1
|
Clarke R, Wright N, Lin K, Yu C, Walters RG, Lv J, Hill M, Kartsonaki C, Millwood IY, Bennett DA, Avery D, Yang L, Chen Y, Du H, Sherliker P, Yang X, Sun D, Li L, Qu C, Marcovina S, Collins R, Chen Z, Parish S. Causal Relevance of Lp(a) for Coronary Heart Disease and Stroke Types in East Asian and European Ancestry Populations: A Mendelian Randomization Study. Circulation 2025. [PMID: 40297899 DOI: 10.1161/circulationaha.124.072086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Elevated plasma levels of Lp(a) [lipoprotein(a)] are a causal risk factor for coronary heart disease and stroke in European individuals, but the causal relevance of Lp(a) for different stroke types and in East Asian individuals with different Lp(a) genetic architecture is uncertain. METHODS We measured plasma levels of Lp(a) in a nested case-control study of 18 174 adults (mean [SD] age, 57 [10] years; 49% female) in the China Kadoorie Biobank (CKB) and performed a genome-wide association analysis to identify genetic variants affecting Lp(a) levels, with replication in ancestry-specific subsets in UK Biobank. We further performed 2-sample Mendelian randomization analyses, associating ancestry-specific Lp(a)-associated instrumental variants derived from CKB or from published data in European individuals with risk of myocardial infarction (n=17 091), ischemic stroke (IS [n=29 233]) and its subtypes, or intracerebral hemorrhage (n=5845) in East Asian and European individuals using available data from CKB and genome-wide association analysis consortia. RESULTS In CKB observational analyses, plasma levels of Lp(a) were log-linearly and positively associated with higher risks of myocardial infarction and IS, but not with ICH. In genome-wide association analysis, we identified 29 single nucleotide polymorphisms independently associated with Lp(a) that together explained 33% of variance in Lp(a) in Chinese individuals. In UK Biobank, the lead Chinese variants identified in CKB were replicated in 1260 Chinese individuals, but explained only 10% of variance in Lp(a) in European individuals. In Mendelian randomization analyses, however, there were highly concordant effects of Lp(a) across both ancestries for all cardiovascular disease outcomes examined. In combined analyses of both ancestries, the proportional reductions in risk per 100 nmol/L lower genetically predicted Lp(a) levels for myocardial infarction were 3-fold greater than for total IS (rate ratio, 0.78 [95% CI, 0.76-0.81] versus 0.94 [0.92-0.96]), but were similar to those for large-artery IS (0.80 [0.73-0.87]; n=8134). There were weaker associations with cardioembolic IS (0.92 [95% CI, 0.86-0.98]; n=11 730), and no association with small-vessel IS (0.99 [0.91-1.07]; n=12 343) or with intracerebral hemorrhage (1.08 [0.96-1.21]; n=5845). CONCLUSIONS The effects of Lp(a) on risk of myocardial infarction and large-artery IS were comparable in East Asian and European individuals, suggesting that people with either ancestry could expect comparable proportional benefits for equivalent reductions in Lp(a), but there was little effect on other stroke types.
Collapse
Affiliation(s)
- Robert Clarke
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Neil Wright
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Kuang Lin
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Robin G Walters
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Michael Hill
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Christiana Kartsonaki
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Iona Y Millwood
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Derrick A Bennett
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Daniel Avery
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Ling Yang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Yiping Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Huaidong Du
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Paul Sherliker
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Xiaoming Yang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Chan Qu
- NCDs Prevention and Control Department, Liuyang CDC, China (C.Q.)
| | | | - Rory Collins
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Zhengming Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Sarah Parish
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| |
Collapse
|
2
|
Kaur G, Rosenson RS, Gencer B, López JAG, Lepor NE, Baum SJ, Stout E, Gaudet D, Knusel B, Park JG, Wang H, Wu Y, Kassahun H, Sabatine MS, O'Donoghue ML. Olpasiran lowering of lipoprotein(a) according to baseline levels: insights from the OCEAN(a)-DOSE study. Eur Heart J 2025; 46:1162-1164. [PMID: 39565305 DOI: 10.1093/eurheartj/ehae781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/03/2024] [Accepted: 10/27/2024] [Indexed: 11/21/2024] Open
Affiliation(s)
- Gurleen Kaur
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Baris Gencer
- Cardiology Division, Geneva University Hospitals, Geneva, Switzerland
- The Institute of Primary Health Care, University of Bern, Bern, Switzerland
| | | | - Norman E Lepor
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Elmer Stout
- Crossroads Clinical Research, Mooresville, NC
| | - Daniel Gaudet
- Department of Medicine, Université de Montréal, Montréal, Canada
| | - Beat Knusel
- Global Development, Amgen, Thousand Oaks, CA, USA
| | - Jeong-Gun Park
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Rd, 7th Floor, Boston, MA 02115, USA
| | - Huei Wang
- Global Development, Amgen, Thousand Oaks, CA, USA
| | - You Wu
- Global Development, Amgen, Thousand Oaks, CA, USA
| | | | - Marc S Sabatine
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Rd, 7th Floor, Boston, MA 02115, USA
| | - Michelle L O'Donoghue
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Rd, 7th Floor, Boston, MA 02115, USA
| |
Collapse
|
3
|
Doherty S, Hernandez S, Rikhi R, Mirzai S, De Los Reyes C, McIntosh S, Block RC, Shapiro MD. Lipoprotein(a) as a Causal Risk Factor for Cardiovascular Disease. CURRENT CARDIOVASCULAR RISK REPORTS 2025; 19:8. [PMID: 39980866 PMCID: PMC11836235 DOI: 10.1007/s12170-025-00760-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 02/22/2025]
Abstract
Purpose of Review Lipoprotein(a) [Lp(a)], an atherogenic low-density lipoprotein cholesterol (LDL-C)-like molecule, has emerged as an important risk factor for the development of atherosclerotic cardiovascular disease (ASCVD). This review summarizes the evidence supporting Lp(a) as a causal risk factor for ASCVD and calcific aortic valve stenosis (CAVS). Recent Findings Lp(a) is largely (~ 90%) genetically determined and approximately 20% of the global population has elevated Lp(a). The unique structure of Lp(a) leads to proatherogenic, proinflammatory, and antifibrinolytic properties. Data from epidemiological, genome-wide association, Mendelian randomization, and meta-analyses have shown a clear association between Lp(a) and ASCVD, as well as CAVS. There are emerging data on the association between Lp(a) and ischemic stroke, peripheral arterial disease, and heart failure; however, the associations are not as strong. Summary Several lines of evidence support Lp(a) as a causal risk factor for ASCVD and CAVS. The 2024 National Lipid Association guidelines, 2022 European Atherosclerosis Society, and 2021 Canadian Cardiology Society guidelines recommend testing Lp(a) once in all adults to guide primary prevention efforts. Further studies on cardiovascular outcomes with Lp(a) targeted therapies will provide more insight on causal relationship between Lp(a) and cardiovascular disease.
Collapse
Affiliation(s)
- Sean Doherty
- Department of Internal Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Sebastian Hernandez
- Department of Internal Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Rishi Rikhi
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Department of Internal Medicine, Medical Center Boulevard, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Saeid Mirzai
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Department of Internal Medicine, Medical Center Boulevard, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Chris De Los Reyes
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, 14642 Rochester, NY USA
| | - Scott McIntosh
- Department of Public Health Sciences, Division of Public Health Sciences, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Robert C. Block
- Department of Public Health Sciences, Division of Epidemiology, Department of Medicine’s Cardiology Division, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642 USA
| | - Michael D. Shapiro
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Department of Internal Medicine, Medical Center Boulevard, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| |
Collapse
|
4
|
Wierzbicki AS. Advances in the pharmacological management of hyperlipidemia through the use of combination therapies. Expert Opin Pharmacother 2025; 26:157-165. [PMID: 39709627 DOI: 10.1080/14656566.2024.2444986] [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: 09/16/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Lipid-lowering therapies are well established for the treatment of cardiovascular disease (CVD). Historically monotherapy studies have been performed, but the introduction of statins has led to these drugs being recognized as baseline therapies and to the investigation of combination therapy of both older and newer medications with them. AREAS COVERED Surrogate marker studies have shown additive effects on LDL-C, triglycerides and HDL-C of combination therapies with statins and these have extended to lipoprotein (a). Imaging studies have often shown benefits paralleling lipid studies. However, outcome studies have failed to show added benefits with niacin or fibrates while confirming the benefits of ezetimibe, bempedoic acid and proprotein convertase subtilisin kexin-9 (PCSK-9) inhibitors and icosapent ethyl. EXPERT OPINION Combination therapy for LDL-C in dual combinations is well validated. Data for intervention on triglycerides is limited to icosapent ethyl, but this may exert effects independent of lipids. New drugs targeting triglycerides through apolipoprotein C3 and angiopoietin-like peptides are in development. Studies on combination therapy raising HDL-C have generally disappointed, though cholesterol ester transfer protein (CETP) inhibition remains a target. Lipoprotein (a) is recognized as a CVD risk factor and effective therapies are in development but results on CVD events are lacking.
Collapse
Affiliation(s)
- Anthony S Wierzbicki
- Department of Metabolic Medicine/Chemical Pathology Guy's, St Thomas' Hospitals, London, UK
| |
Collapse
|
5
|
Sheashaa H, Mousa H, Abbas MT, Farina JM, Awad K, Pereyra M, Scalia IG, Ali NB, Javadi N, Bismee NN, Esfahani SA, Ibrahim O, Abdelfattah F, Ibrahim R, Abdelnabi M, Ayoub C, Arsanjani R. Interaction Between Lipoprotein(a) and Other Lipid Molecules: A Review of the Current Literature. Biomolecules 2025; 15:162. [PMID: 40001465 PMCID: PMC11853184 DOI: 10.3390/biom15020162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Lipoprotein(a) [Lp(a)] is a well-established causal risk factor for cardiovascular diseases (CVDs), as reported by multiple Mendelian randomization studies and large epidemiological studies. When elevated Lp(a) is combined with other risk factors, most notably elevated low-density lipoprotein cholesterol (LDL-C), a synergistic atherogenic effect has been reported. However, the current literature is conflicting regarding how Lp(a) interacts in the context of controlled LDL-C levels (e.g., <70 mg/dL) and whether reducing LDL-C can modify the atherogenic effect of Lp(a). In some studies, elevated Lp(a) was still significantly associated with a higher risk of cardiovascular events, despite controlled levels of LDL-C. In contrast, multiple studies have reported attenuation of the cardiovascular risk mediated by elevated Lp(a) with lower LDL-C levels. Moreover, the relationship between Lp(a) and triglycerides, high-density lipoprotein, and very low-density lipoprotein remains unclear. In this literature review, we summarize and discuss the current evidence regarding the interactions between Lp(a) and other lipid molecules, how they contribute to the pathogenesis of CVD, and future perspectives, particularly in the current era where promising targeted Lp(a)-lowering therapies are under development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (H.S.); (H.M.); (M.T.A.); (J.M.F.); (K.A.); (M.P.); (I.G.S.); (N.B.A.); (N.J.); (N.N.B.); (S.A.E.); (O.I.); (F.A.); (R.I.); (M.A.); (C.A.)
| |
Collapse
|
6
|
Akinlonu A, Boffa MB, Lyu C, Zhong J, Jindal M, Fadzan M, Garshick MS, Schwartzbard A, Weintraub HS, Bredefeld C, Newman JD, Fisher EA, Koschinsky ML, Goldberg IJ, Berger JS. Variation in lipoprotein(a) response to potent lipid lowering: The role of apolipoprotein (a) isoform size. J Clin Lipidol 2025; 19:39-50. [PMID: 39828454 PMCID: PMC11908881 DOI: 10.1016/j.jacl.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is a driver of residual cardiovascular risk. Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) decrease Lp(a) with significant heterogeneity in response. We investigated contributors to the heterogeneous response. METHODS Cholesterol Reduction and Residual Risk in Diabetes (CHORD) was a prospective study examining lipid lowering in participants with a low-density lipoprotein cholesterol (LDL-C) > 100 mg/dL with and without diabetes (DM) on lipid lowering therapy (LLT) for 30-days with evolocumab 140 mg every 14 days combined with either atorvastatin 80 mg or ezetimibe 10 mg daily. Lp(a) level was measured by immunoturbidometry, and the apolipoprotein(a) [apo(a)] isoform size was measured by denaturing agarose gel electrophoresis and Western blotting. We examined the change in Lp(a) levels from baseline to 30 days. RESULTS Among 150 participants (mean age 50 years, 58% female, 50% non-White, 17% Hispanic, 50% DM), median (interquartile range) Lp(a) was 27.5 (8-75) mg/dL at baseline and 23 (3-68) mg/dL at 30 days, leading to a 10% (0-36) median reduction (P < .001). Among 73 (49%) participants with Lp(a) ≥ 30 mg/dL at baseline, there was a 15% (3-25) median reduction in Lp(a) (P < .001). While baseline Lp(a) level was not correlated with change in Lp(a) (r = 0.04, P = .59), apo(a) size directly correlated with Lp(a) reduction (P < .001). After adjustment for age, sex, race/ethnicity, DM, and type of LLT, apo(a) size remained positively associated with a reduction in Lp(a) (Beta 0.95, 95% confidence interval, 0.93-0.97, P < .001). CONCLUSION Our data demonstrate variation in Lp(a) reduction with potent LLT. Change in Lp(a) was strongly associated with apo(a) isoform size.
Collapse
Affiliation(s)
- Adedoyin Akinlonu
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Akinlonu, Jindal, Fadzan, Garshick, Schwartzbard, Weintraub, Newman, Fisher, Goldberg and Berger).
| | - Michael B Boffa
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada (Dr Boffa and Koschinsky)
| | - Chen Lyu
- Division of Biostatistics, Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA (Dr Lyu, Zhong)
| | - Judy Zhong
- Division of Biostatistics, Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA (Dr Lyu, Zhong)
| | - Manila Jindal
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Akinlonu, Jindal, Fadzan, Garshick, Schwartzbard, Weintraub, Newman, Fisher, Goldberg and Berger)
| | - Maja Fadzan
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Akinlonu, Jindal, Fadzan, Garshick, Schwartzbard, Weintraub, Newman, Fisher, Goldberg and Berger)
| | - Michael S Garshick
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Akinlonu, Jindal, Fadzan, Garshick, Schwartzbard, Weintraub, Newman, Fisher, Goldberg and Berger)
| | - Arthur Schwartzbard
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Akinlonu, Jindal, Fadzan, Garshick, Schwartzbard, Weintraub, Newman, Fisher, Goldberg and Berger)
| | - Howard S Weintraub
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Akinlonu, Jindal, Fadzan, Garshick, Schwartzbard, Weintraub, Newman, Fisher, Goldberg and Berger)
| | - Cindy Bredefeld
- Department of Medicine, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, USA (Dr Bredefeld)
| | - Jonathan D Newman
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Akinlonu, Jindal, Fadzan, Garshick, Schwartzbard, Weintraub, Newman, Fisher, Goldberg and Berger)
| | - Edward A Fisher
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Akinlonu, Jindal, Fadzan, Garshick, Schwartzbard, Weintraub, Newman, Fisher, Goldberg and Berger)
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada (Dr Boffa and Koschinsky)
| | - Ira J Goldberg
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Akinlonu, Jindal, Fadzan, Garshick, Schwartzbard, Weintraub, Newman, Fisher, Goldberg and Berger); Holman Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Goldberg)
| | - Jeffrey S Berger
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA (Dr Akinlonu, Jindal, Fadzan, Garshick, Schwartzbard, Weintraub, Newman, Fisher, Goldberg and Berger).
| |
Collapse
|
7
|
Schuth U, Gill K, Telyuk P, Bawamia BR, Austin D, Zaman A. Role of Lipoprotein(a) Reduction in Cardiovascular Disease. J Clin Med 2024; 13:6311. [PMID: 39518451 PMCID: PMC11546410 DOI: 10.3390/jcm13216311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Recent studies have shown that lipoprotein(a) (Lp(a)) is an important risk factor for a plethora of different cardiovascular diseases. It has been proven that Lp(a) levels are genetically determined and correlate with risk of cardiovascular disease, independent of lifestyle factors. As of yet, treatment options to reduce Lp(a) levels are limited, but new research into Lp(a) reduction yields promising results. This review delves into Lp(a)'s biochemistry and mechanism of effect, the association between Lp(a) and cardiovascular diseases, and possible therapies to minimise cardiovascular disease.
Collapse
Affiliation(s)
- Uma Schuth
- Faculty of Medicine, St Andrews University, St Andrews KY16 9ST, UK;
| | - Kieran Gill
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4AX, UK;
| | - Pyotr Telyuk
- Academic Cardiovascular Unit, The James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough TS4 3BW, UK; (P.T.); (D.A.)
| | | | - David Austin
- Academic Cardiovascular Unit, The James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough TS4 3BW, UK; (P.T.); (D.A.)
- Population Health Sciences Institute, Newcastle University, Newcastle-upon-Tyne NE2 4AX, UK
| | - Azfar Zaman
- Cardiology, Freeman Hospital and Vascular Biology and Medicine, Newcastle University, Newcastle upon Tyne NE2 4AX, UK
| |
Collapse
|
8
|
Boffa MB, Koschinsky ML. Lipoprotein(a) and cardiovascular disease. Biochem J 2024; 481:1277-1296. [PMID: 39302109 PMCID: PMC11555715 DOI: 10.1042/bcj20240037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
Elevated plasma levels of lipoprotein(a) (Lp(a)) are a prevalent, independent, and causal risk factor for atherosclerotic cardiovascular disease and calcific aortic valve disease. Lp(a) consists of a lipoprotein particle resembling low density lipoprotein and the covalently-attached glycoprotein apolipoprotein(a) (apo(a)). Novel therapeutics that specifically and potently lower Lp(a) levels are currently in advanced stages of clinical development, including in large, phase 3 cardiovascular outcomes trials. However, fundamental unanswered questions remain concerning some key aspects of Lp(a) biosynthesis and catabolism as well as the true pathogenic mechanisms of the particle. In this review, we describe the salient biochemical features of Lp(a) and apo(a) and how they underlie the disease-causing potential of Lp(a), the factors that determine plasma Lp(a) concentrations, and the mechanism of action of Lp(a)-lowering drugs.
Collapse
Affiliation(s)
- Michael B. Boffa
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Marlys L. Koschinsky
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
9
|
Delgado-Lista J, Mostaza JM, Arrobas-Velilla T, Blanco-Vaca F, Masana L, Pedro-Botet J, Perez-Martinez P, Civeira F, Cuende-Melero JI, Gomez-Barrado JJ, Lahoz C, Pintó X, Suarez-Tembra M, Lopez-Miranda J, Guijarro C. Consensus on lipoprotein(a) of the Spanish Society of Arteriosclerosis. Literature review and recommendations for clinical practice. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:243-266. [PMID: 38599943 DOI: 10.1016/j.arteri.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
The irruption of lipoprotein(a) (Lp(a)) in the study of cardiovascular risk factors is perhaps, together with the discovery and use of proprotein convertase subtilisin/kexin type 9 (iPCSK9) inhibitor drugs, the greatest novelty in the field for decades. Lp(a) concentration (especially very high levels) has an undeniable association with certain cardiovascular complications, such as atherosclerotic vascular disease (AVD) and aortic stenosis. However, there are several current limitations to both establishing epidemiological associations and specific pharmacological treatment. Firstly, the measurement of Lp(a) is highly dependent on the test used, mainly because of the characteristics of the molecule. Secondly, Lp(a) concentration is more than 80% genetically determined, so that, unlike other cardiovascular risk factors, it cannot be regulated by lifestyle changes. Finally, although there are many promising clinical trials with specific drugs to reduce Lp(a), currently only iPCSK9 (limited for use because of its cost) significantly reduces Lp(a). However, and in line with other scientific societies, the SEA considers that, with the aim of increasing knowledge about the contribution of Lp(a) to cardiovascular risk, it is relevant to produce a document containing the current status of the subject, recommendations for the control of global cardiovascular risk in people with elevated Lp(a) and recommendations on the therapeutic approach to patients with elevated Lp(a).
Collapse
Affiliation(s)
- Javier Delgado-Lista
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España.
| | - Jose M Mostaza
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario La Paz, Madrid, España
| | - Teresa Arrobas-Velilla
- Sociedad Española de Medicina de Laboratorio (SEQCML), Laboratorio de Bioquímica Clínica, Hospital Universitario Virgen Macarena, Sevilla, España
| | - Francisco Blanco-Vaca
- Departamento de Bioquímica Clínica, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB Sant Pau), Barcelona; Departamento de Bioquímica y Biología Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, España
| | - Luis Masana
- Unidad de Medicina Vascular y Metabolismo, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Tarragona, España
| | - Juan Pedro-Botet
- Unidad de Lípidos y Riesgo Vascular, Servicio de Endocrinología y Nutrición, Hospital del Mar, Barcelona; Departamento de Medicina, Universidad Autónoma de Barcelona, Barcelona, España
| | - Pablo Perez-Martinez
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España
| | - Fernando Civeira
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Servicio de Medicina Interna, Hospital Universitario Miguel Servet, IIS Aragón, Universidad de Zaragoza, Zaragoza; CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, España
| | - Jose I Cuende-Melero
- Consulta de Riesgo Vascular, Servicio de Medicina Interna, Complejo Asistencial Universitario de Palencia, Palencia; Departamento de Medicina, Dermatología y Toxicología, Facultad de Medicina, Universidad de Valladolid, Valladolid, España
| | - Jose J Gomez-Barrado
- Unidad de Cuidados Cardiológicos Agudos y Riesgo Cardiovascular, Servicio de Cardiología, Hospital Universitario San Pedro de Alcántara, Cáceres, España
| | - Carlos Lahoz
- Unidad de Lípidos y Arteriosclerosis, Servicio de Medicina Interna, Hospital La Paz-Carlos III, Madrid, España
| | - Xavier Pintó
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell-Universidad de Barcelona-CiberObn, España
| | - Manuel Suarez-Tembra
- Unidad de Lípidos y RCV, Servicio de Medicina Interna, Hospital San Rafael, A Coruña, España
| | - Jose Lopez-Miranda
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España.
| | - Carlos Guijarro
- Unidad de Medicina Interna, Hospital Universitario Fundación Alcorcón, Universidad Rey Juan Carlos, Alcorcón, Madrid, España
| |
Collapse
|
10
|
Volgman AS, Koschinsky ML, Mehta A, Rosenson RS. Genetics and Pathophysiological Mechanisms of Lipoprotein(a)-Associated Cardiovascular Risk. J Am Heart Assoc 2024; 13:e033654. [PMID: 38879448 PMCID: PMC11255763 DOI: 10.1161/jaha.123.033654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Abstract
Elevated lipoprotein(a) is a genetically transmitted codominant trait that is an independent risk driver for cardiovascular disease. Lipoprotein(a) concentration is heavily influenced by genetic factors, including LPA kringle IV-2 domain size, single-nucleotide polymorphisms, and interleukin-1 genotypes. Apolipoprotein(a) is encoded by the LPA gene and contains 10 subtypes with a variable number of copies of kringle -2, resulting in >40 different apolipoprotein(a) isoform sizes. Genetic loci beyond LPA, such as APOE and APOH, have been shown to impact lipoprotein(a) levels. Lipoprotein(a) concentrations are generally 5% to 10% higher in women than men, and there is up to a 3-fold difference in median lipoprotein(a) concentrations between racial and ethnic populations. Nongenetic factors, including menopause, diet, and renal function, may also impact lipoprotein(a) concentration. Lipoprotein(a) levels are also influenced by inflammation since the LPA promoter contains an interleukin-6 response element; interleukin-6 released during the inflammatory response results in transient increases in plasma lipoprotein(a) levels. Screening can identify elevated lipoprotein(a) levels and facilitate intensive risk factor management. Several investigational, RNA-targeted agents have shown promising lipoprotein(a)-lowering effects in clinical studies, and large-scale lipoprotein(a) testing will be fundamental to identifying eligible patients should these agents become available. Lipoprotein(a) testing requires routine, nonfasting blood draws, making it convenient for patients. Herein, we discuss the genetic determinants of lipoprotein(a) levels, explore the pathophysiological mechanisms underlying the association between lipoprotein(a) and cardiovascular disease, and provide practical guidance for lipoprotein(a) testing.
Collapse
Affiliation(s)
| | - Marlys L. Koschinsky
- Robarts Research Institute, Schulich School of Medicine and DentistryWestern UniversityLondonONCanada
| | | | - Robert S. Rosenson
- Metabolism and Lipids Program, Mount Sinai Fuster Heart HospitalIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| |
Collapse
|
11
|
Sosnowska B, Stepinska J, Mitkowski P, Bielecka-Dabrowa A, Bobrowska B, Budzianowski J, Burchardt P, Chlebus K, Dobrowolski P, Gasior M, Jankowski P, Kubica J, Mickiewicz A, Mysliwiec M, Osadnik T, Prejbisz A, Rajtar-Salwa R, Wita K, Witkowski A, Gil R, Banach M. Recommendations of the Experts of the Polish Cardiac Society (PCS) and the Polish Lipid Association (PoLA) on the diagnosis and management of elevated lipoprotein(a) levels. Arch Med Sci 2024; 20:8-27. [PMID: 38414479 PMCID: PMC10895977 DOI: 10.5114/aoms/183522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 02/29/2024] Open
Abstract
Lipoprotein(a) [Lp(a)] is made up of a low-density lipoprotein (LDL) particle and a specific apolipoprotein(a). The blood concentration of Lp(a) is approximately 90% genetically determined, and the main genetic factor determining Lp(a) levels is the size of the apo(a) isoform, which is determined by the number of KIV2 domain repeats. The size of the apo(a) isoform is inversely proportional to the blood concentration of Lp(a). Lp(a) is a strong and independent cardiovascular risk factor. Elevated Lp(a) levels ≥ 50 mg/dl (≥ 125 nmol/l) are estimated to occur in more than 1.5 billion people worldwide. However, determination of Lp(a) levels is performed far too rarely, including Poland, where, in fact, it is only since the 2021 guidelines of the Polish Lipid Association (PoLA) and five other scientific societies that Lp(a) measurements have begun to be performed. Determination of Lp(a) concentrations is not easy due to, among other things, the different sizes of the apo(a) isoforms; however, the currently available certified tests make it possible to distinguish between people with low and high cardiovascular risk with a high degree of precision. In 2022, the first guidelines for the management of patients with elevated lipoprotein(a) levels were published by the European Atherosclerosis Society (EAS) and the American Heart Association (AHA). The first Polish guidelines are the result of the work of experts from the two scientific societies and their aim is to provide clear, practical recommendations for the determination and management of elevated Lp(a) levels.
Collapse
Affiliation(s)
- Bożena Sosnowska
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
| | | | - Przemyslaw Mitkowski
- 1 Department of Cardiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Agata Bielecka-Dabrowa
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Department of Cardiology and Adult Congenital Defects, Polish Mother’s Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Beata Bobrowska
- Department of Clinical Cardiology and Cardiovascular Interventions, University Hospital in Krakow, Krakow, Poland
| | - Jan Budzianowski
- Department of Interventional Cardiology and Cardiac Surgery, University of Zielona Gora, Collegium Medicum, Zielona Gora, Poland
- Multidisciplinary Hospital, Nowa Sol, Poland
| | - Pawel Burchardt
- Department of Cardiology, J. Strus Hospital, Poznan, Poland
- Department of Hypertension, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Krzysztof Chlebus
- National Center for Familial Hypercholesterolemia, 1 Chair and Department of Cardiology, Medical University of Gdansk, Gdansk, Poland
| | - Piotr Dobrowolski
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, National Institute of Cardiology, Warsaw, Poland
| | - Mariusz Gasior
- 3 Department of Cardiology, Silesian Centre for Heart Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| | - Piotr Jankowski
- Department of Internal Medicine and Geriatric Cardiology, Medical Centre for Postgraduate Education, Warsaw, Poland
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Agnieszka Mickiewicz
- Lipoprotein Apheresis Laboratory, 1 Department of Cardiology, Medical University of Gdansk, Gdansk, Poland
| | - Malgorzata Mysliwiec
- Department of Paediatrics, Diabetology and Endocrinology, Medical University of Gdansk, Gdansk, Poland
| | - Tadeusz Osadnik
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Aleksander Prejbisz
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, National Institute of Cardiology, Warsaw, Poland
| | - Renata Rajtar-Salwa
- Department of Clinical Cardiology and Cardiovascular Interventions, University Hospital in Krakow, Krakow, Poland
| | - Kristian Wita
- 1 Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Adam Witkowski
- Department of Interventional Cardiology and Angiology, National Institute of Cardiology, Warsaw, Poland
| | - Robert Gil
- Department of Cardiology, National Medical Institute of the Ministry of Internal Affairs and Administration, Warsaw, Poland
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Department of Cardiology and Adult Congenital Defects, Polish Mother’s Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Pasławska A, Tomasik PJ. Lipoprotein(a)-60 Years Later-What Do We Know? Cells 2023; 12:2472. [PMID: 37887316 PMCID: PMC10605347 DOI: 10.3390/cells12202472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/17/2023] [Accepted: 10/14/2023] [Indexed: 10/28/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) molecule includes two protein components: apolipoprotein(a) and apoB100. The molecule is the main transporter of oxidized phospholipids (OxPL) in plasma. The concentration of this strongly atherogenic lipoprotein is predominantly regulated by the LPA gene expression. Lp(a) is regarded as a risk factor for several cardiovascular diseases. Numerous epidemiological, clinical and in vitro studies showed a strong association between increased Lp(a) and atherosclerotic cardiovascular disease (ASCVD), calcific aortic valve disease/aortic stenosis (CAVD/AS), stroke, heart failure or peripheral arterial disease (PAD). Although there are acknowledged contributions of Lp(a) to the mentioned diseases, clinicians struggle with many inconveniences such as a lack of well-established treatment lowering Lp(a), and common guidelines for diagnosing or assessing cardiovascular risk among both adult and pediatric patients. Lp(a) levels are different with regard to a particular race or ethnicity and might fluctuate during childhood. Furthermore, the lack of standardization of assays is an additional impediment. The review presents the recent knowledge on Lp(a) based on clinical and scientific research, but also highlights relevant aspects of future study directions that would approach more suitable and effective managing risk associated with increased Lp(a), as well as control the Lp(a) levels.
Collapse
Affiliation(s)
- Anna Pasławska
- Tuchow Health Center, Medical Hospital Laboratory, Szpitalna St. 1, 33-170 Tuchow, Poland;
| | - Przemysław J. Tomasik
- Department of Clinical Biochemistry, Pediatric Institute, College of Medicine, Jagiellonian University, Wielicka St. 265, 30-663 Cracow, Poland
| |
Collapse
|
13
|
Steg PG, Szarek M, Valgimigli M, Islam S, Zeiher AM, Bhatt DL, Bittner VA, Chiang CE, Diaz R, Goodman SG, Gotcheva N, Harrington RA, Jukema JW, Kim HS, Kim SH, Morais J, Pordy R, Scemama M, White HD, Schwartz GG. Lipoprotein(a) and the Effect of Alirocumab on Revascularization After Acute Coronary Syndrome. Can J Cardiol 2023; 39:1315-1324. [PMID: 37116789 DOI: 10.1016/j.cjca.2023.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Many patients require revascularization after index acute coronary syndrome (ACS). Lipoprotein(a) is thought to play a pathogenic role in atherothrombosis. In ODYSSEY OUTCOMES, alirocumab reduced major adverse cardiovascular events after ACS, with greater reduction among those with higher lipoprotein(a) levels. We explored whether risk of revascularization after ACS was modified by the level of lipoprotein(a) and treatment with alirocumab or placebo. METHODS In ODYSSEY OUTCOMES alirocumab was compared with placebo in 18,924 patients with ACS and elevated atherogenic lipoprotein levels despite optimized statin treatment. In this post hoc analysis, treatment effects are summarized using competing risks proportional hazard models. RESULTS A total of 1559 (8.2%) patients had coronary, 204 (1.1%) had limb, and 40 (0.2%) had carotid revascularization. Alirocumab reduced coronary revascularization (2.8 vs 3.2 events per 100 patient-years; hazard ratio [HR], 0.88 [95% confidence interval (CI), 0.80-0.97]; P = 0.01) and any revascularization (3.2 vs 3.7 events per 100 patient-years; HR, 0.85 [95% CI, 0.78-0.94]; P = 0.001). Baseline lipoprotein(a) quartile was directly associated with risk of coronary or any revascularization in the placebo arm and inversely related to treatment HRs (all P for trend < 0.01). Alirocumab produced the greatest reduction of coronary revascularization in patients with baseline lipoprotein(a) in the top quartile (≥ 59.6 mg/dL; HR, 0.69 [95% CI, 0.57-0.84]), but no apparent reduction in the bottom quartile (HR, 1.00 [95% CI, 0.82-1.22]). Findings were similar for the effect of alirocumab on any revascularization. CONCLUSIONS Alirocumab reduced revascularization rates after ACS. The risk of revascularization and reduction in that risk with alirocumab were greatest in patients with elevated lipoprotein(a) at baseline.
Collapse
Affiliation(s)
- P Gabriel Steg
- Department of Cardiology, Université Paris-Cité, INSERM-UMR1148, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, French Alliance for Cardiovascular Trials, and Institut Universitaire de France, Paris, France.
| | - Michael Szarek
- CPC Clinical Research and Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA; State University of New York, Downstate Health Sciences University, Brooklyn, New York, USA
| | - Marco Valgimigli
- Division of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland; Department of Cardiology,University of Bern, Bern, Switzerland
| | - Shahidul Islam
- Division of Health Services Research, NYU Long Island School of Medicine, Mineola, New York, USA
| | - Andreas M Zeiher
- Department of Medicine III, Goethe University, Frankfurt am Main, Germany
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Health System, New York, New York, USA
| | - Vera A Bittner
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chern-En Chiang
- General Clinical Research Center, Division of Cardiology, Taipei Veterans General Hospital and National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Rafael Diaz
- Estudios Cardiológicos Latinoamérica, Instituto Cardiovascular de Rosario, Rosario, Argentina
| | - Shaun G Goodman
- Department of Medicine, Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiology, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Nina Gotcheva
- Department of Cardiology, MHAT "National Cardiology Hospital" EAD, Sofia, Bulgaria
| | - Robert A Harrington
- Stanford Center for Clinical Research, Department of Medicine, Stanford University, Stanford, California, USA
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands; Netherlands Heart Institute, Utrecht, The Netherlands
| | - Hyo-Soo Kim
- Departmentof Cardiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sang-Hyun Kim
- Division of Cardiology, SMG Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Joao Morais
- Division of Cardiology, Leiria Hospital Center, Leiria, Portugal; ciTechCare, Polytechnic of Leiria, Leiria, Portugal
| | - Robert Pordy
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | - Harvey D White
- Green Lane Cardiovascular Services, Auckland City Hospital and Auckland University, Auckland, New Zealand
| | - Gregory G Schwartz
- Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
14
|
Fogacci F, Di Micoli V, Avagimyan A, Giovannini M, Imbalzano E, Cicero AFG. Assessment of Apolipoprotein(a) Isoform Size Using Phenotypic and Genotypic Methods. Int J Mol Sci 2023; 24:13886. [PMID: 37762189 PMCID: PMC10531419 DOI: 10.3390/ijms241813886] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Apolipoprotein(a) (apo(a)) is the protein component that defines lipoprotein(a) (Lp(a)) particles and is encoded by the LPA gene. The apo(a) is extremely heterogeneous in size due to the copy number variations in the kringle-IV type 2 (KIV2) domains. In this review, we aim to discuss the role of genetics in establishing Lp(a) as a risk factor for coronary heart disease (CHD) by examining a series of molecular biology techniques aimed at identifying the best strategy for a possible application in clinical research and practice, according to the current gold standard.
Collapse
Affiliation(s)
- Federica Fogacci
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.F.); (V.D.M.); (M.G.)
| | - Valentina Di Micoli
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.F.); (V.D.M.); (M.G.)
| | - Ashot Avagimyan
- Pathological Anatomy Department, Yerevan State Medical University, Yerevan 0025, Armenia;
| | - Marina Giovannini
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.F.); (V.D.M.); (M.G.)
| | - Egidio Imbalzano
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Arrigo F. G. Cicero
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.F.); (V.D.M.); (M.G.)
- Cardiovascular Medicine Unit, Heart, Thoracic and Vascular Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40100 Bologna, Italy
| |
Collapse
|
15
|
Kosmas CE, Bousvarou MD, Papakonstantinou EJ, Tsamoulis D, Koulopoulos A, Echavarria Uceta R, Guzman E, Rallidis LS. Novel Pharmacological Therapies for the Management of Hyperlipoproteinemia(a). Int J Mol Sci 2023; 24:13622. [PMID: 37686428 PMCID: PMC10487774 DOI: 10.3390/ijms241713622] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Lipoprotein(a) [Lp(a)] is a well-established risk factor for cardiovascular disease, predisposing to major cardiovascular events, including coronary heart disease, stroke, aortic valve calcification and abdominal aortic aneurysm. Lp(a) is differentiated from other lipoprotein molecules through apolipoprotein(a), which possesses atherogenic and antithrombolytic properties attributed to its structure. Lp(a) levels are mostly genetically predetermined and influenced by the size of LPA gene variants, with smaller isoforms resulting in a greater synthesis rate of apo(a) and, ultimately, elevated Lp(a) levels. As a result, serum Lp(a) levels may highly vary from extremely low to extremely high. Hyperlipoproteinemia(a) is defined as Lp(a) levels > 30 mg/dL in the US and >50 mg/dL in Europe. Because of its association with CVD, Lp(a) levels should be measured at least once a lifetime in adults. The ultimate goal is to identify individuals with increased risk of CVD and intervene accordingly. Traditional pharmacological interventions like niacin, statins, ezetimibe, aspirin, PCSK-9 inhibitors, mipomersen, estrogens and CETP inhibitors have not yet yielded satisfactory results. The mean Lp(a) reduction, if any, is barely 50% for all agents, with statins increasing Lp(a) levels, whereas a reduction of 80-90% appears to be required to achieve a significant decrease in major cardiovascular events. Novel RNA-interfering agents that specifically target hepatocytes are aimed in this direction. Pelacarsen is an antisense oligonucleotide, while olpasiran, LY3819469 and SLN360 are small interfering RNAs, all conjugated with a N-acetylgalactosamine molecule. Their ultimate objective is to genetically silence LPA, reduce apo(a) production and lower serum Lp(a) levels. Evidence thus so far demonstrates that monthly subcutaneous administration of a single dose yields optimal results with persisting substantial reductions in Lp(a) levels, potentially enhancing CVD risk reduction. The Lp(a) reduction achieved with novel RNA agents may exceed 95%. The results of ongoing and future clinical trials are eagerly anticipated, and it is hoped that guidelines for the tailored management of Lp(a) levels with these novel agents may not be far off.
Collapse
Affiliation(s)
- Constantine E. Kosmas
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Bronx, NY 10467, USA;
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY 10033, USA;
| | - Maria D. Bousvarou
- School of Medicine, University of Crete, 710 03 Heraklion, Greece; (M.D.B.); (A.K.)
| | | | - Donatos Tsamoulis
- First Department of Internal Medicine, Thriasio General Hospital of Eleusis, 196 00 Athens, Greece;
| | - Andreas Koulopoulos
- School of Medicine, University of Crete, 710 03 Heraklion, Greece; (M.D.B.); (A.K.)
| | | | - Eliscer Guzman
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Bronx, NY 10467, USA;
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY 10033, USA;
| | - Loukianos S. Rallidis
- 2nd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, University General Hospital ATTIKON, 124 62 Athens, Greece;
| |
Collapse
|
16
|
Michaeli DT, Michaeli JC, Albers S, Boch T, Michaeli T. Established and Emerging Lipid-Lowering Drugs for Primary and Secondary Cardiovascular Prevention. Am J Cardiovasc Drugs 2023; 23:477-495. [PMID: 37486464 PMCID: PMC10462544 DOI: 10.1007/s40256-023-00594-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/02/2023] [Indexed: 07/25/2023]
Abstract
Despite treatment with statins, patients with elevated low-density lipoprotein cholesterol (LDL-C) and triglycerides remain at increased risk for adverse cardiovascular events. Consequently, novel pharmaceutical drugs have been developed to control and modify the composition of blood lipids to ultimately prevent fatal cardiovascular events in patients with dyslipidaemia. This article reviews established and emerging lipid-lowering drugs regarding their mechanism of action, development stage, ongoing clinical trials, side effects, effect on blood lipids and reduction in cardiovascular morbidity and mortality. We conducted a keyword search to identify studies on established and emerging lipid modifying drugs. Results were summarized in a narrative overview. Established pharmaceutical treatment options include the Niemann-Pick-C1 like-1 protein (NPC1L1) inhibitor ezetimibe, the protein convertase subtilisin-kexin type 9 (PCSK9) inhibitors alirocumab and evolocumab, fibrates as peroxisome proliferator receptor alpha (PPAR-α) activators, and the omega-3 fatty acid icosapent ethyl. Statins are recommended as the first-line therapy for primary and secondary cardiovascular prevention in patients with hypercholesterinaemia and hypertriglyceridemia. For secondary prevention in hypercholesterinaemia, second-line options such as statin add-on or statin-intolerant treatments are ezetimibe, alirocumab and evolocumab. For secondary prevention in hypertriglyceridemia, second-line options such as statin add-on or statin-intolerant treatments are icosapent ethyl and fenofibrate. Robust data for these add-on therapeutics in primary cardiovascular prevention remains scarce. Recent biotechnological advances have led to the development of innovative small molecules (bempedoic acid, lomitapide, pemafibrate, docosapentaenoic and eicosapentaenoic acid), antibodies (evinacumab), antisense oligonucleotides (mipomersen, volanesorsen, pelcarsen, olezarsen), small interfering RNA (inclisiran, olpasiran), and gene therapies for patients with dyslipidemia. These molecules specifically target new cellular pathways, such as the adenosine triphosphate-citrate lyase (bempedoic acid), PCSK9 (inclisiran), angiopoietin-like 3 (ANGPTL3: evinacumab), microsomal triglyceride transfer protein (MTP: lomitapide), apolipoprotein B-100 (ApoB-100: mipomersen), apolipoprotein C-III (ApoC-III: volanesorsen, olezarsen), and lipoprotein (a) (Lp(a): pelcarsen, olpasiran). The authors are hopeful that the development of new treatment modalities alongside new therapeutic targets will further reduce patients' risk of adverse cardiovascular events. Apart from statins, data on new drugs' use in primary cardiovascular prevention remain scarce. For their swift adoption into clinical routine, these treatments must demonstrate safety and efficacy as well as cost-effectiveness in randomized cardiovascular outcome trials.
Collapse
Affiliation(s)
- Daniel Tobias Michaeli
- Department of Medical Oncology, National Center for Tumour Diseases, Heidelberg University Hospital, Heidelberg, Germany.
| | - Julia Caroline Michaeli
- Department of Obstetrics and Gynaecology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Albers
- Department of Orthopaedics and Sport Orthopaedics, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Boch
- Department of Medical Oncology, National Center for Tumour Diseases, Heidelberg University Hospital, Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Heidelberg University, Heidelberg, Germany
| | - Thomas Michaeli
- Department of Medical Oncology, National Center for Tumour Diseases, Heidelberg University Hospital, Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
17
|
Koutsogianni AD, Liamis G, Liberopoulos E, Adamidis PS, Florentin M. Effects of Lipid-Modifying and Other Drugs on Lipoprotein(a) Levels-Potent Clinical Implications. Pharmaceuticals (Basel) 2023; 16:ph16050750. [PMID: 37242533 DOI: 10.3390/ph16050750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The past few years have shown an ongoing interest in lipoprotein(a) (Lp(a)), a lipid molecule that has been proven to have atherogenic, thrombogenic, and inflammatory properties. Several lines of evidence, indeed, have demonstrated an increased risk of cardiovascular disease as well as calcific aortic valve stenosis in patients with elevated Lp(a) levels. Statins, the mainstay of lipid-lowering therapy, slightly increase Lp(a) levels, while most other lipid-modifying agents do not significantly alter Lp(a) concentrations, except for proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors. The latter have been shown to reduce Lp(a) levels; however, the clinical significance of this effect has not been clearly elucidated. Of note, the pharmaceutical lowering of Lp(a) may be achieved with novel treatments specifically designed for this purpose (i.e., antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs)). Large clinical trials with cardiovascular outcomes with these agents are ongoing, and their results are eagerly awaited. Furthermore, several non-lipid-modifying drugs of various classes may influence Lp(a) concentrations. We have searched MEDLINE, EMBASE, and CENTRAL databases up to 28 January 2023 and summarized the effects of established and emerging lipid-modifying drugs and other medications on Lp(a) levels. We also discuss the potent clinical implications of these alterations.
Collapse
Affiliation(s)
| | - George Liamis
- Department of Internal Medicine, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Evangelos Liberopoulos
- 1st Propaideutic Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece
| | | | - Matilda Florentin
- Department of Internal Medicine, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| |
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: 42] [Impact Index Per Article: 21.0] [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
|
Lampsas S, Xenou M, Oikonomou E, Pantelidis P, Lysandrou A, Sarantos S, Goliopoulou A, Kalogeras K, Tsigkou V, Kalpis A, Paschou SA, Theofilis P, Vavuranakis M, Tousoulis D, Siasos G. Lipoprotein(a) in Atherosclerotic Diseases: From Pathophysiology to Diagnosis and Treatment. Molecules 2023; 28:969. [PMID: 36770634 PMCID: PMC9918959 DOI: 10.3390/molecules28030969] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is a low-density lipoprotein (LDL) cholesterol-like particle bound to apolipoprotein(a). Increased Lp(a) levels are an independent, heritable causal risk factor for atherosclerotic cardiovascular disease (ASCVD) as they are largely determined by variations in the Lp(a) gene (LPA) locus encoding apo(a). Lp(a) is the preferential lipoprotein carrier for oxidized phospholipids (OxPL), and its role adversely affects vascular inflammation, atherosclerotic lesions, endothelial function and thrombogenicity, which pathophysiologically leads to cardiovascular (CV) events. Despite this crucial role of Lp(a), its measurement lacks a globally unified method, and, between different laboratories, results need standardization. Standard antilipidemic therapies, such as statins, fibrates and ezetimibe, have a mediocre effect on Lp(a) levels, although it is not yet clear whether such treatments can affect CV events and prognosis. This narrative review aims to summarize knowledge regarding the mechanisms mediating the effect of Lp(a) on inflammation, atherosclerosis and thrombosis and discuss current diagnostic and therapeutic potentials.
Collapse
Affiliation(s)
- Stamatios Lampsas
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Maria Xenou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Panteleimon Pantelidis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Antonios Lysandrou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Savvas Sarantos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Athina Goliopoulou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, 11527 Athens, Greece
| | - Vasiliki Tsigkou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Athanasios Kalpis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Stavroula A. Paschou
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, 11527 Athens, Greece
| | - Panagiotis Theofilis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, 11527 Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
20
|
Efficacy and safety of pelacarsen in lowering Lp(a) in healthy Japanese subjects. J Clin Lipidol 2023; 17:181-188. [PMID: 36529659 DOI: 10.1016/j.jacl.2022.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pelacarsen is a liver-targeted antisense oligonucleotide that potently lowers lipoprotein(a) [Lp(a)] levels. Its safety and efficacy in diverse populations has not been extensively studied. OBJECTIVE To assess the effect of pelacarsen, including monthly dosing of 80 mg, in subjects of Japanese ancestry. METHODS A randomized double-blind, placebo-controlled, study was performed in 29 healthy Japanese subjects treated with single ascending doses (SAD) of pelacarsen 20, 40 and 80 mg subcutaneously or multiple doses (MD) of pelacarsen 80 mg monthly for 4 doses. The primary objective was to assess the safety and tolerability in healthy Japanese subjects; secondary objectives to assess the pharmacokinetics of pelacarsen; and exploratory objective to determine the effect of pelacarsen on plasma Lp(a) levels. RESULTS No serious adverse events or clinically relevant abnormalities in any laboratory parameters were noted. In the MD cohort, mean plasma concentrations of pelacarsen peaked at ∼4 hours and declined in a bi-exponential manner thereafter. In the SAD cohorts, the placebo-corrected least-square mean (PCLSM) percent changes in Lp(a) at Day 30 were: -55.4% (p=0.0008), -58.9% (p=0.0003) and -73.7% (p<0.0001) for the 20 mg, 40 mg, and 80 mg pelacarsen-treated groups, respectively. In the MD cohort, the PCLSM at Days 29, 85, 113, 176 and 204 were -84.0% (p=0.0003), -106.2% (p<0.0001), -70.0 (p<0.0001), -80.0% (p=0.0104) and -55.8% (p=0.0707), respectively. CONCLUSIONS Pelacarsen demonstrates an acceptable safety and tolerability profile and potently lowers plasma levels of Lp(a) in healthy Japanese subjects, including with the 80 mg monthly dose being evaluated in the Lp(a) HORIZON trial.
Collapse
|
21
|
Luo Y, Peng D. Residual Atherosclerotic Cardiovascular Disease Risk: Focus on Non-High-Density Lipoprotein Cholesterol. J Cardiovasc Pharmacol Ther 2023; 28:10742484231189597. [PMID: 37641208 DOI: 10.1177/10742484231189597] [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: 08/31/2023]
Abstract
Cardiovascular disease (CVD) caused by atherosclerosis is the leading cause of death worldwide. The level of low-density lipoprotein cholesterol (LDL-C), considered as the initiator of atherosclerosis, is the most widely used predictor for CVD risk and LDL-C has been the primary target for lipid-lowering therapies. However, residual CVD risk remains high even with very low levels of LDL-C. This residual CVD risk may be due to remnant cholesterol, high triglyceride levels, and low high-density lipoprotein cholesterol (HDL-C). Non-high density lipoprotein cholesterol (non-HDL-C), which is calculated as total cholesterol minus HDL-C (and represents the cholesterol content of all atherogenic apolipoprotein B-containing lipoproteins), has emerged as a better risk predictor for CVD than LDL-C and an alternative target for CVD risk reduction. Major international guidelines recommend evaluating non-HDL-C as part of atherosclerotic CVD risk assessment, especially in people with high triglycerides, diabetes, obesity, or very low LDL-C. A non-HDL-C target of <130 mg/dL (3.4 mmol/L) has been recommended for patients at very high risk, which is 30 mg/dL (0.8 mmol/L) higher than the corresponding LDL-C target goal. Non-HDL-C lowering approaches include reducing LDL-C and triglyceride levels, increasing HDL-C, or targeting multiple risk factors simultaneously. However, despite the growing evidence for the role of non-HDL-C in residual CVD risk, and recommendations for its assessment in major guidelines, non-HDL-C testing is not routinely done in clinical practice. Thus, there is a need for increased awareness of the need for non-HDL-C testing for ascertaining CVD risk and concomitant prevention of CVD.
Collapse
Affiliation(s)
- Yonghong Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
22
|
Di Fusco SA, Arca M, Scicchitano P, Alonzo A, Perone F, Gulizia MM, Gabrielli D, Oliva F, Imperoli G, Colivicchi F. Lipoprotein(a): a risk factor for atherosclerosis and an emerging therapeutic target. Heart 2022; 109:18-25. [PMID: 35288443 DOI: 10.1136/heartjnl-2021-320708] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is a complex circulating lipoprotein, and increasing evidence has demonstrated its role as a risk factor for atherosclerotic cardiovascular disease (ASCVD) and as a possible therapeutic target. Lp(a) atherogenic effects are attributed to several potential mechanisms in addition to cholesterol accumulation in the arterial wall, including proinflammatory effects mainly mediated by oxidised phospholipids. Several studies have found a causal and independent relationship between Lp(a) levels and cardiovascular risk. Furthermore, several studies also suggest a causal association between Lp(a) levels and calcific aortic valve stenosis. Available lipid-lowering agents have at best moderate impact on Lp(a) levels. Among available therapies, antibody proprotein convertase subtilisin/kexin type 9 inhibitors are the most effective in reducing Lp(a). Potent Lp(a)-lowering treatments that target LPA expression are under development. Lp(a) level measurement poses some challenges due to the absence of a definitive reference method and the reporting of Lp(a) values as molar (nanomoles per litre (nmol/L)) or mass concentrations (milligrams per decilitre (mg/dL)) by different assays. Currently, Lp(a) measurement is recommended to refine cardiovascular risk in specific clinical settings, that is, in individuals with a family history of premature ASCVD, in patients with ASCVD not explained by standard risk factors or in those with recurrent events despite optimal management of traditional risk factors. Patients with high Lp(a) levels should be managed with more intensive approaches to treat other modifiable cardiovascular risk factors. Overall, this review focuses on Lp(a) as an ASCVD risk factor and therapeutic target. Furthermore, it reports practical recommendations for Lp(a) measurement and interpretation and updated evidence on Lp(a)-lowering approaches.
Collapse
Affiliation(s)
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | | | | | | | - Michele Massimo Gulizia
- Cardiology Division, Garibaldi-Nesima Hospital, Garibaldi-Nesima Hospital, Catania, Italy
- Heart Care Foundation, Florence, Italy
| | | | - Fabrizio Oliva
- De Gasperis Cardio Center, Niguarda Hospital, Milan, Italy
| | | | | |
Collapse
|
23
|
Li T, Yuan D, Wang P, Jia S, Zhang C, Zhu P, Song Y, Tang X, Zhao X, Gao Z, Yang Y, Gao R, Xu B, Yuan J. Associations of lipid measures with total occlusion in patients with established coronary artery disease: a cross-sectional study. Lipids Health Dis 2022; 21:118. [DOI: 10.1186/s12944-022-01733-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Total occlusion is the most severe coronary lesion, indicating heavy ischemic burden and poor prognosis. The lipid profile is central to the development of atherosclerotic coronary lesions. Evidence on the optimal lipid measure to be monitored and managed in patients with established coronary artery disease (CAD) is inconclusive.
Methods
Total cholesterol (TC), total triglyceride (TG), low-density lipoprotein cholesterol (LDL-c), nonhigh-density lipoprotein cholesterol (non-HDL-c), lipoprotein (a) [Lp(a)], apolipoprotein B (apoB), non-HDL-c/HDL-c, and apoB/apoA-1 were analyzed in quintiles and as continuous variables. The associations of lipid measures with total occlusion were tested using logistic regression models, visualized with restricted cubic splines, and compared by areas under the receiver operating characteristic curves (AUROC). Discordance analysis was performed when apoB/apoA-1 and non-HDL-c/HDL-c were not in concordance.
Results
The prospective cohort study included 10,003 patients (mean age: 58 years; women: 22.96%), with 1879 patients having total occlusion. The risks of total occlusion significantly increased with quintiles of Lp(a), non-HDL-c/HDL-c, and apoB/apoA-1 (all p for trend < 0.001). TG had no association with total occlusion. Restricted cubic splines indicate significant positive linear relations between the two ratios and total occlusion [odds ratio per 1-standard deviation increase (95% confidence interval): non-HDL-c/HDL-c: 1.135 (1.095–1.176), p < 0.001; apoB/apoA-1: 2.590 (2.049–3.274), p < 0.001]. The AUROCs of apoB/apoA-1 and non-HDL-c/HDL-c were significantly greater than those of single lipid measures. Elevation in the apoB/apoA-1 tertile significantly increased the risk of total occlusion at a given non-HDL-c/HDL-c tertile but not vice versa.
Conclusion
ApoB/apoA-1 confers better predictive power for total occlusion than non-HDL-c/HDL-c and single lipid measures in established CAD patients.
Collapse
|
24
|
Yanai H, Adachi H, Hakoshima M, Katsuyama H. Atherogenic Lipoproteins for the Statin Residual Cardiovascular Disease Risk. Int J Mol Sci 2022; 23:ijms232113499. [PMID: 36362288 PMCID: PMC9657259 DOI: 10.3390/ijms232113499] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Randomized controlled trials (RCTs) show that decreases in low-density lipoprotein cholesterol (LDL-C) by the use of statins cause a significant reduction in the development of cardiovascular disease (CVD). However, one of our previous studies showed that, among eight RCTs that investigated the effect of statins vs. a placebo on CVD development, 56–79% of patients had residual CVD risk after the trials. In three RCTs that investigated the effect of a high dose vs. a usual dose of statins on CVD development, 78–87% of patients in the high-dose statin arms still had residual CVD risk. The risk of CVD development remains even when statins are used to strongly reduce LDL-C, and this type of risk is now regarded as statin residual CVD risk. Our study shows that elevated triglyceride (TG) levels, reduced high-density lipoprotein cholesterol (HDL-C), and the existence of obesity/insulin resistance and diabetes may be important metabolic factors that determine statin residual CVD risk. Here, we discuss atherogenic lipoproteins that were not investigated in such RCTs, such as lipoprotein (a) (Lp(a)), remnant lipoproteins, malondialdehyde-modified LDL (MDA-LDL), and small-dense LDL (Sd-LDL). Lp(a) is under strong genetic control by apolipoprotein (a), which is an LPA gene locus. Variations in the LPA gene account for 91% of the variability in the plasma concentration of Lp(a). A meta-analysis showed that genetic variations at the LPA locus are associated with CVD events during statin therapy, independent of the extent of LDL lowering, providing support for exploring strategies targeting circulating concentrations of Lp(a) to reduce CVD events in patients receiving statins. Remnant lipoproteins and small-dense LDL are highly associated with high TG levels, low HDL-C, and obesity/insulin resistance. MDA-LDL is a representative form of oxidized LDL and plays important roles in the formation and development of the primary lesions of atherosclerosis. MDA-LDL levels were higher in CVD patients and diabetic patients than in the control subjects. Furthermore, we demonstrated the atherogenic properties of such lipoproteins and their association with CVD as well as therapeutic approaches.
Collapse
Affiliation(s)
- Hidekatsu Yanai
- Correspondence: ; Tel.: +81-473-72-3501; Fax: +81-473-72-1858
| | | | | | | |
Collapse
|
25
|
Esau D, Abramson BL. Approach to risk stratification of atherosclerotic cardiovascular disease: Use of biomarkers and imaging in a Canadian context. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2022; 68:654-660. [PMID: 36100373 PMCID: PMC9470181 DOI: 10.46747/cfp.6809654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To outline the 2021 Canadian Cardiovascular Society (CCS) dyslipidemia guidelines and to present the current approaches to cardiovascular risk stratification, including the incorporation of biomarkers and imaging tests. SOURCES OF INFORMATION Current guidelines were reviewed and an Ovid MEDLINE literature search was performed. MAIN MESSAGE Cardiovascular disease (CVD) is the leading cause of global mortality, with ischemic heart disease contributing to nearly half of these deaths. Risk stratification is undertaken to identify patients who would benefit from primary prevention for atherosclerotic CVD (ASCVD), but commonly used methods for risk stratification are imperfect. The CCS guidelines endorse that the presence of risk modifiers (family history of premature ASCVD, high-sensitivity C-reactive protein level ≥2.0 mg/L, lipoprotein[a] level ≥500 mg/L [≥50 mg/dL], or coronary artery calcium >0) supports the use of statin therapy in those at intermediate risk (Framingham risk score 10% to 19.9%) who do not otherwise meet the recommendations for statin use. The CCS guidelines recommend statin therapy in patients at intermediate risk when cholesterol levels are elevated (low-density lipoprotein cholesterol level ≥3.5 mmol/L, non-high-density lipoprotein cholesterol level ≥4.2 mmol/L, or apolipoprotein B level ≥1.05 g/L). In addition, statin therapy should be considered for patients at low risk (Framingham risk score 5% to 9.9%) with elevated cholesterol levels, especially if risk modifiers are present. When cholesterol levels are not elevated, evidence still favours the use of statins in intermediate-risk patients when risk modifiers are present and in men 50 years and older and women 60 years and older with 1 additional risk factor. CONCLUSION Biomarkers and imaging tests have the potential to improve ASCVD risk stratification by reclassifying any patient whose risk has been inaccurately estimated by traditional methods. Recently published guidelines by the CCS suggest the use of biomarkers and imaging in certain patient groups.
Collapse
Affiliation(s)
- Daniel Esau
- Recently completed a fellowship in ambulatory and preventive cardiology in the Division of Cardiology at St Michael's Hospital in Toronto, Ont, and is now working in Victoria, BC
| | - Beth L Abramson
- Paul Albrechtsen Professor in Cardiac Prevention and Women's Health in the Division of Cardiology at St Michael's Hospital and Associate Professor of Medicine at the University of Toronto.
| |
Collapse
|
26
|
Esau D, Abramson BL. Approche de la stratification du risque de maladies cardiovasculaires athéroscléreuses. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2022; 68:e256-e263. [PMID: 36100384 PMCID: PMC9470202 DOI: 10.46747/cfp.6809e256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Objectif Donner un aperçu des lignes directrices de la Société canadienne de cardiologie (SCC) de 2021 sur la dyslipidémie et présenter les approches actuelles relatives à la stratification du risque cardiovasculaire, y compris l’intégration de la mesure des biomarqueurs et de l’imagerie. Sources de l’information Les lignes directrices actuelles ont été passées en revue, et une recherche documentaire dans Ovid MEDLINE a été effectuée. Message principal Les maladies cardiovasculaires (MCV) sont la principale cause de mortalité dans le monde, et les cardiopathies ischémiques contribuent à près de la moitié de ces décès. Une stratification du risque est entreprise pour identifier les patients susceptibles de bénéficier d’une prévention primaire de la MCV athéroscléreuse (MCVAS), mais les méthodes habituellement utilisées pour la stratification du risque sont imparfaites. Les lignes directrices de la SCC soutiennent que la présence de modificateurs du risque (antécédents familiaux de MCVAS prématurée, dosage de la protéine C réactive hypersensible ≥2,0 mg/L, dosage de la lipoprotéine [a] ≥500 mg/L [≥50 mg/dL] ou score calcique coronarien >0) justifie le recours à une thérapie aux statines chez les personnes à risque moyen (score de risque de Framingham de 10 à 19,9 %) qui ne sont pas autrement visées par les recommandations en faveur de l’utilisation de statines. Les lignes directrices de la SCC recommandent une thérapie aux statines chez les patients à risque modéré, lorsque leurs taux de cholestérol sont élevés (taux de cholestérol à lipoprotéines de basse densité ≥3,5 mmol/L, taux de cholestérol lié aux lipoprotéines autres que celles de haute densité ≥4,2 mmol/L ou taux d’apolipoprotéines B ≥1,05 g/L). De plus, une thérapie aux statines devrait être envisagée pour les patients à faible risque (score de risque de Framingham de 5 à 9,9 %) dont les taux de cholestérol sont élevés, surtout en présence de modificateurs du risque. Lorsque les taux de cholestérol ne sont pas élevés, des données probantes favorisent quand même le recours aux statines chez les patients à risque modéré lorsque des modificateurs du risque sont présents, de même que chez les hommes de 50 ans et plus et chez les femmes de 60 ans et plus ayant 1 facteur de risque additionnel. Conclusion La mesure des biomarqueurs et l’imagerie ont le potentiel d’améliorer la stratification du risque de MCVAS en reclassant les patients dont le risque avait été estimé de manière inexacte par les méthodes traditionnelles. Les lignes directrices récemment publiées par la SCC suggèrent de se servir des biomarqueurs et de l’imagerie chez certains groupes de patients.
Collapse
Affiliation(s)
- Daniel Esau
- Récemment terminé une bourse d'études en cardiologie ambulatoire et préventive à la Division de cardiologie de l'Hôpital St Michael's à Toronto (Ontario) et travaille maintenant à Victoria (C.-B.)
| | - Beth L Abramson
- Professeure Paul-Albrechtsen en prévention cardiaque et en santé des femmes à la Division de cardiologie de l'Hôpital St Michael's et professeure agrégée de médecine à l'Université de Toronto.
| |
Collapse
|
27
|
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
|
28
|
Wilson DP, Jacobson TA, Jones PH, Koschinsky ML, McNeal CJ, Nordestgaard BG, Orringer CE. Use of Lipoprotein(a) in clinical practice: A biomarker whose time has come. A scientific statement from the National Lipid Association. J Clin Lipidol 2022; 16:e77-e95. [PMID: 36068139 DOI: 10.1016/j.jacl.2022.08.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Lipoprotein(a) [Lp(a)] is a well-recognized, independent risk factor for atherosclerotic cardiovascular disease, with elevated levels estimated to be prevalent in 20% of the population. Observational and genetic evidence strongly support a causal relationship between high plasma concentrations of Lp(a) and increased risk of atherosclerotic cardiovascular disease-related events, such as myocardial infarction and stroke, and valvular aortic stenosis. In this scientific statement, we review an array of evidence-based considerations for testing of Lp(a) in clinical practice and the utilization of Lp(a) levels to inform treatment strategies in primary and secondary prevention.
Collapse
Affiliation(s)
- Don P Wilson
- Department of Pediatric Endocrinology and Diabetes, Cook Children''s Medical Center, Fort Worth, TX, USA.
| | - Terry A Jacobson
- Department of Medicine, Lipid Clinic and Cardiovascular Risk Reduction Program, Emory University, Atlanta, GA, USA
| | - Peter H Jones
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Catherine J McNeal
- Division of Cardiology, Department of Internal Medicine, Baylor Scott & White Health, Temple, TX, USA
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Carl E Orringer
- Division of Cardiology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
29
|
Duan Y, Gong K, Xu S, Zhang F, Meng X, Han J. Regulation of cholesterol homeostasis in health and diseases: from mechanisms to targeted therapeutics. Signal Transduct Target Ther 2022; 7:265. [PMID: 35918332 PMCID: PMC9344793 DOI: 10.1038/s41392-022-01125-5] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
Disturbed cholesterol homeostasis plays critical roles in the development of multiple diseases, such as cardiovascular diseases (CVD), neurodegenerative diseases and cancers, particularly the CVD in which the accumulation of lipids (mainly the cholesteryl esters) within macrophage/foam cells underneath the endothelial layer drives the formation of atherosclerotic lesions eventually. More and more studies have shown that lowering cholesterol level, especially low-density lipoprotein cholesterol level, protects cardiovascular system and prevents cardiovascular events effectively. Maintaining cholesterol homeostasis is determined by cholesterol biosynthesis, uptake, efflux, transport, storage, utilization, and/or excretion. All the processes should be precisely controlled by the multiple regulatory pathways. Based on the regulation of cholesterol homeostasis, many interventions have been developed to lower cholesterol by inhibiting cholesterol biosynthesis and uptake or enhancing cholesterol utilization and excretion. Herein, we summarize the historical review and research events, the current understandings of the molecular pathways playing key roles in regulating cholesterol homeostasis, and the cholesterol-lowering interventions in clinics or in preclinical studies as well as new cholesterol-lowering targets and their clinical advances. More importantly, we review and discuss the benefits of those interventions for the treatment of multiple diseases including atherosclerotic cardiovascular diseases, obesity, diabetes, nonalcoholic fatty liver disease, cancer, neurodegenerative diseases, osteoporosis and virus infection.
Collapse
Affiliation(s)
- Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Suowen Xu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Feng Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xianshe Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China. .,College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.
| |
Collapse
|
30
|
Patel N, Mittal N, Choubdar PA, Taub PR. Lipoprotein(a)—When to Screen and How to Treat. CURRENT CARDIOVASCULAR RISK REPORTS 2022. [DOI: 10.1007/s12170-022-00698-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
31
|
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]
|
32
|
Lipoprotein(a) Serum Levels Predict Pulse Wave Velocity in Subjects in Primary Prevention for Cardiovascular Disease with Large Apo(a) Isoforms: Data from the Brisighella Heart Study. Biomedicines 2022; 10:biomedicines10030656. [PMID: 35327457 PMCID: PMC8945189 DOI: 10.3390/biomedicines10030656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 11/17/2022] Open
Abstract
In the last decades, high serum levels of lipoprotein(a) (Lp(a)) have been associated with increased cardiovascular disease (CVD) risk, in particular among individuals with smaller apolipoprotein(a) (apo(a)) isoforms than those with larger sizes. The aim of our analysis was to evaluate whether Lp(a) levels could predict early vascular aging, and whether smaller apo(a) isoforms had a predictive value for vascular aging different than larger apo(a) isoforms in a cohort of subjects free from CVD. We considered the data of a subset of Brisighella Heart Study (BHS) participants free from CVD (462 men and 516 women) who were clinically evaluated during the 2012 BHS population survey. Predictors of arterial stiffness, measured as carotid-femoral pulse wave velocity (cfPWV) were estimated by the application of a step-wise linear regression model. In our cohort, there were 511 subjects with small apo(a) size and 467 subjects with large apo(a) isoforms. Subjects with larger apo(a) isoform sizes had significantly lower serum levels of Lp(a). In the BHS subpopulation sample, cfPWV was predicted by age, systolic blood pressure (SBP), serum levels of high-density lipoprotein cholesterol (HDL-C), triglycerides (TG) and sex, higher HDL-C serum levels and female sex associated with lower values of cfPWV. In subjects with smaller apo(a) isoform sizes, predictors of cfPWV were age, SBP, sex and serum levels of HDL-C, being higher HDL-C serum levels and female sex associated to lower values of cfPWV. In subjects with larger apo(a) isoform sizes, cfPWV was predicted by age, SBP, serum levels of Lp(a) and sex, with female sex associated with lower values of cfPWV. In our subpopulation sample, Lp(a) did not predict cfPWV. However, in subjects with large apo(a) isoform sizes, Lp(a) was a significant predictor of arterial stiffness.
Collapse
|
33
|
de Boer LM, Hof MH, Wiegman A, Stroobants AK, Kastelein JJ, Hutten BA. Lipoprotein(a) levels from childhood to adulthood: Data in nearly 3,000 children who visited a pediatric lipid clinic. Atherosclerosis 2022; 349:227-232. [DOI: 10.1016/j.atherosclerosis.2022.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/14/2022] [Accepted: 03/01/2022] [Indexed: 02/02/2023]
|
34
|
Durlach V, Bonnefont-Rousselot D, Boccara F, Varret M, Di-Filippo Charcosset M, Cariou B, Valero R, Charriere S, Farnier M, Morange PE, Meilhac O, Lambert G, Moulin P, Gillery P, Beliard-Lasserre S, Bruckert E, Carrié A, Ferrières J, Collet X, Chapman MJ, Anglés-Cano E. Lipoprotein(a): Pathophysiology, measurement, indication and treatment in cardiovascular disease. A consensus statement from the Nouvelle Société Francophone d'Athérosclérose (NSFA). Arch Cardiovasc Dis 2021; 114:828-847. [PMID: 34840125 DOI: 10.1016/j.acvd.2021.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 10/19/2022]
Abstract
Lipoprotein(a) is an apolipoprotein B100-containing low-density lipoprotein-like particle that is rich in cholesterol, and is associated with a second major protein, apolipoprotein(a). Apolipoprotein(a) possesses structural similarity to plasminogen but lacks fibrinolytic activity. As a consequence of its composite structure, lipoprotein(a) may: (1) elicit a prothrombotic/antifibrinolytic action favouring clot stability; and (2) enhance atherosclerosis progression via its propensity for retention in the arterial intima, with deposition of its cholesterol load at sites of plaque formation. Equally, lipoprotein(a) may induce inflammation and calcification in the aortic leaflet valve interstitium, leading to calcific aortic valve stenosis. Experimental, epidemiological and genetic evidence support the contention that elevated concentrations of lipoprotein(a) are causally related to atherothrombotic risk and equally to calcific aortic valve stenosis. The plasma concentration of lipoprotein(a) is principally determined by genetic factors, is not influenced by dietary habits, remains essentially constant over the lifetime of a given individual and is the most powerful variable for prediction of lipoprotein(a)-associated cardiovascular risk. However, major interindividual variations (up to 1000-fold) are characteristic of lipoprotein(a) concentrations. In this context, lipoprotein(a) assays, although currently insufficiently standardized, are of considerable interest, not only in stratifying cardiovascular risk, but equally in the clinical follow-up of patients treated with novel lipid-lowering therapies targeted at lipoprotein(a) (e.g. antiapolipoprotein(a) antisense oligonucleotides and small interfering ribonucleic acids) that markedly reduce circulating lipoprotein(a) concentrations. We recommend that lipoprotein(a) be measured once in subjects at high cardiovascular risk with premature coronary heart disease, in familial hypercholesterolaemia, in those with a family history of coronary heart disease and in those with recurrent coronary heart disease despite lipid-lowering treatment. Because of its clinical relevance, the cost of lipoprotein(a) testing should be covered by social security and health authorities.
Collapse
Affiliation(s)
- Vincent Durlach
- Champagne-Ardenne University, UMR CNRS 7369 MEDyC & Cardio-Thoracic Department, Reims University Hospital, 51092 Reims, France
| | - Dominique Bonnefont-Rousselot
- Metabolic Biochemistry Department, Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France; Université de Paris, CNRS, INSERM, UTCBS, 75006 Paris, France
| | - Franck Boccara
- Sorbonne University, GRC n(o) 22, C(2)MV, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, IHU ICAN, 75012 Paris, France; Service de Cardiologie, Hôpital Saint-Antoine, AP-HP, 75012 Paris, France
| | - Mathilde Varret
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalier Universitaire Xavier Bichat, 75018 Paris, France; Université de Paris, 75018 Paris, France
| | - Mathilde Di-Filippo Charcosset
- Hospices Civils de Lyon, UF Dyslipidémies, 69677 Bron, France; Laboratoire CarMen, INSERM, INRA, INSA, Université Claude-Bernard Lyon 1, 69495 Pierre-Bénite, France
| | - Bertrand Cariou
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'Institut du Thorax, 44000 Nantes, France
| | - René Valero
- Endocrinology Department, La Conception Hospital, AP-HM, Aix-Marseille University, INSERM, INRAE, C2VN, 13005 Marseille, France
| | - Sybil Charriere
- Hospices Civils de Lyon, INSERM U1060, Laboratoire CarMeN, Université Lyon 1, 69310 Pierre-Bénite, France
| | - Michel Farnier
- PEC2, EA 7460, University of Bourgogne Franche-Comté, 21079 Dijon, France; Department of Cardiology, CHU Dijon Bourgogne, 21000 Dijon, France
| | - Pierre E Morange
- Aix-Marseille University, INSERM, INRAE, C2VN, 13385 Marseille, France
| | - Olivier Meilhac
- INSERM, UMR 1188 DéTROI, Université de La Réunion, 97744 Saint-Denis de La Réunion, Reunion; CHU de La Réunion, CIC-EC 1410, 97448 Saint-Pierre, Reunion
| | - Gilles Lambert
- INSERM, UMR 1188 DéTROI, Université de La Réunion, 97744 Saint-Denis de La Réunion, Reunion; CHU de La Réunion, CIC-EC 1410, 97448 Saint-Pierre, Reunion
| | - Philippe Moulin
- Hospices Civils de Lyon, INSERM U1060, Laboratoire CarMeN, Université Lyon 1, 69310 Pierre-Bénite, France
| | - Philippe Gillery
- Laboratory of Biochemistry-Pharmacology-Toxicology, Reims University Hospital, University of Reims Champagne-Ardenne, UMR CNRS/URCA n(o) 7369, 51092 Reims, France
| | - Sophie Beliard-Lasserre
- Endocrinology Department, La Conception Hospital, AP-HM, Aix-Marseille University, INSERM, INRAE, C2VN, 13005 Marseille, France
| | - Eric Bruckert
- Service d'Endocrinologie-Métabolisme, Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France; IHU ICAN, Sorbonne University, 75013 Paris, France
| | - Alain Carrié
- Sorbonne University, UMR INSERM 1166, IHU ICAN, Laboratory of Endocrine and Oncological Biochemistry, Obesity and Dyslipidaemia Genetic Unit, Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France
| | - Jean Ferrières
- Department of Cardiology and INSERM UMR 1295, Rangueil University Hospital, TSA 50032, 31059 Toulouse, France
| | - Xavier Collet
- INSERM U1048, Institute of Metabolic and Cardiovascular Diseases, Rangueil University Hospital, BP 84225, 31432 Toulouse, France
| | - M John Chapman
- Sorbonne University, Hôpital Pitié-Salpêtrière and National Institute for Health and Medical Research (INSERM), 75013 Paris, France
| | - Eduardo Anglés-Cano
- Université de Paris, INSERM, Innovative Therapies in Haemostasis, 75006 Paris, France.
| |
Collapse
|
35
|
Ruscica M, Sirtori CR, Corsini A, Watts GF, Sahebkar A. Lipoprotein(a): Knowns, unknowns and uncertainties. Pharmacol Res 2021; 173:105812. [PMID: 34450317 DOI: 10.1016/j.phrs.2021.105812] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
Over the last 10 years, there have been advances on several aspects of lipoprotein(a) which are reviewed in the present article. Since the standard immunoassays for measuring lipoprotein(a) are not fully apo(a) isoform-insensitive, the application of an LC-MS/MS method for assaying molar concentrations of lipoprotein(a) has been advocated. Genome wide association, epidemiological, and clinical studies have established high lipoprotein(a) as a causal risk factor for atherosclerotic cardiovascular diseases (ASCVD). However, the relative importance of molar concentration, apo(a) isoform size or variants within the LPA gene is still controversial. Lipoprotein(a)-raising single nucleotide polymorphisms has not been shown to add on value in predicting ASCVD beyond lipoprotein(a) concentrations. Although hyperlipoproteinemia(a) represents an important confounder in the diagnosis of familial hypercholesterolemia (FH), it enhances the risk of ASCVD in these patients. Thus, identification of new cases of hyperlipoproteinemia(a) during cascade testing can increase the identification of high-risk individuals. However, it remains unclear whether FH itself increases lipoprotein(a). The ASCVD risk associated with lipoprotein(a) seems to follow a linear gradient across the distribution, regardless of racial subgroups and other risk factors. The inverse association with the risk of developing type 2 diabetes needs consideration as effective lipoprotein(a) lowering therapies are progressing towards the market. Considering that Mendelian randomization analyses have identified the degree of lipoprotein(a)-lowering that is required to achieve ASCVD benefit, the findings of the ongoing outcome trial with pelacarsen will clarify whether dramatically lowering lipoprotein(a) levels can reduce the risk of ASCVD.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy.
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Cardiometabolic Services, Department of Cardiology, Royal Perth Hospital, Australia
| | - Amirhossein Sahebkar
- School of Medicine, University of Western Australia, Perth, Australia; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
36
|
Lipoprotein (a) and Cardiovascular Disease: A Missing Link for Premature Atherosclerotic Heart Disease and/or Residual Risk. J Cardiovasc Pharmacol 2021; 79:e18-e35. [PMID: 34694242 DOI: 10.1097/fjc.0000000000001160] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/30/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Lipoprotein(a) or lipoprotein "little a" is an under-recognized causal risk factor for cardiovascular (CV) disease (CVD), including coronary atherosclerosis, aortic valvular stenosis, ischemic stroke, heart failure and peripheral arterial disease. Elevated plasma Lp(a) (≥50 mg/dL or ≥100 nmol/L) is commonly encountered in almost 1 in 5 individuals and confers a higher CV risk compared to those with normal Lp(a) levels, although such normal levels have not been generally agreed upon. Elevated Lp(a) is considered a cause of premature and accelerated atherosclerotic CVD. Thus, in patients with a positive family or personal history of premature coronary artery disease (CAD), Lp(a) should be measured. However, elevated Lp(a) may confer increased risk for incident CAD even in the absence of a family history of CAD, and even in those who have guideline-lowered LDL-cholesterol (<70 mg/dl) and continue to have a persisting CV residual risk. Thus, measurement of Lp(a) will have a significant clinical impact on the assessment of atherosclerotic CVD risk, and will assume a more important role in managing patients with CVD with the advent and clinical application of specific Lp(a)-lowering therapies. Conventional therapeutic approaches like lifestyle modification and statin therapy remain ineffective at lowering Lp(a). Newer treatment modalities, such as gene silencing via RNA interference with use of antisense oligonucleotide(s) or small interfering RNA molecules targeting Lp(a) seem very promising. These issues are herein reviewed, accumulated data are scrutinized, meta-analyses and current guidelines are tabulated and Lp(a)-related CVDs and newer therapeutic modalities are pictorially illustrated.
Collapse
|
37
|
Handhle A, Viljoen A, Wierzbicki AS. Elevated Lipoprotein(a): Background, Current Insights and Future Potential Therapies. Vasc Health Risk Manag 2021; 17:527-542. [PMID: 34526771 PMCID: PMC8436116 DOI: 10.2147/vhrm.s266244] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
Lipoprotein(a) forms a subfraction of the lipid profile and is characterized by the addition of apolipprotein(a) (apo(a)) to apoB100 derived particles. Its levels are mostly genetically determined inversely related to the number of protein domain (kringle) repeats in apo(a). In epidemiological studies, it shows consistent association with cardiovascular disease (CVD) and most recently with extent of aortic stenosis. Issues with standardizing the measurement of Lp(a) are being resolved and consensus statements favor its measurement in patients at high risk of, or with family histories of CVD events. Major lipid-lowering therapies such as statin, fibrates, and ezetimibe have little effect on Lp(a) levels. Therapies such as niacin or cholesterol ester transfer protein (CETP) inhibitors lower Lp(a) as well as reducing other lipid-related risk factors but have failed to clearly reduce CVD events. Proprotein convertase subtilisin kexin-9 (PCSK9) inhibitors reduce cholesterol and Lp(a) as well as reducing CVD events. New antisense therapies specifically targeting apo(a) and hence Lp(a) have greater and more specific effects and will help clarify the extent to which intervention in Lp(a) levels will reduce CVD events.
Collapse
Affiliation(s)
- Ahmed Handhle
- Department of Metabolic Medicine/Chemical Pathology, Addenbrookes Hospital, Cambridge, UK
| | - Adie Viljoen
- Department of Metabolic Medicine/Chemical Pathology, North & East Hertfordshire Hospitals Trust, Lister Hospital, Hertfordshire, UK
| | - Anthony S Wierzbicki
- Department of Metabolic Medicine/Chemical Pathology, Guy's & St Thomas', Hospitals, London, SE1 7EH, UK
| |
Collapse
|
38
|
Noels H, Lehrke M, Vanholder R, Jankowski J. Lipoproteins and fatty acids in chronic kidney disease: molecular and metabolic alterations. Nat Rev Nephrol 2021; 17:528-542. [PMID: 33972752 DOI: 10.1038/s41581-021-00423-5] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) induces modifications in lipid and lipoprotein metabolism and homeostasis. These modifications can promote, modulate and/or accelerate CKD and secondary cardiovascular disease (CVD). Lipid and lipoprotein abnormalities - involving triglyceride-rich lipoproteins, LDL and/or HDL - not only involve changes in concentration but also changes in molecular structure, including protein composition, incorporation of small molecules and post-translational modifications. These alterations modify the function of lipoproteins and can trigger pro-inflammatory and pro-atherogenic processes, as well as oxidative stress. Serum fatty acid levels are also often altered in patients with CKD and lead to changes in fatty acid metabolism - a key process in intracellular energy production - that induce mitochondrial dysfunction and cellular damage. These fatty acid changes might not only have a negative impact on the heart, but also contribute to the progression of kidney damage. The presence of these lipoprotein alterations within a biological environment characterized by increased inflammation and oxidative stress, as well as the competing risk of non-atherosclerotic cardiovascular death as kidney function declines, has important therapeutic implications. Additional research is needed to clarify the pathophysiological link between lipid and lipoprotein modifications, and kidney dysfunction, as well as the genesis and/or progression of CVD in patients with kidney disease.
Collapse
Affiliation(s)
- Heidi Noels
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, University Hospital, Aachen, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Michael Lehrke
- Department of Internal Medicine I, RWTH Aachen University, University Hospital, Aachen, Germany
| | - Raymond Vanholder
- Nephrology Section, Department of Internal Medicine and Pediatrics, University Hospital, Ghent, Belgium
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, University Hospital, Aachen, Germany.
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
39
|
Blanchard V, Chemello K, Hollstein T, Hong-Fong CC, Schumann F, Grenkowitz T, Nativel B, Coassin S, Croyal M, Kassner U, Lamina C, Steinhagen-Thiessen E, Lambert G. The size of apolipoprotein (a) is an independent determinant of the reduction in lipoprotein (a) induced by PCSK9 inhibitors. Cardiovasc Res 2021; 118:2103-2111. [PMID: 34314498 DOI: 10.1093/cvr/cvab247] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/24/2021] [Indexed: 12/14/2022] Open
Abstract
AIMS Lipoprotein (a) [Lp(a)] is a lipoprotein species causatively associated with atherosclerosis. Unlike statins, PCSK9 inhibitors (PCSK9i) reduce Lp(a), but this reduction is highly variable. Levels of Lp(a) are chiefly governed by the size of its signature protein, apolipoprotein (a) [apo(a)]. Whether this parameter determines some of the reduction in Lp(a) induced by PCSK9i remains unknown. We aimed to investigate if the Lp(a) lowering efficacy of PCSK9i is modulated by the size of apo(a), which is genetically determined by the variable number of KIV domains present on that protein. METHODS AND RESULTS The levels of Lp(a) and the size of apo(a) were assessed in plasma samples from 268 patients before and after treatment with PCSK9i. Patients were recruited at the Outpatient Lipid Clinic of the Charité Hospital (Berlin) between 2015 and 2020. They were hypercholesterolemic at very high CVD risk with LDL-cholesterol levels above therapeutic targets despite maximally tolerated lipid-lowering therapy. Patients received either Alirocumab (75 or 150 mg) or Evolocumab (140 mg) every 2 weeks. Apo(a), apoB100, and apoE concentrations as well as apoE major isoforms were determined by liquid chromatography high-resolution mass spectrometry. Apo(a) isoforms sizes were determined by Western Blot. PCSK9i sharply reduced LDL-cholesterol (-57%), apoB100 (-47%) and Lp(a) (-36%). There was a positive correlation between the size of apo(a) and the relative reduction in Lp(a) induced by PCSK9i (r = 0.363, p = 0.0001). The strength of this association remained unaltered after adjustment for baseline Lp(a) levels and all other potential confounding factors. In patients with two detectable apo(a) isoforms, there was also a positive correlation between the size of apo(a) and the reduction in Lp(a), separately for the smaller (r = 0.350, p = 0.0001) and larger (r = 0.324, p = 0.0003) isoforms. The relative contribution of the larger isoform to the total concentration of apo(a) was reduced from 29% to 15% (p < 0.0001). CONCLUSIONS The size of apo(a) is an independent determinant of the response to PCSK9i. Each additional kringle domain is associated with a 3% additional reduction in Lp(a). This explains in part the variable efficacy of PCSK9i and allows to identify patients who will benefit most from these therapies in terms of Lp(a) lowering. TRANSLATIONAL PERSPECTIVE Unlike statins, PCSK9 inhibitors reduce the circulating levels of the highly atherogenic Lipoprotein (a). The underlying mechanism remains a matter of considerable debate. The size of apo(a), the signature protein of Lp(a), is extremely variable (300 to more than 800 kDa) and depends on its number of kringle domains. We now show that each increase in apo(a) size by one kringle domain is associated with a 3% additional reduction in Lp(a) following PCSK9i treatment and that apo(a) size polymorphism is an independent predictor of the reduction in Lp(a) induced by these drugs. In an era of personalized medicine, this allows to identify patients who will benefit most from PCSK9i in terms of Lp(a) lowering.
Collapse
Affiliation(s)
- Valentin Blanchard
- Université de La Réunion, INSERM UMR 1188 DéTROI, Sainte-Clotilde, France.,Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada; Department of Medicine, UBC, Vancouver, Canada
| | - Kévin Chemello
- Université de La Réunion, INSERM UMR 1188 DéTROI, Sainte-Clotilde, France
| | - Tim Hollstein
- Department of Endocrinology, Campus Virchow-Klinikum, Charité Universitätsmedizin, Berlin, Germany.,Division of Endocrinology, Diabetology and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | | | - Friederike Schumann
- Department of Endocrinology, Campus Virchow-Klinikum, Charité Universitätsmedizin, Berlin, Germany
| | - Thomas Grenkowitz
- Department of Endocrinology, Campus Virchow-Klinikum, Charité Universitätsmedizin, Berlin, Germany
| | - Brice Nativel
- Université de La Réunion, INSERM UMR 1188 DéTROI, Sainte-Clotilde, France
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbrück, Innsbrück, Austria
| | - Mikaël Croyal
- NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, Nantes, France
| | - Ursula Kassner
- Department of Endocrinology, Campus Virchow-Klinikum, Charité Universitätsmedizin, Berlin, Germany
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbrück, Innsbrück, Austria
| | | | - Gilles Lambert
- Université de La Réunion, INSERM UMR 1188 DéTROI, Sainte-Clotilde, France
| |
Collapse
|
40
|
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
|
41
|
Kamstrup PR. Lipoprotein(a) and Cardiovascular Disease. Clin Chem 2021; 67:154-166. [PMID: 33236085 DOI: 10.1093/clinchem/hvaa247] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/25/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND High lipoprotein(a) concentrations present in 10%-20% of the population have long been linked to increased risk of ischemic cardiovascular disease. It is unclear whether high concentrations represent an unmet medical need. Lipoprotein(a) is currently not a target for treatment to prevent cardiovascular disease. CONTENT The present review summarizes evidence of causality for high lipoprotein(a) concentrations gained from large genetic epidemiologic studies and discusses measurements of lipoprotein(a) and future treatment options for high values found in an estimated >1 billion individuals worldwide. SUMMARY Evidence from mechanistic, observational, and genetic studies support a causal role of lipoprotein(a) in the development of cardiovascular disease, including coronary heart disease and peripheral arterial disease, as well as aortic valve stenosis, and likely also ischemic stroke. Effect sizes are most pronounced for myocardial infarction, peripheral arterial disease, and aortic valve stenosis where high lipoprotein(a) concentrations predict 2- to 3-fold increases in risk. Lipoprotein(a) measurements should be performed using well-validated assays with traceability to a recognized calibrator to ensure common cut-offs for high concentrations and risk assessment. Randomized cardiovascular outcome trials are needed to provide final evidence of causality and to assess the potential clinical benefit of novel, potent lipoprotein(a) lowering therapies.
Collapse
Affiliation(s)
- Pia R Kamstrup
- Department of Clinical Biochemistry.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark
| |
Collapse
|
42
|
Mathew RO, Rosenson RS, Lyubarova R, Chaudhry R, Costa SP, Bangalore S, Sidhu MS. Concepts and Controversies: Lipid Management in Patients with Chronic Kidney Disease. Cardiovasc Drugs Ther 2021; 35:479-489. [PMID: 32556851 DOI: 10.1007/s10557-020-07020-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains an important contributor of morbidity and mortality in patients with chronic kidney disease (CKD). CKD is recognized as an important risk enhancer that identifies patients as candidates for more intensive low-density lipoprotein (LDL) cholesterol lowering. However, there is controversy regarding the efficacy of lipid-lowering therapy, especially in patients on dialysis. Among patients with CKD, not yet on dialysis, there is clinical trial evidence for the use of statins with or without ezetimibe to reduce ASCVD events. Newer cholesterol lowering agents have been introduced for the management of hyperlipidemia to reduce ASCVD, but these therapies have not been tested in the CKD population except in secondary analyses of patients with primarily CKD stage 3. This review summarizes the role of hyperlipidemia in ASCVD and treatment strategies for hyperlipidemia in the CKD population.
Collapse
Affiliation(s)
- Roy O Mathew
- Columbia V.A. Health Care System, 6439 Garners Ferry Road, Columbia, SC, 29209, USA.
- University of South Carolina School of Medicine, Columbia, SC, USA.
| | | | | | | | | | | | - Mandeep S Sidhu
- Albany Medical College and Albany Medical Center, Albany, NY, USA
| |
Collapse
|
43
|
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
|
44
|
Yelnik CM, Bruckert É. [Hypercholesterolemia, from screening to treatment: Who, why and how to manage]. Rev Med Interne 2020; 42:707-713. [PMID: 33308860 DOI: 10.1016/j.revmed.2020.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/26/2020] [Accepted: 11/08/2020] [Indexed: 10/22/2022]
Abstract
Hypercholesterolemia refers to dyslipidemia with an increased circulating cholesterol levels. This is the most common dyslipidemia and is associated with an increased risk of developing atheromatous cardiovascular diseases. One of the major challenges in primary prevention is to define the threshold for therapeutic intervention that allow to obtain a significant clinical benefit without unnecessarily expose the patient to potential side effects of lipid-lowering treatments. It is also important to recall to screen patient for heterozygous familial hypercholesterolemia, a common genetic disease of lipid metabolism responsible for particularly severe and early coronary disease. In this article, the issues of hypercholesterolemia screening, the definition of therapeutic targets and expected benefits as well as the modalities of therapeutic management (by also addressing the problem of statin intolerance) will be addressed.
Collapse
Affiliation(s)
- C M Yelnik
- Université Lille, Inserm, U1167 Risk factors and molecular determinants of aging-related diseases, 59000 Lille, France; Service de médecine interne, d'immunologie clinique et de médecine polyvalente-post urgence, centre national de référence maladies systémiques et auto-immunes rares, European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases (ReCONNECT), CHU de Lille, 59000 Lille, France.
| | - É Bruckert
- Hôpital Pitié-Salpêtrière, Sorbonne université, institut hôpital-universitaire cardiométabolique et service d'endocrinologie-métabolisme, 75000 Paris, France
| |
Collapse
|
45
|
Pearson K, Rodriguez F. Lipoprotein(a) and Cardiovascular Disease Prevention across Diverse Populations. Cardiol Ther 2020; 9:275-292. [PMID: 32451810 PMCID: PMC7584702 DOI: 10.1007/s40119-020-00177-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Indexed: 12/16/2022] Open
Abstract
Lipoprotein(a) (Lp(a)) is a highly proatherogenic lipid fraction that is genetically determined and minimally responsive to lifestyle or behavior changes. Mendelian randomization studies have suggested a causal link between elevated Lp(a) and heart disease, stroke, and aortic stenosis. There is substantial inter-ethnic variation in Lp(a) levels, with persons of African descent having the highest median values. Monitoring of Lp(a) has historically been limited by lack of standardization of assays. With the advent of novel therapeutic modalities to lower Lp(a) levels including proprotein convertase subtilisin/kexin 9 (PCSK9) inhibitors and targeted antisense oligonucleotides, it is increasingly important to screen patients who have family or personal history of atherosclerotic cardiovascular disease for elevations in Lp(a). Further study is needed to establish a causal relationship between elevated Lp(a) and cardiovascular disease across diverse ethnic populations.
Collapse
Affiliation(s)
- Keon Pearson
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford University, Stanford, USA
| | - Fatima Rodriguez
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford University, Stanford, USA.
| |
Collapse
|
46
|
Increased cardiovascular risk associated with hyperlipoproteinemia (a) and the challenges of current and future therapeutic possibilities. Anatol J Cardiol 2020; 23:60-69. [PMID: 32011323 PMCID: PMC7040869 DOI: 10.14744/anatoljcardiol.2019.56068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Population, genetic, and clinical studies demonstrated a causative and continuous, from other plasma lipoproteins independent relationship between elevated plasma lipoprotein (a) [Lp(a)] concentration and the development of cardiovascular disease (CVD), mainly those related to athe-rosclerotic CVD, and calcific aortic stenosis. Currently, a strong international consensus is still lacking regarding the single value which would be commonly used to define hyperlipoproteinemia (a). Its prevalence in the general population is estimated to be in the range of 10%–35% in accordance with the most commonly used threshold levels (>30 or >50 mg/dL). Since elevated Lp(a) can be of special importance in patients with some genetic disorders, as well as in individuals with otherwise controlled major risk factors, the identification and establishment of the proper therapeutic interventions that would lower Lp(a) levels and lead to CVD risk reduction could be very important. The majority of the classical lipid-lowering agents (statins, ezetimibe, and fibrates), as well as nutraceuticals (CoQ10 and garlic), appear to have no significant effect on its plasma levels, whereas for the drugs with the demonstrated Lp(a)-lowering effects (aspirin, niacin, and estrogens), their clinical efficacy in reducing cardiovascular (CV) events has not been unequivocally proven yet. Both Lp(a) apheresis and proprotein convertase subtilisin/kexin type 9 inhibitors can reduce the plasma Lp(a) by approximately 20%–30% on average, in parallel with much larger reduction of low-density lipoprotein cholesterol (up to 70%), what puts us in a difficulty to conclude about the true contribution of lowered Lp(a) to the reduction of CV events. The most recent advancement in the field is the introduction of the novel apolipoprotein (a) [apo(a)] antisense oligonucleotide therapy targeting apo(a), which has already proven itself as being very effective in decreasing plasma Lp(a) (by even >90%), but should be further tested in clinical trials. The aim of this review was to present some of the most important accessible scientific data, as well as dilemmas related to the currently and potentially in the near future more widely available therapeutic options for the management of hyperlipoproteinemia (a).
Collapse
|
47
|
Abstract
Abstract
Collapse
Affiliation(s)
- Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, Milan, Italy
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Alberico Luigi Catapano
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
48
|
Bittner VA, Szarek M, Aylward PE, Bhatt DL, Diaz R, Edelberg JM, Fras Z, Goodman SG, Halvorsen S, Hanotin C, Harrington RA, Jukema JW, Loizeau V, Moriarty PM, Moryusef A, Pordy R, Roe MT, Sinnaeve P, Tsimikas S, Vogel R, White HD, Zahger D, Zeiher AM, Steg PG, Schwartz GG. Effect of Alirocumab on Lipoprotein(a) and Cardiovascular Risk After Acute Coronary Syndrome. J Am Coll Cardiol 2020; 75:133-144. [PMID: 31948641 DOI: 10.1016/j.jacc.2019.10.057] [Citation(s) in RCA: 329] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/07/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Lipoprotein(a) concentration is associated with cardiovascular events. Alirocumab, a proprotein convertase subtilisin/kexin type 9 inhibitor, lowers lipoprotein(a) and low-density lipoprotein cholesterol (LDL-C). OBJECTIVES A pre-specified analysis of the placebo-controlled ODYSSEY Outcomes trial in patients with recent acute coronary syndrome (ACS) determined whether alirocumab-induced changes in lipoprotein(a) and LDL-C independently predicted major adverse cardiovascular events (MACE). METHODS One to 12 months after ACS, 18,924 patients on high-intensity statin therapy were randomized to alirocumab or placebo and followed for 2.8 years (median). Lipoprotein(a) was measured at randomization and 4 and 12 months thereafter. The primary MACE outcome was coronary heart disease death, nonfatal myocardial infarction, ischemic stroke, or hospitalization for unstable angina. RESULTS Baseline lipoprotein(a) levels (median: 21.2 mg/dl; interquartile range [IQR]: 6.7 to 59.6 mg/dl) and LDL-C [corrected for cholesterol content in lipoprotein(a)] predicted MACE. Alirocumab reduced lipoprotein(a) by 5.0 mg/dl (IQR: 0 to 13.5 mg/dl), corrected LDL-C by 51.1 mg/dl (IQR: 33.7 to 67.2 mg/dl), and reduced the risk of MACE (hazard ratio [HR]: 0.85; 95% confidence interval [CI]: 0.78 to 0.93). Alirocumab-induced reductions of lipoprotein(a) and corrected LDL-C independently predicted lower risk of MACE, after adjustment for baseline concentrations of both lipoproteins and demographic and clinical characteristics. A 1-mg/dl reduction in lipoprotein(a) with alirocumab was associated with a HR of 0.994 (95% CI: 0.990 to 0.999; p = 0.0081). CONCLUSIONS Baseline lipoprotein(a) and corrected LDL-C levels and their reductions by alirocumab predicted the risk of MACE after recent ACS. Lipoprotein(a) lowering by alirocumab is an independent contributor to MACE reduction, which suggests that lipoprotein(a) should be an independent treatment target after ACS. (ODYSSEY Outcomes: Evaluation of Cardiovascular Outcomes After an Acute Coronary Syndrome During Treatment With Alirocumab; NCT01663402).
Collapse
Affiliation(s)
- Vera A Bittner
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama.
| | - Michael Szarek
- State University of New York, Downstate School of Public Health, Brooklyn, New York
| | - Philip E Aylward
- South Australian Health and Medical Research Institute, Flinders University and Medical Centre, Adelaide, South Australia, Australia
| | - Deepak L Bhatt
- Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, Massachusetts. https://twitter.com/DLBHATTMD
| | - Rafael Diaz
- Estudios Cardiológicos Latinoamérica, Instituto Cardiovascular de Rosario, Rosario, Argentina
| | | | - Zlatko Fras
- Division of Medicine, Department of Vascular Medicine, Preventive Cardiology Unit, University Medical Centre Ljubljana, Ljubljana, Slovenia; Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Sigrun Halvorsen
- Department of Cardiology, Oslo University Hospital, Oslo, Norway; University of Oslo, Oslo, Norway
| | | | - Robert A Harrington
- Stanford Center for Clinical Research, Department of Medicine, Stanford University, Stanford, California
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Patrick M Moriarty
- Division of Clinical Pharmacology, University of Kansas Medical Center, Kansas City, Kansas
| | | | - Robert Pordy
- Regeneron Pharmaceuticals Inc., Tarrytown, New York
| | - Matthew T Roe
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina; Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Peter Sinnaeve
- Department of Cardiovascular Medicine, University Hospitals Leuven, Leuven, Belgium; University of Leuven, Leuven, Belgium
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, California
| | - Robert Vogel
- Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Harvey D White
- Green Lane Cardiovascular Services Auckland City Hospital, Auckland, New Zealand
| | - Doron Zahger
- Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Andreas M Zeiher
- Department of Medicine III, Goethe University, Frankfurt am Main, Germany
| | - Ph Gabriel Steg
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Université de Paris, FACT (French Alliance for Cardiovascular Trials), INSERM U1148, Paris, France; National Heart and Lung Institute, Imperial College, Royal Brompton Hospital, London, United Kingdom. https://twitter.com/gabrielsteg
| | - Gregory G Schwartz
- Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado
| | | |
Collapse
|
49
|
Rehberger Likozar A, Zavrtanik M, Šebeštjen M. Lipoprotein(a) in atherosclerosis: from pathophysiology to clinical relevance and treatment options. Ann Med 2020; 52:162-177. [PMID: 32453609 PMCID: PMC7877976 DOI: 10.1080/07853890.2020.1775287] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lipoprotein(a) (Lp(a)) was discovered more than 50 years ago, and a decade later, it was recognized as a risk factor for coronary artery disease. However, it has gained importance only in the past 10 years, with emergence of drugs that can effectively decrease its levels. Lp(a) is a low-density lipoprotein (LDL) with an added apolipoprotein(a) attached to the apolipoprotein B component via a disulphide bond. Circulating levels of Lp(a) are mainly genetically determined. Lp(a) has many functions, which include proatherosclerotic, prothrombotic and pro-inflammatory roles. Here, we review recent data on the role of Lp(a) in the atherosclerotic process, and treatment options for patients with cardiovascular diseases. Currently 'Proprotein convertase subtilisin/kexin type 9' (PCSK9) inhibitors that act through non-specific reduction of Lp(a) are the only drugs that have shown effectiveness in clinical trials, to provide reductions in cardiovascular morbidity and mortality. The effects of PCSK9 inhibitors are not purely through Lp(a) reduction, but also through LDL cholesterol reduction. Finally, we discuss new drugs on the horizon, and gene-based therapies that affect transcription and translation of apolipoprotein(a) mRNA. Clinical trials in patients with high Lp(a) and low LDL cholesterol might tell us whether Lp(a) lowering per se decreases cardiovascular morbidity and mortality.KEY MESSAGESLipoprotein(a) is an important risk factor in patients with cardiovascular diseases.Lipoprotein(a) has many functions, which include proatherosclerotic, prothrombotic and pro-inflammatory roles.Treatment options to lower lipoprotein(a) levels are currently scarce, but new drugs are on the horizon.
Collapse
Affiliation(s)
| | - Mark Zavrtanik
- Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Miran Šebeštjen
- Department of Vascular Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Department of Cardiology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
50
|
Greco MF, Sirtori CR, Corsini A, Ezhov M, Sampietro T, Ruscica M. Lipoprotein(a) Lowering-From Lipoprotein Apheresis to Antisense Oligonucleotide Approach. J Clin Med 2020; 9:jcm9072103. [PMID: 32635396 PMCID: PMC7408876 DOI: 10.3390/jcm9072103] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
It is well-known that elevated lipoprotein(a)—Lp(a)—levels are associated with a higher risk of cardiovascular (CV) mortality and all-cause mortality, although a standard pharmacotherapeutic approach is still undefined for patients with high CV risk dependent on hyperlipoproteinemia(a). Combined with high Lp(a) levels, familial hypercholesterolemia (FH) leads to a greater CVD risk. In suspected FH patients, the proportion of cases explained by a rise of Lp(a) levels ranges between 5% and 20%. In the absence of a specific pharmacological approach able to lower Lp(a) to the extent required to achieve CV benefits, the most effective strategy today is lipoprotein apheresis (LA). Although limited, a clear effect on Lp(a) is exerted by PCSK9 antagonists, with apparently different mechanisms when given with statins (raised catabolism) or as monotherapy (reduced production). In the era of RNA-based therapies, a new dawn is represented by the use of antisense oligonucleotides APO(a)Lrx, able to reduce Lp(a) from 35% to over 80%, with generally modest injection site reactions. The improved knowledge of Lp(a) atherogenicity and possible prevention will be of benefit for patients with residual CV risk remaining after the most effective available lipid-lowering agents.
Collapse
Affiliation(s)
- Maria Francesca Greco
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
| | - Cesare R. Sirtori
- Dyslipidemia Center, A.S.S.T. Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Alberto Corsini
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
- IRCCS Multimedica, 20099 Milan, Italy
| | - Marat Ezhov
- National Medical Research Center of Cardiology of the Ministry of Health, Moscow, Russia;
| | - Tiziana Sampietro
- U.O. Lipoapheresis and Center for Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, 56126 Pisa, Italy;
| | - Massimiliano Ruscica
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
- Correspondence: ; Tel.: +39-0250318220
| |
Collapse
|