; Universitat Rovira i Virgili; Institut Investigació Sanitària Pere Virgili (IISPV)-CERCA, Spain; Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Meral Kayikcioglu
- Department of Cardiology, Medical Faculty, Ege University, 35100 Izmir, Turkey
| | - Reinhard Klingel
- Apheresis Research Institute, Stadtwaldguertel 77, 50935 Cologne, Germany (www.apheresis-research.org)
| | - Genovefa D. Kolovou
- Metropolitan Hospital, Department of Preventive Cardiology. 9, Ethn. Makariou & 1, El. Venizelou, N. Faliro, 185 47, Athens, Greece
| | - Jun Oh
- University Medical Center Hamburg/Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - R. Nils Planken
- Department of Radiology and nuclear medicine, Amsterdam UMC, location AMC, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands
| | - Claudia Stefanutti
- Department of Molecular Medicine, Lipid Clinic and Atherosclerosis Prevention Centre, ‘Umberto I’ Hospital ‘Sapienza’ University of Rome, I-00161 Rome, Italy
| | - Christina Taylan
- Paediatric Nephrology, Children’s and Adolescents’ Hospital, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Albert Wiegman
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - Claus Peter Schmitt
- Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University of Heidelberg, Germany
| |
Collapse
|
15
|
Nurmohamed NS, Moriarty PM, Stroes ESG. Considerations for routinely testing for high lipoprotein(a). Curr Opin Lipidol 2023; 34:174-179. [PMID: 35942815 PMCID: PMC10328534 DOI: 10.1097/mol.0000000000000838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Lipoprotein (a) [Lp(a)] is a likely causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and aortic valve disease, confirmed by Mendelian randomization. With reliable assays, it has been established that Lp(a) is linearly associated with ASCVD. Current low-density lipoprotein cholesterol (LDL-C) lowering therapies do not or minimally lower Lp(a). This review focuses on the clinical importance and therapeutic consequences of Lp(a) measurement. RECENT FINDINGS Development of RNA-based Lp(a) lowering therapeutics has positioned Lp(a) as one of the principal residual risk factors to target in the battle against lipid-driven ASCVD risk. Pelacarsen, which is a liver-specific antisense oligonucleotide, has shown Lp(a) reductions up to 90% and its phase 3 trial is currently underway. Olpasiran is a small interfering RNA targeting LPA messenger RNA, which is being investigated in phase 2 and has already shown dose-dependent Lp(a) reductions up to 90%. SUMMARY Lp(a) should be measured in every patient at least once to identify patients with very high Lp(a) levels. These patients could benefit from Lp(a) lowering therapies when approved. In the meantime, therapy in high Lp(a) patients should focus on further reducing LDL-C and other ASCVD risk factors.
Collapse
Affiliation(s)
- Nick S Nurmohamed
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, The Netherlands
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Patrick M Moriarty
- Atherosclerosis and Lipid-apheresis Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Erik SG Stroes
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, The Netherlands
| |
Collapse
|
16
|
Alankar A, Brar PC, Kohn B. Lipoprotein(a): a Case for Universal Screening in Youth. Curr Atheroscler Rep 2023; 25:487-493. [PMID: 37405555 DOI: 10.1007/s11883-023-01120-3] [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: 06/14/2023] [Indexed: 07/06/2023]
Abstract
PURPOSE OF REVIEW Lipoprotein(a) has emerged as a strong independent risk factor for cardiovascular disease. Targeted screening recommendations for Lp(a) measurement exist for adults and youth known to be at high-risk. However, Lp(a) measurements are not included in universal screening guidelines in the US; hence, most families in the US with high Lp(a) levels who are at risk of future atherosclerotic heart disease, stroke, or aortic stenosis are not recognized. Lp(a) measurement included as part of routine universal lipid screening in youth would identify those children at risk of ASCVD and enable family cascade screening with identification and early intervention for affected family members. RECENT FINDINGS Lp(a) levels can be reliably measured in children as young as two years of age. Lp(a) levels are genetically determined. The Lp(a) gene is inherited in a co-dominant fashion. Serum Lp(a) attains adult levels by two years of age and is stable for the lifetime of the individual. Novel therapies that aim to specifically target Lp(a) are in the pipeline, including nucleic acid-based molecules such as antisense oligonucleotides and siRNAs. Inclusion of a single Lp(a) measurement performed as part of routine universal lipid screening in youth (ages 9-11; or at ages 17-21) is feasible and cost effective. Lp(a) screening would identify youth at-risk of ASCVD and enable family cascade screening with identification and early intervention for affected family members.
Collapse
Affiliation(s)
- Aparna Alankar
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Preneet C Brar
- NYU-Langone Medical Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Brenda Kohn
- NYU-Langone Medical Center, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
17
|
Farina JM, Pereyra M, Mahmoud AK, Chao CJ, Barry T, Halli Demeter SM, Ayoub C, Arsanjani R. Current Management and Future Perspectives in the Treatment of Lp(a) with a Focus on the Prevention of Cardiovascular Diseases. Pharmaceuticals (Basel) 2023; 16:919. [PMID: 37513831 PMCID: PMC10385436 DOI: 10.3390/ph16070919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Lipoprotein(a) [Lp(a)] is a lipid molecule with atherogenic, inflammatory, thrombotic, and antifibrinolytic effects, whose concentrations are predominantly genetically determined. The association between Lp(a) and cardiovascular diseases (CVDs) has been well-established in numerous studies, and the ability to measure Lp(a) levels is widely available in the community. As such, there has been increasing interest in Lp(a) as a therapeutic target for the prevention of CVD. The impact of the currently available lipid-modifying agents on Lp(a) is modest and heterogeneous, except for the monoclonal antibody proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), which demonstrated a significant reduction in Lp(a) levels. However, the absolute reduction in Lp(a) to significantly decrease CVD outcomes has not been definitely established, and the magnitude of the effect of PCSK9i seems insufficient to directly reduce the Lp(a)-related CVD risk. Therefore, emerging therapies are being developed that specifically aim to lower Lp(a) levels and the risk of CVD, including RNA interference (RNAi) agents, which have the capacity for temporary and reversible downregulation of gene expression. This review article aims to summarize the effects of Lp(a) on CVD and to evaluate the available evidence on established and emerging therapies targeting Lp(a) levels, focusing on the potential reduction of CVD risk attributable to Lp(a) concentrations.
Collapse
Affiliation(s)
- Juan M. Farina
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Ahmed K. Mahmoud
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Chieh-Ju Chao
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Timothy Barry
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Susan M. Halli Demeter
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| |
Collapse
|
18
|
Malick WA, Goonewardena SN, Koenig W, Rosenson RS. Clinical Trial Design for Lipoprotein(a)-Lowering Therapies: JACC Focus Seminar 2/3. J Am Coll Cardiol 2023; 81:1633-1645. [PMID: 37076218 DOI: 10.1016/j.jacc.2023.02.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 04/21/2023]
Abstract
Lipoprotein(a) [Lp(a)] is a source of residual risk in patients with atherosclerotic cardiovascular disease (ASCVD). Clinical trials of fully human monoclonal antibodies targeting proprotein convertase subtilisin kexin 9 have shown that reductions in Lp(a) concentrations may be a predictor of event reduction with this class of cholesterol-lowering therapy. With the advent of selective therapies targeting Lp(a) such as antisense oligonucleotides, small-interfering RNA-based therapies, and gene editing, lowering of Lp(a) may lead to reduction in ASCVD. The phase 3 Lp(a)HORIZON (Assessing the Impact of Lipoprotein(a) Lowering with TQJ230 on Major Cardiovascular Events in Patients With CVD) outcomes trial is currently testing the effect of pelacarsen, an antisense oligonucleotide, on ASCVD risk. Olpasiran is a small-interfering RNA that is in a phase 3 clinical trial. As these therapies enter clinical trials, challenges in trial design will have to be addressed to optimize patient selection and outcomes.
Collapse
Affiliation(s)
- Waqas A Malick
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Wolfgang Koenig
- Deutsches Herzzentrum Muenchen, Technische Universitat Muenchen, Munich, DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany; Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Robert S Rosenson
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
19
|
van den Bosch SE, Corpeleijn WE, Hutten BA, Wiegman A. How Genetic Variants in Children with Familial Hypercholesterolemia Not Only Guide Detection, but Also Treatment. Genes (Basel) 2023; 14:669. [PMID: 36980941 PMCID: PMC10048736 DOI: 10.3390/genes14030669] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Familial hypercholesterolemia (FH) is a hereditary disorder that causes severely elevated low-density lipoprotein (LDL-C) levels, which leads to an increased risk for premature cardiovascular disease. A variety of genetic variants can cause FH, namely variants in the genes for the LDL receptor (LDLR), apolipoprotein B (APOB), proprotein convertase subtilisin/kexin type 9 (PCSK9), and/or LDL-receptor adaptor protein 1 (LDLRAP1). Variants can exist in a heterozygous form (HeFH) or the more severe homozygous form (HoFH). If affected individuals are diagnosed early (through screening), they benefit tremendously from early initiation of lipid-lowering therapy, such as statins, and cardiovascular imaging to detect possible atherosclerosis. Over the last years, due to intensive research on the genetic basis of LDL-C metabolism, novel, promising therapies have been developed to reduce LDL-C levels and subsequently reduce cardiovascular risk. Results from studies on therapies focused on inhibiting PCSK9, a protein responsible for degradation of the LDLR, are impressive. As the effect of PCSK9 inhibitors (PCSK9-i) is dependent of residual LDLR activity, this medication is less potent in patients without functional LDLR (e.g., null/null variant). Novel therapies that are expected to become available in the near future focused on inhibition of another major regulatory protein in lipid metabolism (angiopoietin-like 3 (ANGPTL3)) might dramatically reduce the frequency of apheresis in children with HoFH, independently of their residual LDLR. At present, another independent risk factor for premature cardiovascular disease, elevated levels of lipoprotein(a) (Lp(a)), cannot be effectively treated with medication. Further understanding of the genetic basis of Lp(a) metabolism, however, offers a possibility for the development of novel therapies.
Collapse
Affiliation(s)
- Sibbeliene E. van den Bosch
- Department of Pediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Willemijn E. Corpeleijn
- Department of Pediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Barbara A. Hutten
- Department of Epidemiology and Data Science, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Albert Wiegman
- Department of Pediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
20
|
Tsamoulis D, Siountri I, Rallidis LS. Lipoprotein(a): Its Association with Calcific Aortic Valve Stenosis, the Emerging RNA-Related Treatments and the Hope for a New Era in “Treating” Aortic Valve Calcification. J Cardiovasc Dev Dis 2023; 10:jcdd10030096. [PMID: 36975859 PMCID: PMC10056331 DOI: 10.3390/jcdd10030096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
The treatment of patients with aortic valve calcification (AVC) and calcific aortic valve stenosis (CAVS) remains challenging as, until today, all non-invasive interventions have proven fruitless in preventing the disease’s onset and progression. Despite the similarities in the pathogenesis of AVC and atherosclerosis, statins failed to show a favorable effect in preventing AVC progression. The recognition of lipoprotein(a) [Lp(a)] as a strong and potentially modifiable risk factor for the development and, perhaps, the progression of AVC and CAVS and the evolution of novel agents leading in a robust Lp(a) reduction, have rekindled hope for a promising future in the treatment of those patients. Lp(a) seems to promote AVC via a ‘three hit’ mechanism including lipid deposition, inflammation and autotaxin transportation. All of these lead to valve interstitial cells transition into osteoblast-like cells and, thus, to parenchymal calcification. Currently available lipid-lowering therapies have shown a neutral or mild effect on Lp(a), which was proven insufficient to contribute to clinical benefits. The short-term safety and the efficacy of the emerging agents in reducing Lp(a) have been proven; nevertheless, their effect on cardiovascular risk is currently under investigation in phase 3 clinical trials. A positive result of these trials will probably be the spark to test the hypothesis of the modification of AVC’s natural history with the novel Lp(a)-lowering agents.
Collapse
Affiliation(s)
- Donatos Tsamoulis
- 1st Department of Internal Medicine, Thriasio General Hospital of Eleusis, 192 00 Athens, Greece
- Society of Junior Doctors, 5 Menalou Str., 151 23 Athens, Greece
| | - Iliana Siountri
- 1st Department of Internal Medicine, General Hospital of Nikaia “Agios Panteleimon”, 184 54 Nikaia, Greece
| | - Loukianos S. Rallidis
- Second Department of Cardiology, National & Kapodistrian University of Athens, School of Medicine, University General Hospital ATTIKON, 124 62 Athens, Greece
- Correspondence:
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Apheresis is a treatment option for severe dyslipidemia which has been introduced approximately 40 years ago to clinical practice. This article reviews recent apheresis research progresses, including apheresis for elevated LDL-cholesterol and elevated lipoprotein(a). RECENT FINDINGS While the role of apheresis in treating more common forms of LDL-hypercholesterolemia has been reduced due to the development of new, very potent LDL-lowering drugs, it still plays an important role in treating patients with homozygous familial hypercholesterolemia and patients with severe lipoprotein(a) elevation. One apheresis session can decrease LDL-cholesterol, apoB, and lipoprotein(a) by approximately 65%, which results in a time averaged reduction of 30-50%. Although time-consuming, and expensive regular apheresis is very well tolerated and has been proven safe for decades. Apheresis remains a treatment option for severe dyslipidemia, especially in homozygous familial hypercholesterolemia and elevated lipoprotein(a), if other forms of therapy fail to achieve targets.
Collapse
Affiliation(s)
- Klaus G Parhofer
- Medical Department IV - Grosshadern, University Munich, Marchioninistraße 15, 81377, Munich, Germany.
| |
Collapse
|
22
|
Waitz G, Atiye S, Gauly A, Prophet H. Comparison of plasma separation using centrifugation or filtration for MONET lipoprotein apheresis in patients with cardiovascular disease and severe dyslipidemia. Ther Apher Dial 2022; 26:1281-1288. [PMID: 35322939 PMCID: PMC9790347 DOI: 10.1111/1744-9987.13840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/11/2022] [Accepted: 03/21/2022] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Homozygous or severe heterozygous familial hypercholesterolemia and elevated lipoprotein(a) levels may be treated with membrane filtration. The MONET system (Fresenius Medical Care, Bad Homburg, Germany) involves plasma separation by centrifugation or filtration. METHODS Whether the method of plasma separation affects lipoprotein lowering and treatment safety was investigated in a single-center retrospective study. RESULTS The centrifugation-based plasma separation achieved a higher plasma flow and shorter time to treat 1 L of plasma (46.2 ± 8.6 min), than the filtration-based system (71.5 ± 40.0 min; p = 0.001). The mean reduction of LDL-cholesterol was 69% and 67% with centrifugation and filtration and was 75% for lipoprotein(a) with both plasma separation methods. A reduction of IgM by more than 60%, of albumin and total protein by approximately 20% and low frequency of side effects was observed. CONCLUSIONS The efficacy of lowering atherogenic lipoproteins was comparable with both plasma separation methods. Centrifugation was more time-efficient compared to filtration.
Collapse
Affiliation(s)
| | - Saynab Atiye
- Global Medical Office, Fresenius Medical CareBad HomburgGermany
| | - Adelheid Gauly
- Global Medical Office, Fresenius Medical CareBad HomburgGermany
| | | |
Collapse
|
23
|
Wang L, Liu L, Zhao Y, Chu M, Teng J. Lipoprotein(a) and residual vascular risk in statin-treated patients with first acute ischemic stroke: A prospective cohort study. Front Neurol 2022; 13:1004264. [PMID: 36408516 PMCID: PMC9671150 DOI: 10.3389/fneur.2022.1004264] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/17/2022] [Indexed: 08/26/2023] Open
Abstract
OBJECTIVES Statins either barely affect or increase lipoprotein(a) [Lp(a)] levels. This study aimed to explore the factors correlated to the change of Lp(a) levels as well as the relationship between Lp(a) and the recurrent vascular events in statin-treated patients with first acute ischemic stroke (AIS). METHODS Patients who were admitted to the hospital with first AIS from October 2018 to September 2020 were eligible for inclusion. Correlation between the change of Lp(a) levels and potential influencing factors was assessed by linear regression analysis. Cox proportional regression models were used to estimate the association between Lp(a) and recurrent vascular events including AIS, transient ischemic attack, myocardial infarction and coronary revascularization. RESULTS In total, 303 patients, 69.6% males with mean age 64.26 ± 11.38 years, completed the follow-up. During the follow-up period, Lp(a) levels increased in 50.5% of statin-treated patients and the mean percent change of Lp(a) levels were 14.48% (95% CI 6.35-22.61%). Creatinine (β = 0.152, 95% CI 0.125-0.791, P = 0.007) and aspartate aminotransferase (AST) (β = 0.160, 95% CI 0.175-0.949, P = 0.005) were positively associated with the percent change of Lp(a) levels. During a median follow-up of 26 months, 66 (21.8%) patients had a recurrent vascular event. The median time period between AIS onset and vascular events recurrence was 9.5 months (IQR 2.0-16.3 months). The on-statin Lp(a) level ≥70 mg/dL (HR 2.539, 95% CI 1.076-5.990, P = 0.033) and the change of Lp(a) levels (HR 1.003, 95% CI 1.000-1.005, P = 0.033) were associated with the recurrent vascular events in statin-treated patients with first AIS. Furthermore, the on-statin Lp(a) levels ≥70 mg/dL (HR 3.612, 95% CI 1.018-12.815, P = 0.047) increased the risk of recurrent vascular events in patients with low-density lipoprotein cholesterol (LDL-C) levels < 1.8 mmol/L. CONCLUSIONS Lp(a) levels increased in half of statin-treated patients with first AIS. Creatinine and AST were positively associated with the percent change of Lp(a) levels. Lp(a) is a determinant of residual vascular risk and the change of Lp(a) is positively associated with the risk of recurrent vascular events in these patients.
Collapse
Affiliation(s)
- Lanjing Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Lijun Liu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yanhong Zhao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Min Chu
- Department of Neurology, Minhang Hospital, Fudan University, Qingdao, China
| | - Jijun Teng
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
24
|
Bhatia HS, Wilkinson MJ. Lipoprotein(a): Evidence for Role as a Causal Risk Factor in Cardiovascular Disease and Emerging Therapies. J Clin Med 2022; 11:6040. [PMID: 36294361 PMCID: PMC9604626 DOI: 10.3390/jcm11206040] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/20/2022] [Accepted: 10/07/2022] [Indexed: 08/03/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is an established risk factor for multiple cardiovascular diseases. Several lines of evidence including mechanistic, epidemiologic, and genetic studies support the role of Lp(a) as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and aortic stenosis/calcific aortic valve disease (AS/CAVD). Limited therapies currently exist for the management of risk associated with elevated Lp(a), but several targeted therapies are currently in various stages of clinical development. In this review, we detail evidence supporting Lp(a) as a causal risk factor for ASCVD and AS/CAVD, and discuss approaches to managing Lp(a)-associated risk.
Collapse
Affiliation(s)
| | - Michael J. Wilkinson
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
25
|
Abstract
Purpose of Review Over the past decades, genetic and observational evidence has positioned lipoprotein(a) as novel important and independent risk factor for cardiovascular disease (ASCVD) and aortic valve stenosis. Recent Findings As Lp(a) levels are determined genetically, lifestyle interventions have no effect on Lp(a)-mediated ASCVD risk. While traditional low-density lipoprotein cholesterol (LDL-C) can now be effectively lowered in the vast majority of patients, current lipid lowering therapies have no clinically relevant Lp(a) lowering effect. Summary There are multiple Lp(a)-directed therapies in clinical development targeting LPA mRNA that have shown to lower Lp(a) plasma levels for up to 90%: pelacarsen, olpasiran, and SLN360. Pelacarsen is currently investigated in a phase 3 cardiovascular outcome trial expected to finish in 2024, while olpasiran is about to proceed to phase 3 and SLN360’s phase 1 outcomes were recently published. If proven efficacious, Lp(a) will soon become the next pathway to target in ASCVD risk management.
Collapse
|
26
|
de Boer LM, Wiegman A, Swerdlow DI, Kastelein JJP, Hutten BA. Pharmacotherapy for children with elevated levels of lipoprotein(a): future directions. Expert Opin Pharmacother 2022; 23:1601-1615. [PMID: 36047306 DOI: 10.1080/14656566.2022.2118522] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Elevated lipoprotein(a) [Lp(a)] is an independent risk factor for atherosclerotic cardiovascular disease (ASCVD). With the advent of the antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs) targeted at LPA, the gene encoding apolipoprotein(a), that are highly effective for lowering Lp(a) levels, this risk factor might be managed in the near future. Given that Lp(a) levels are mostly genetically determined and once elevated, present from early age, we have evaluated future directions for the treatment of children with high Lp(a) levels. AREAS COVERED In the current review, we discuss different pharmacological treatments in clinical development and provide an in-depth overview of the effects of ASOs and siRNAs targeted at LPA. EXPERT OPINION Since high Lp(a) is an important risk factor for ASCVD and given the promising effects of both ASOs and siRNAs targeted at apo(a), there is an urgent need for well-designed prospective studies to assess the impact of elevated Lp(a) in childhood. If the Lp(a)-hypothesis is confirmed in adults, and also in children, the rationale might arise for treating children with high Lp(a) levels. However, we feel that this should be limited to children with the highest cardiovascular risk including familial hypercholesterolemia and potentially pediatric stroke.
Collapse
Affiliation(s)
- Lotte M de Boer
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Department of Pediatrics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Albert Wiegman
- Department of Pediatrics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - John J P Kastelein
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Barbara A Hutten
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
27
|
A cross-sectional study to assess proteinuria and lipoprotein (a) levels in chronic kidney disease. EUREKA: HEALTH SCIENCES 2022. [DOI: 10.21303/2504-5679.2022.002567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Chronic kidney disease (CKD) is a reduced glomerular filtration rate and/or increased urinary albumin excretion. The worldwide prevalence of chronic kidney disease (CKD) ranges from 8 to 16 %, and the prevalence of CKD is rising.
The aim: To study the association between CKD stages, proteinuria, and lipoprotein (a) levels among the study participants.
Materials and methods: This study was an institution-based observational case-control study involving CKD patients as study group and healthy volunteers as control one. Blood samples were tested for urea, serum creatinine, uric acid levels, triglycerides, total cholesterol, HDL cholesterol, VLDL cholesterol and serum lipoprotein. Statistical analysis was done with SPSS version 20.0.
Result: In our study, the most common age group affected among cases was 41 to 50 years (5th decade), and there was a male preponderance in CKD. CKD patients had a higher mean protein creatinine ratio than controls, and this difference was statistically significant. In addition, CKD patients had significantly higher total cholesterol, triglyceride levels and lower HDL cholesterol levels than controls. Also, they had significantly elevated serum lipoprotein (a) levels than controls.
Conclusion: Based on our study findings, we can conclude that because of the potential role of lipoprotein (a) in the development of cardiovascular disease, it is imperative to include an estimation of lipoprotein (a) levels in all CKD patients, especially in later stages to give a targeted therapy for dyslipidemia among CKD patients
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW Lipoprotein(a) (Lp[a]) is a likely causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and aortic valve disease, confirmed by Mendelian randomization. With reliable assays, it has been established that Lp(a) is linearly associated with ASCVD. Current low-density lipoprotein cholesterol (LDL-C) lowering therapies do not or minimally lower Lp(a). This review focuses on the clinical importance and therapeutic consequences of Lp(a) measurement. RECENT FINDINGS Development of RNA-based Lp(a) lowering therapeutics has positioned Lp(a) as one of the principal residual risk factors to target in the battle against lipid-driven ASCVD risk. Pelacarsen, which is a liver-specific antisense oligonucleotide, has shown Lp(a) reductions up to 90% and its phase 3 trial is currently underway. Olpasiran is a small interfering RNA targeting LPA messenger RNA which is being investigated in phase 2 and has already shown dose-dependent Lp(a) reductions up to 90%. SUMMARY Lp(a) should be measured in every patient at least once to identify patients with very high Lp(a) levels. These patients could benefit from Lp(a) lowering therapies when approved. In the meantime, therapy in high Lp(a) patients should focus on further reducing LDL-C and other ASCVD risk factors.
Collapse
Affiliation(s)
- Nick S Nurmohamed
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, The Netherlands
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Patrick M Moriarty
- Atherosclerosis and Lipid-apheresis Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, The Netherlands
| |
Collapse
|
29
|
Schwartz GG, Ballantyne CM. Existing and emerging strategies to lower Lipoprotein(a). Atherosclerosis 2022; 349:110-122. [DOI: 10.1016/j.atherosclerosis.2022.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 12/24/2022]
|
30
|
Kong P, Cui ZY, Huang XF, Zhang DD, Guo RJ, Han M. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther 2022; 7:131. [PMID: 35459215 PMCID: PMC9033871 DOI: 10.1038/s41392-022-00955-7] [Citation(s) in RCA: 500] [Impact Index Per Article: 166.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease driven by traditional and nontraditional risk factors. Genome-wide association combined with clonal lineage tracing and clinical trials have demonstrated that innate and adaptive immune responses can promote or quell atherosclerosis. Several signaling pathways, that are associated with the inflammatory response, have been implicated within atherosclerosis such as NLRP3 inflammasome, toll-like receptors, proprotein convertase subtilisin/kexin type 9, Notch and Wnt signaling pathways, which are of importance for atherosclerosis development and regression. Targeting inflammatory pathways, especially the NLRP3 inflammasome pathway and its regulated inflammatory cytokine interleukin-1β, could represent an attractive new route for the treatment of atherosclerotic diseases. Herein, we summarize the knowledge on cellular participants and key inflammatory signaling pathways in atherosclerosis, and discuss the preclinical studies targeting these key pathways for atherosclerosis, the clinical trials that are going to target some of these processes, and the effects of quelling inflammation and atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiao-Fu Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Dan-Dan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Rui-Juan Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
31
|
Rhainds D, Brodeur MR, Tardif JC. Lipoprotein (a): When to Measure and How to Treat? Curr Atheroscler Rep 2021; 23:51. [PMID: 34235598 DOI: 10.1007/s11883-021-00951-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review current evidence for lipoprotein (a) (Lp(a)) as a risk factor for multiple cardiovascular (CV) disease phenotypes, provide a rationale for Lp(a) lowering to reduce CV risk, identify therapies that lower Lp(a) levels that are available clinically and under investigation, and discuss future directions. RECENT FINDINGS Mendelian randomization and epidemiological studies have shown that elevated Lp(a) is an independent and causal risk factor for atherosclerosis and major CV events. Lp(a) is also associated with non-atherosclerotic endpoints such as venous thromboembolism and calcific aortic valve disease. It contributes to residual CV risk in patients receiving standard-of-care LDL-lowering therapy. Plasma Lp(a) levels present a skewed distribution towards higher values and vary widely between individuals and according to ethnic background due to genetic variants in the LPA gene, but remain relatively constant throughout a person's life. Thus, elevated Lp(a) (≥50 mg/dL) is a prevalent condition affecting >20% of the population but is still underdiagnosed. Treatment guidelines have begun to advocate measurement of Lp(a) to identify patients with very high levels that have a family history of premature CVD or elevated Lp(a). Lipoprotein apheresis (LA) efficiently lowers Lp(a) and was recently associated with a reduction of incident CV events. Statins have neutral or detrimental effects on Lp(a), while PCSK9 inhibitors significantly reduce its level by up to 30%. Specific lowering of Lp(a) with antisense oligonucleotides (ASO) shows good safety and strong efficacy with up to 90% reductions. The ongoing CV outcomes study Lp(a)HORIZON will provide a first answer as to whether selective Lp(a) lowering with ASO reduces the risk of major CV events. Given the recently established association between Lp(a) level and CV risk, guidelines now recommend Lp(a) measurement in specific clinical conditions. Accordingly, Lp(a) is a current target for drug development to reduce CV risk in patients with elevated levels, and lowering Lp(a) with ASO represents a promising avenue.
Collapse
Affiliation(s)
- David Rhainds
- Montreal Heart Institute Research Center, 5000 Belanger Street, Montréal, Canada
| | - Mathieu R Brodeur
- Montreal Heart Institute Research Center, 5000 Belanger Street, Montréal, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute Research Center, 5000 Belanger Street, Montréal, Canada. .,Faculty of Medicine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
32
|
Abstract
Lipoprotein(a) [Lp(a)] is an atherogenic lipoprotein with a strong genetic regulation. Up to 90% of the concentrations are explained by a single gene, the LPA gene. The concentrations show a several-hundred-fold interindividual variability ranging from less than 0.1 mg/dL to more than 300 mg/dL. Lp(a) plasma concentrations above 30 mg/dL and even more above 50 mg/dL are associated with an increased risk for cardiovascular disease including myocardial infarction, stroke, aortic valve stenosis, heart failure, peripheral arterial disease, and all-cause mortality. Since concentrations above 50 mg/dL are observed in roughly 20% of the Caucasian population and in an even higher frequency in African-American and Asian-Indian ethnicities, it can be assumed that Lp(a) is one of the most important genetically determined risk factors for cardiovascular disease.Carriers of genetic variants that are associated with high Lp(a) concentrations have a markedly increased risk for cardiovascular events. Studies that used these genetic variants as a genetic instrument to support a causal role for Lp(a) as a cardiovascular risk factor are called Mendelian randomization studies. The principle of this type of studies has been introduced and tested for the first time ever with Lp(a) and its genetic determinants.There are currently no approved pharmacologic therapies that specifically target Lp(a) concentrations. However, some therapies that target primarily LDL cholesterol have also an influence on Lp(a) concentrations. These are mainly PCSK9 inhibitors that lower LDL cholesterol by 60% and Lp(a) by 25-30%. Furthermore, lipoprotein apheresis lowers both, Lp(a) and LDL cholesterol, by about 60-70%. Some sophisticated study designs and statistical analyses provided support that lowering Lp(a) by these therapies also lowers cardiovascular events on top of the effect caused by lowering LDL cholesterol, although this was not the main target of the therapy. Currently, new therapies targeting RNA such as antisense oligonucleotides (ASO) or small interfering RNA (siRNA) against apolipoprotein(a), the main protein of the Lp(a) particle, are under examination and lower Lp(a) concentrations up to 90%. Since these therapies specifically lower Lp(a) concentrations without influencing other lipoproteins, they will serve the last piece of the puzzle whether a decrease of Lp(a) results also in a decrease of cardiovascular events.
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Genetic, epidemiological, and translational data indicate that Lipoprotein (a) [Lp(a)] is likely in the causal pathway for atherosclerotic cardiovascular diseases as well as calcification of the aortic valves. RECENT FINDINGS Lp(a) is structurally similar to low-density lipoprotein, but in addition to apolipoprotein B-100, it has a glycoprotein apolipoprotein(a) [apo(a)], which is attached to the apolipoprotein B-100. Several distinctive properties of Lp(a) can be attributed to the presence of apo(a). This review discusses the current state of literature on pathophysiological and clinical aspects of Lp(a). After five decades of research, the understanding of Lp(a) structure, biochemistry, and pathophysiology of its cardiovascular manifestations still remains less than fully understood. Universally, Lp(a) elevation may be the most predominant monogenetic lipid disorder with approximate prevalence of Lp(a)>50 mg/dL among estimated >1.4 billion people. This makes a compelling rationale for diagnosing and managing Lp(a)-mediated risk. In addition to discussing various cardiovascular phenotypes of Lp(a) and associated morbidity, we also outline current and emerging therapies aimed at identifying a definitive treatment for elevated Lp(a) levels.
Collapse
Affiliation(s)
- Anum Saeed
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA USA
| | - Sina Kinoush
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX USA
| | - Salim S. Virani
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX USA
- Health Policy, Quality & Informatics Program, Michael E. DeBakey Veterans Affairs Medical Center Health Services Research and Development Center for Innovations, Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, 2002 Holcombe Blvd, Houston, TX 77030 USA
- Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston, TX USA
| |
Collapse
|
34
|
Özdemir ZN, Şahin U, Yıldırım Y, Kaya CT, İlhan O. Lipoprotein apheresis efficacy and challenges: single center experience. Hematol Transfus Cell Ther 2021; 44:56-62. [PMID: 33745887 PMCID: PMC8885363 DOI: 10.1016/j.htct.2021.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/05/2020] [Accepted: 01/12/2021] [Indexed: 11/17/2022] Open
Abstract
Introduction Lipoprotein apheresis (LA) is an extracorporeal therapy which removes apolipoprotein B-containing particles from the circulation. We evaluated techniques and efficiency of lipoprotein apheresis procedures applied to patients with familial and non-familial hypercholesterolemia (FH) at our center. Methods We retrospectively evaluated 250 LA procedures applied to 27 patients with dyslipidemia between March 2011 and August 2019. Results A total of 27 patients, of whom 19 (70.4%) were male and 8 (29.6%), female, were included. Eighteen (66.7%), 6 (22.2%) and 3 (11.1%) patients were diagnosed with non-FH, homozygous FH (HoFH) and heterozygous FH (HeFH), respectively. Two different apheresis techniques, direct adsorption of lipoproteins (DALI) (48.8%) and double filtration plasmapheresis (DFPP) (51.2%), were used. The change in the serum total cholesterol (TC) level was the median 302 mg/dl (171–604 mg/dl) (60.4%) in HoFH patients, 305 mg/dl (194–393 mg/dl) (60.8%) in HeFH patients and 227 mg/dl (75–749 mg/dl) (65.3%) in non-FH patients. The change in the serum low-density lipoprotein (LDL) level was the median 275 mg/dl (109–519 mg/dl) (64.2%), 232 mg/dl (207–291 mg/dl) (64.5%) and 325 mg/dl (22–735 mg/dl) (70.9%) in patients with HoFH, HeFH and non-FH, respectively. A significantly effective reduction in serum lipid levels, including TC, LDL and triglycerides, was achieved in all patients, regardless of the technique, p < .001. The decrease in the serum TC and LDL levels was significantly higher in the DFPP, compared to the DALI, being 220 mg/dl (−300 to 771) vs 184 mg/dl (64–415), p < .001 and 196 mg/dl (11–712) vs 157 mg/dl (54–340), p < .001, respectively. Conclusions Our results showed that LA is a highly effective treatment in reducing serum lipid levels and safe, without any major adverse event.
Collapse
Affiliation(s)
| | - Uğur Şahin
- Medicana International Ankara Hospital, Ankara, Turkey
| | | | | | - Osman İlhan
- Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
35
|
Rosenson RS. Existing and emerging therapies for the treatment of familial hypercholesterolemia. J Lipid Res 2021; 62:100060. [PMID: 33716107 PMCID: PMC8065289 DOI: 10.1016/j.jlr.2021.100060] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/12/2021] [Accepted: 02/21/2021] [Indexed: 12/30/2022] Open
Abstract
Familial hypercholesterolemia (FH), an autosomal dominant disorder of LDL metabolism that is characterized by elevated LDL-cholesterol, is commonly encountered in patients with atherosclerotic coronary heart disease. Combinations of cholesterol-lowering therapies are often used to lower LDL-cholesterol in patients with FH; however, current treatment goals for LDL-cholesterol are rarely achieved in patients with homozygous FH (HoFH) and are difficult to achieve in patients with heterozygous FH (HeFH). Therapies that lower LDL-cholesterol through LDL receptor-mediated mechanisms have thus far been largely ineffective in patients with HoFH, particularly in those with negligible (<2%) LDL receptor activity. Among patients with HeFH who were at very high risk for atherosclerotic cardiovascular disease events, combined therapy consisting of a high dose of high-intensity statin, ezetimibe, and proprotein convertase subtilisin Kexin type 9 inhibitor failed to lower LDL-cholesterol to minimal acceptable goals in more than 50%. This article provides a framework for the use of available and emerging treatments that lower LDL-cholesterol in adult patients with HoFH and HeFH. A framework is provided for the use of angiopoietin-like protein 3 inhibitors in the treatment of HoFH and HeFH.
Collapse
Affiliation(s)
- Robert S Rosenson
- Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health. Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
36
|
Tsimikas S, Moriarty PM, Stroes ES. Emerging RNA Therapeutics to Lower Blood Levels of Lp(a): JACC Focus Seminar 2/4. J Am Coll Cardiol 2021; 77:1576-1589. [PMID: 33766265 DOI: 10.1016/j.jacc.2021.01.051] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/30/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022]
Abstract
Lipoprotein(a) [Lp(a)] has risen to the level of an accepted cardiovascular disease risk factor, but final proof of causality awaits a randomized trial of Lp(a) lowering. Inhibiting apolipoprotein(a) production in the hepatocyte with ribonucleic acid therapeutics has emerged as an elegant and effective solution to reduce plasma Lp(a) levels. Phase 2 clinical trials have shown that the antisense oligonucleotide pelacarsen reduced mean Lp(a) levels by 80%, allowing 98% of subjects to reach on-treatment levels of <125 nmol/l (∼50 mg/dl). The phase 3 Lp(a)HORIZON (Assessing the Impact of Lipoprotein(a) Lowering With TQJ230 on Major Cardiovascular Events in Patients With CVD) outcomes trial is currently enrolling approximately 7,680 patients with history of myocardial infarction, ischemic stroke, and symptomatic peripheral arterial disease and controlled low-density lipoprotein cholesterol to pelacarsen versus placebo. The co-primary endpoints are major adverse cardiovascular events in subjects with Lp(a) >70 mg/dl and >90 mg/dl, in which either of the two being positive will lead to a successful trial. Additional ribonucleic acid-targeted therapies to lower Lp(a) are in preclinical and clinical development. The testing of the Lp(a) hypothesis will provide proof whether Lp(a)-mediated risk can be abolished by potent Lp(a) lowering.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, California, USA.
| | - Patrick M Moriarty
- Division of Clinical Pharmacology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Missouri, USA
| | - Erik S Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
37
|
Liu Y, Wang W, Song J, Zhang K, Xu B, Li P, Shao C, Yang M, Chen J, Tang YD. Association Between Lipoprotein(a) and Peri-procedural Myocardial Infarction in Patients With Diabetes Mellitus Who Underwent Percutaneous Coronary Intervention. Front Endocrinol (Lausanne) 2021; 11:603922. [PMID: 33613445 PMCID: PMC7888338 DOI: 10.3389/fendo.2020.603922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
Background High lipoprotein(a) (Lp[a]) levels are associated with increased risks of cardiovascular events in Percutaneous Coronary Intervention (PCI) patients with diabetes mellitus (DM). Peri-procedural myocardial infarction (PMI) occurs commonly during the PCI, whereas the relationship between Lp(a) and PMI remains unclear. Our study aimed to evaluate the association between Lp(a) value and the incidence of PMI in a larger-scale diabetic cohort undergoing PCI throughout 2013. Methods A total of 2,190 consecutive patients with DM were divided into two groups according to the median Lp(a) level of 175 mg/L: Low Lp(a) group (N = 1095) and high Lp(a) group (N = 1095). PMI was defined based on the 2018 universal definition of myocardial infarction. Results Patients with high Lp(a) levels exhibited higher rates of PMI compared to those with low Lp(a) levels (2.3% versus 0.8%, P = 0.006). The multivariable logistic analysis showed that PMI was independently predicted by Lp(a) as a dichotomous variable (OR 2.64, 95%CI 1.22-5.70) and as a continuous variable (OR 1.57, 95% CI 1.12-2.20). However, further investigation found that this association was only maintained in men, whose Lp(a) levels were significantly associated with the frequency of PMI, both as a dichotomous variable (OR 3.66, 95%CI 1.34-10.01) and as a continuous variable (OR 1.81, 95%CI 1.18-2.78). Lp(a) wasn't a risk factor of PMI in women. Conclusions High Lp(a) levels had forceful correlations with the increased frequency of PMI in male diabetic patients undergoing PCI. Lp(a) might act as a marker of risk stratification and a therapeutic target to reduce PCI-related ischemic events.
Collapse
Affiliation(s)
- Yupeng Liu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Peking University Third Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenyao Wang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Song
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Peking University Third Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kuo Zhang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Xu
- Department of Cardiac Catheterization Laboratory, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ping Li
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunli Shao
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Chen
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Da Tang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Peking University Third Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
38
|
Mickiewicz A, Marlega J, Kuchta A, Bachorski W, Cwiklinska A, Raczak G, Gruchala M, Fijalkowski M. Cardiovascular events in patients with familial hypercholesterolemia and hyperlipoproteinaemia (a): Indications for lipoprotein apheresis in Poland. J Clin Apher 2021; 36:370-378. [PMID: 33386762 DOI: 10.1002/jca.21872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/07/2020] [Accepted: 12/21/2020] [Indexed: 11/07/2022]
Abstract
BACKGROUND Lipoprotein apheresis (LA) is a safe method of reducing atherogenic lipoproteins and improving cardiovascular (CV) outcomes. We aimed to assess the reductions in low-density lipoprotein cholesterol (LDL-C) and lipoprotein (a) [Lp(a)] levels in patients undergoing regular LA therapy and to evaluate its influence on the incidence rate of adverse cardiac and vascular events (ACVE) and major adverse cardiac events (MACE). METHODS A longitudinal study in Poland evaluated the prospective and retrospective observational data of 23 patients with hyperlipoproteinaemia (a) [hyper-Lp(a)] and familial hypercholesterolemia (FH), undergoing 1014 LA sessions between 2013 and 2020. Their pre- and post-apheresis LDL-C and Lp(a) levels were assessed to calculate the acute percent reductions. The time period used to evaluate annual rates of ACVE and MACE before and after initiation of LA was matched in each patient. RESULTS The pre-apheresis LDL-C and Lp(a) concentrations were 155 (107-228) (mg/dL) (median and interquartile range) and 0.56 (0.14-1.37) (g/L), respectively. LA therapy resulted in a reduction of LDL-C to 50 (30-73.5) (mg/dL) and of Lp(a) to 0.13 (0.05-0.34) (g/L), representing a percent reduction of 70.0% and 72.7% for LDL-C and Lp(a), respectively. We found a significant reduction in the annual rate of ACVE (0.365[0.0-0.585] vs (0.0[0.0-0.265]; P = .047) and MACE (0.365[0.0-0.585] vs 0.0[0.0-0.265]; P = .031). CONCLUSIONS The findings of our study indicate that LA treatment in patients with hyperlipoproteinaemia (a) and FH on maximally tolerated lipid lowering therapies leads to a substantial reduction in LDL-C and Lp(a) concentrations and lowers CV event rates in Polish patients.
Collapse
Affiliation(s)
| | - Joanna Marlega
- Department of Cardiology I, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Kuchta
- Department of Clinical Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Witold Bachorski
- Department of Cardiology I, Medical University of Gdansk, Gdansk, Poland
| | | | - Grzegorz Raczak
- Department of Cardiology & Electrotherapy, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Gruchala
- Department of Cardiology I, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Fijalkowski
- Department of Cardiology I, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
39
|
Pokrovsky SN, Afanasieva OI, Ezhov MV. Therapeutic Apheresis for Management of Lp(a) Hyperlipoproteinemia. Curr Atheroscler Rep 2020; 22:68. [DOI: 10.1007/s11883-020-00886-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
40
|
Moriarty PM, Gorby LK, Stroes ES, Kastelein JP, Davidson M, Tsimikas S. Lipoprotein(a) and Its Potential Association with Thrombosis and Inflammation in COVID-19: a Testable Hypothesis. Curr Atheroscler Rep 2020; 22:48. [PMID: 32710255 PMCID: PMC7381416 DOI: 10.1007/s11883-020-00867-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The COVID-19 pandemic has infected over > 11 million as of today people worldwide and is associated with significant cardiovascular manifestations, particularly in subjects with preexisting comorbidities and cardiovascular risk factors. Recently, a predisposition for arterial and venous thromboses has been reported in COVID-19 infection. We hypothesize that besides conventional risk factors, subjects with elevated lipoprotein(a) (Lp(a)) may have a particularly high risk of developing cardiovascular complications. RECENT FINDINGS The Lp(a) molecule has the propensity for inhibiting endogenous fibrinolysis through its apolipoprotein(a) component and for enhancing proinflammatory effects such as through its content of oxidized phospholipids. The LPA gene contains an interleukin-6 (IL-6) response element that may induce an acute phase-type increase in Lp(a) levels following a cytokine storm from COVID-19. Thus, subjects with either baseline elevated Lp(a) or those who have an increase following COVID-19 infection, or both, may be at very high risk of developing thromboses. Elevated Lp(a) may also lead to acute destabilization of preexisting but quiescent atherosclerotic plaques, which might induce acute myocardial infarction and stroke. Ongoing studies with IL-6 antagonists may be informative in understanding this relationship, and registries are being initiated to measure Lp(a) in subjects infected with COVID-19. If indeed an association is suggestive of being causal, consideration can be given to systematic testing of Lp(a) and prophylactic systemic anticoagulation in infected inpatients. Therapeutic lipid apheresis and pharmacotherapy for the reduction of Lp(a) levels may minimize thrombogenic potential and proinflammatory effects. We propose studies to test the hypothesis that Lp(a) may contribute to cardiovascular complications of COVID-19.
Collapse
Affiliation(s)
- Patrick M Moriarty
- Division of Clinical Pharmacology, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Lauryn K Gorby
- Division of Clinical Pharmacology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Erik S Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - John P Kastelein
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Michael Davidson
- Lipid Clinic, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA.
- Vascular Medicine Program, Sulpizio Cardiovascular Center, University of California San Diego, 9500 Gilman Drive, BSB 1080, La Jolla, CA, 92093-0682, USA.
| |
Collapse
|
41
|
Xiao L, Bai T, Zeng J, Yang R, Yang L. Nonalcoholic fatty liver disease, a potential risk factor of non-specific ST-T segment changes: data from a cross-sectional study. PeerJ 2020; 8:e9090. [PMID: 32440372 PMCID: PMC7229768 DOI: 10.7717/peerj.9090] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/08/2020] [Indexed: 12/26/2022] Open
Abstract
Background Non-specific ST-T segment changes are prevalent and are proven risk factors for early onset of cardiovascular diseases. They can increase all-cause mortality by 100∼200% and are candidate for early signs of cardiovascular changes. Nonalcoholic fatty liver disease (NAFLD) is prevalent worldwide and is one facet of a multisystem disease that confers substantial increases morbidity and mortality of nonalcoholic fatty liver-related cardiovascular diseases. It is unclear whether NAFLD is associated with non-specific ST-T changes warning early signs of cardiovascular changes. Therefore, we investigated this association. Methods A cross-sectional study was designed that included a sample consisting of 32,922 participants who underwent health examinations. Participants with missing information, excessive alcohol intake, viral hepatitis, chronic liver disease or established cardiovascular diseases were excluded. Electrocardiograms were used for analysis of non-specific ST-T segment changes. NAFLD was diagnosed by ultrasonographic detection of hepatic steatosis without other liver diseases. A multivariable logistic regression model was served to calculate the OR and 95% CI for non-specific ST-T segment changes. Results The prevalence of non-specific ST-T segment changes was 6.5% in participants with NAFLD, however, the prevalence of NAFLD was 42.9% in participants with non-specific ST-T segment changes. NAFLD was independently associated with non-specific ST-T segment changes (OR: 1.925, 95% CI: 1.727-2.143, P < 0.001). After adjusting for age, sex, heart rate, hypertension, body mass index, fasting glucose, total cholesterol, triglycerides, HDL-C, NAFLD remained an independent risk factor of non-specific ST-T segment changes (OR: 1.289, 95% CI: 1.122-1.480). Conclusion Non-specific ST-T segment changes were independently associated with the presence of NAFLD after adjusting for potential confounders.
Collapse
Affiliation(s)
- Li Xiao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Bai
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junchao Zeng
- Physical Examination (Health Management) Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Yang
- Physical Examination (Health Management) Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
42
|
Tsimikas S, Stroes ES. The dedicated “Lp(a) clinic”: A concept whose time has arrived? Atherosclerosis 2020; 300:1-9. [DOI: 10.1016/j.atherosclerosis.2020.03.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 03/02/2020] [Accepted: 03/05/2020] [Indexed: 12/24/2022]
|
43
|
From the editor: Margaritaville blues and Philadelphia news. J Clin Lipidol 2019; 13:857-858. [PMID: 31837748 DOI: 10.1016/j.jacl.2019.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|