1
|
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
|
2
|
Abduljabbar MH. PCSK9 Inhibitors: Focus on Evolocumab and Its Impact on Atherosclerosis Progression. Pharmaceuticals (Basel) 2024; 17:1581. [PMID: 39770423 PMCID: PMC11676890 DOI: 10.3390/ph17121581] [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] [Received: 08/29/2024] [Revised: 10/22/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
This paper investigates the therapeutic use of PCSK9 inhibitors, particularly Evolocumab, as monoclonal antibodies for the treatment of atherosclerosis based on recent literature reviews. PCSK9 is an outstanding example of a breakthrough in medical science, with advancements in understanding its biological function driving substantial progress in atherosclerosis treatment. Atherosclerotic cardiovascular disease (ASCVD) is a leading global cause of mortality, imposing substantial financial burdens on healthcare systems. Elevated low-density lipoprotein cholesterol (LDL-C), a modifiable risk factor, plays a pivotal role in the development of ASCVD. Emerging treatments such as PCSK9 inhibitors are now being introduced to combat this issue, with the goal of reducing ASCVD risk by directly targeting LDL-C levels. This discovery highlighted the potential of monoclonal antibodies to inhibit PCSK9, thereby enhancing LDL-C receptor activity. This breakthrough led to the development of Alirocumab and Evolocumab inhibitors, which typically reduce LDL-C levels by approximately 50%. This research underscores the importance of PCSK9 inhibitors in treating ASCVD, drawing on evidence from various randomized controlled trials such as FOURIER, ODYSSEY OUTCOMES, and VESALIUS-CV. These trials have also shown that PCSK9 inhibitors are effective and safe for the treatment of several cardiovascular disorders. PCSK9 inhibitors are therefore useful in patients who do not reach their target LDL-C levels when on the highest doses of statins or patients with very high cardiovascular risk who cannot tolerate statins at all.
Collapse
Affiliation(s)
- Maram H Abduljabbar
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
3
|
Cao Zhang AM, Ziogos E, Harb T, Gerstenblith G, Leucker TM. Emerging clinical role of proprotein convertase subtilisin/kexin type 9 inhibition-Part one: Pleiotropic pro-atherosclerotic effects of PCSK9. Eur J Clin Invest 2024; 54:e14273. [PMID: 38922860 DOI: 10.1111/eci.14273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) is primarily recognized for its role in lipid metabolism, but recent evidence suggests that it may have broader implications due to its diverse tissue expression. OBJECTIVE This review aims to explore the multifaceted functions of PCSK9, highlighting its pro-atherosclerotic effects, including its impact on circulating lipoprotein variables, non-low-density lipoprotein receptors, and various cell types involved in atherosclerotic plaque development. CONCLUSIONS PCSK9 exhibits diverse roles beyond lipid metabolism, potentially contributing to atherosclerosis through multiple pathways. Understanding these mechanisms could offer new insights into therapeutic strategies targeting PCSK9 for cardiovascular disease management.
Collapse
Affiliation(s)
- Alexander M Cao Zhang
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Efthymios Ziogos
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tarek Harb
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gary Gerstenblith
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thorsten M Leucker
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Fogacci F, Yerlitaş Sİ, Giovannini M, Zararsız G, Lido P, Borghi C, Cicero AFG. Sex X Time Interactions in Lp(a) and LDL-C Response to Evolocumab. Biomedicines 2023; 11:3271. [PMID: 38137492 PMCID: PMC10741148 DOI: 10.3390/biomedicines11123271] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
The aim of this study was to evaluate whether there were significant sex x time interactions in lipoprotein(a) (Lp(a)) and low-density lipoprotein cholesterol (LDL-C) response to treatment with the Proprotein Convertase Subtilisin/Kexin type 9 inhibitor (PCSK9i) Evolocumab, in a real-life clinical setting. For this purpose, we pooled data from 176 outpatients (Men: 93; Women: 83) clinically evaluated at baseline and every six months after starting Evolocumab. Individuals who had been on PCSK9i for less than 30 months and nonadherent patients were excluded from the analysis. Over time, absolute values of Lp(a) plasma concentrations significantly decreased in the entire cohort (p-value < 0.001) and by sex (p-value < 0.001 in men and p-value = 0.002 in and women). However, there were no sex-related significant differences. Absolute plasma concentrations of LDL-C significantly decreased over time in the entire cohort and by sex (p-value < 0.001 always), with greater improvements in men compared to women. The sex x time interaction was statistically significant in LDL-C (all p-values < 0.05), while absolute changes in Lp(a) were not influenced by either sex or time (all p-value > 0.05). Our data partially reinforce the presence of differences in response to treatment to PCSK9i between men and women and are essential to gain a better understanding of the relationship between LDL-C and Lp(a) lowering in response to PCSK9i. Further research will clarify whether these sex-related significant differences translate into a meaningful difference in the long-term risk of ASCVD.
Collapse
Affiliation(s)
- Federica Fogacci
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (F.F.); (M.G.); (C.B.)
| | - Serra İlayda Yerlitaş
- Department of Biostatistics, Erciyes University School of Medicine, 38039 Kayseri, Turkey; (S.İ.Y.); (G.Z.)
- Drug Application and Research Center (ERFARMA), Erciyes University, 38280 Kayseri, Turkey
| | - Marina Giovannini
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (F.F.); (M.G.); (C.B.)
| | - Gökmen Zararsız
- Department of Biostatistics, Erciyes University School of Medicine, 38039 Kayseri, Turkey; (S.İ.Y.); (G.Z.)
- Drug Application and Research Center (ERFARMA), Erciyes University, 38280 Kayseri, Turkey
| | - Paolo Lido
- Italian Medicines Agency (AIFA), 00187 Rome, Italy;
| | - Claudio Borghi
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (F.F.); (M.G.); (C.B.)
- Unit of Cardiovascular Internal Medicine, Department of Cardiac, Thoracic, Vascular Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40100 Bologna, Italy
| | - Arrigo F. G. Cicero
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (F.F.); (M.G.); (C.B.)
- Unit of Cardiovascular Internal Medicine, Department of Cardiac, Thoracic, Vascular Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40100 Bologna, Italy
| |
Collapse
|
5
|
Milosavljevic MN, Stefanovic SM, Pejcic AV. Potential Novel RNA-Targeting Agents for Effective Lipoprotein(a) Lowering: A Systematic Assessment of the Evidence From Completed and Ongoing Developmental Clinical Trials. J Cardiovasc Pharmacol 2023; 82:1-12. [PMID: 37070852 DOI: 10.1097/fjc.0000000000001429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 03/25/2023] [Indexed: 04/19/2023]
Abstract
ABSTRACT An increase in blood lipoprotein (a) [Lp(a)] levels, mostly genetically determined, has been identified as an independent risk factor of atherosclerotic cardiovascular disease. No drug has yet been approved that markedly lowers Lp(a) and thereby reduces residual cardiovascular risk. The aim of this article was to critically review the evidence from clinical development studies to date on the efficacy and safety of new RNA-based therapeutics for targeted lowering of Lp(a). PubMed/MEDLINE, Scopus, Web of Science, and ClinicalTrials.gov were searched without any language or date restriction up to November 5, 2022, and a total of 12 publications and 22 trial records were included. Several drugs were found that are currently in various stages of clinical development, such as the antisense oligonucleotide pelacarsen and the small interfering RNA molecule olpasiran and drugs coded as SLN360 and LY3819469. Among them, pelacarsen has progressed the most, currently reaching phase 3. All these drugs have so far shown satisfactory pharmacokinetic properties, consistently high and stable, dose-dependent efficacy in lowering Lp(a) even by more than 90%, with an acceptable safety profile in subjects with highly elevated Lp(a). In addition, reports of early clinical trials with pelacarsen imply a promising suppressive effect on key mechanisms of atherogenesis. Future research should focus on confirming these beneficial clinical effects in patients with lower average Lp(a) levels and clearly demonstrating the association between lowering Lp(a) and reducing adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Milos N Milosavljevic
- Department of Pharmacology and Toxicology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Srdjan M Stefanovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia; and
- Department of Clinical Pharmacology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Ana V Pejcic
- Department of Pharmacology and Toxicology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
6
|
Hardy J, Niman S, Goldfaden RF, Ashchi M, Bisharat M, Huston J, Hartmann H, Choksi R. A Review of the Clinical Pharmacology of Pelacarsen: A Lipoprotein(a)-Lowering Agent. Am J Cardiovasc Drugs 2022; 22:47-54. [PMID: 34490591 DOI: 10.1007/s40256-021-00499-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 11/26/2022]
Abstract
Patients with genetically associated elevated lipoprotein(a) [Lp(a)] levels are at greater risk for coronary artery disease, heart attack, stroke, and peripheral arterial disease. To date, there are no US FDA-approved drug therapies that are designed to target Lp(a) with the goal of lowering the Lp(a) level in patients who have increased risk. The American College of Cardiology (ACC) has provided guidelines on how to use traditional lipid profiles to assess the risk of atherosclerotic cardiovascular disease (ASCVD); however, even with the emergence of statin add-on therapies such as ezetimibe and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, some populations with elevated Lp(a) biomarkers remain at an increased risk for cardiovascular (CV) disease. Residual CV risk has led researchers to inquire about how lowering Lp(a) can be used as a potential preventative therapy in reducing CV events. This review aims to present and discuss the current clinical and scientific evidence pertaining to pelacarsen.
Collapse
Affiliation(s)
- Jennifer Hardy
- East Coast Institute for Research, Jacksonville, FL, USA
| | | | | | - Majdi Ashchi
- Ashchi Heart and Vascular Center, Jacksonville, FL, USA
| | | | - Jessica Huston
- College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | | | - Rushab Choksi
- East Coast Institute for Research, Jacksonville, FL, USA
| |
Collapse
|
7
|
Krittanawong C, Khawaja M, Rosenson RS, Amos CI, Nambi V, Lavie CJ, Virani SS. Association of PCSK9 Variants With the Risk of Atherosclerotic Cardiovascular Disease and Variable Responses to PCSK9 Inhibitor Therapy. Curr Probl Cardiol 2021; 47:101043. [PMID: 34780866 DOI: 10.1016/j.cpcardiol.2021.101043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/29/2022]
Abstract
Genetic polymorphisms or variations, randomly distributed in a population, may cause drug-gene response variations. Investigation into these polymorphisms may identify novel mechanisms contributing to a specific disease process. Such investigation necessitates the use of Mendelian randomization, an analytical method that uses genetic variants as instrumental variables for modifiable risk factors that affect population health.1 In the past decade, advances in our understanding of genetic polymorphisms have enabled the identification of genetic variants in candidate genes that impact low-density lipoprotein cholesterol (LDL-C) regulating pathways and cardiovascular disease (CVD) outcomes. A specific candidate gene of interest is that of the LDL receptor degrading protein, PCSK9. In fact, loss-of-function genetic variants for the PCSK9 gene are what first highlighted this pathway as a candidate for pharmacologic inhibition. PCSK9 inhibitors (PCSK9i) are a class of cholesterol-lowering medications that provide significant reductions in LDL by inhibiting the degradation of LDL receptors (LDLR). These inhibitors have also been found to reduce production and enhance clearance of lipoprotein A (Lp[a]), an LDL-like particle currently under study as a separate risk factor for atherosclerotic CVD. Here, we discuss the promise of personalized medicine in developing a more efficacious and individualized pharmacogenomics-based approach for the use of PCSK9i that considers genetic variation and targets different patient populations. This review explores the pharmacogenomics of PCSK9i in the context of PCSK9 allele variants related to drug-metabolizing enzymes and responses since more studies are demonstrating that some patients are hyporesponsive or non-responsive to PCSK9i.2 In summary, the pharmacogenomics of PCSK9 are a promising therapeutic target and genetic information from prospective randomized clinical trials is warranted to gain a full understanding of the efficacy and cost-effectiveness of such allele and/or gene-guided PCSK9i therapy.
Collapse
Affiliation(s)
- Chayakrit Krittanawong
- The Michael E. DeBakey VA Medical Center, Houston, TX; Section of Cardiology, Baylor College of Medicine, Houston, TX.
| | - Muzamil Khawaja
- The Michael E. DeBakey VA Medical Center, Houston, TX; Section of Cardiology, Baylor College of Medicine, Houston, TX
| | - Robert S Rosenson
- Director, Cardiometabolics Unit, Mount Sinai Hospital, Mount Sinai Heart, NY, NY
| | - Christopher I Amos
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, TX
| | - Vijay Nambi
- The Michael E. DeBakey VA Medical Center, Houston, TX; Section of Cardiology, Baylor College of Medicine, Houston, TX
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School, The University of Queensland School of Medicine, New Orleans, LA
| | - Salim S Virani
- The Michael E. DeBakey VA Medical Center, Houston, TX; Section of Cardiology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
8
|
Korneva VA, Kuznetsova TY, Julius U. Modern Approaches to Lower Lipoprotein(a) Concentrations and Consequences for Cardiovascular Diseases. Biomedicines 2021; 9:biomedicines9091271. [PMID: 34572458 PMCID: PMC8469722 DOI: 10.3390/biomedicines9091271] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 12/11/2022] Open
Abstract
Lipoprotein(a) (Lp(a)) is a low density lipoprotein particle that is associated with poor cardiovascular prognosis due to pro-atherogenic, pro-thrombotic, pro-inflammatory and pro-oxidative properties. Traditional lipid-lowering therapy does not provide a sufficient Lp(a) reduction. For PCSK9 inhibitors a small reduction of Lp(a) levels could be shown, which was associated with a reduction in cardiovascular events, independently of the effect on LDL cholesterol. Another option is inclisiran, for which no outcome data are available yet. Lipoprotein apheresis acutely and in the long run decreases Lp(a) levels and effectively improves cardiovascular prognosis in high-risk patients who cannot be satisfactorily treated with drugs. New drugs inhibiting the synthesis of apolipoprotein(a) (an antisense oligonucleotide (Pelacarsen) and two siRNA drugs) are studied. Unlike LDL-cholesterol, for Lp(a) no target value has been defined up to now. This overview presents data of modern capabilities of cardiovascular risk reduction by lowering Lp(a) level.
Collapse
Affiliation(s)
- Victoria A. Korneva
- Department of Faculty Therapy, Petrozavodsk State University, Lenin Ave. 33, 185000 Petrozavodsk, Russia;
- Correspondence:
| | | | - Ulrich Julius
- Lipidology and Lipoprotein Apheresis Center, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany;
| |
Collapse
|
9
|
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
|
10
|
Mahmood T, Minnier J, Ito MK, Li QH, Koren A, Kam IW, Fazio S, Shapiro MD. Discordant responses of plasma low-density lipoprotein cholesterol and lipoprotein(a) to alirocumab: A pooled analysis from 10 ODYSSEY Phase 3 studies. Eur J Prev Cardiol 2021; 28:816-822. [PMID: 34298554 DOI: 10.1177/2047487320915803] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/09/2020] [Indexed: 12/24/2022]
Abstract
AIMS Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors consistently reduce low-density lipoprotein cholesterol (LDL-C) by 50-60% and lipoprotein(a) (Lp(a)) by 20-30%, but the mechanism of Lp(a) lowering remains unclear. If Lp(a) is cleared by the LDL receptor, similar to LDL-C, then one would expect PCSK9 inhibition to induce a concordant LDL-C/Lp(a) response in an approximately 2:1 ratio. We aim to determine the prevalence of discordant plasma LDL-C/Lp(a) response to the PCSK9 inhibitor alirocumab. METHODS This is a post hoc, pooled analysis of 10 randomized controlled trials from the ODYSSEY Phase 3 clinical trial program for alirocumab. Patients enrolled in the trials were high cardiovascular risk and/or with heterozygous familial hypercholesterolemia. The primary end point was prevalence of discordant LDL-C/Lp(a) response to alirocumab at 24 weeks. Discordant response was defined as LDL-C reduction >35% and Lp(a) reduction ≤10%, or LDL-C reduction ≤35% and Lp(a) reduction >10%. RESULTS Of the 1709 patients in the pooled study cohort, 62.4% were male, and the mean age was 59.2 (SD: 11.0) years. Baseline mean LDL-C was 126.5 (SD: 46.3) mg/dL and baseline median Lp(a) was 46.9 (interquartile range: 21.8-89.0) mg/dL. Total prevalence of discordant LDL-C/Lp(a) response was 21.5% (12.6% with LDL-C >35% reduction and Lp(a) ≤10% reduction; 8.9% with LDL-C ≤35% reduction and Lp(a) >10% reduction). Baseline Lp(a) and familial hypercholesterolemia status did not affect discordance. CONCLUSION A high prevalence of discordant LDL-C/Lp(a) response was observed with alirocumab, further suggesting that PCSK9 inhibitor therapy with alirocumab reduces plasma Lp(a) through alternative pathways to LDL receptor clearance.
Collapse
Affiliation(s)
- Tahir Mahmood
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology, USA
| | - Jessica Minnier
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology, USA
- Oregon Health & Science University, OHSU-PSU School of Public Health, USA
| | | | | | | | | | - Sergio Fazio
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology, USA
| | - Michael D Shapiro
- Center for Preventive Cardiology, Wake Forest University Baptist Medical Center, Section on Cardiovascular Medicine, USA
| |
Collapse
|
11
|
Chakraborty A, Pang J, Chan DC, Barnett W, Woodward AM, Vorster M, Watts GF. Effectiveness of proprotein convertase subtilisin/kexin-9 monoclonal antibody treatment on plasma lipoprotein(a) concentrations in patients with elevated lipoprotein(a) attending a clinic. Clin Cardiol 2021; 44:805-813. [PMID: 33955565 PMCID: PMC8207967 DOI: 10.1002/clc.23607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Lipoprotein(a) (Lp[a]) is a causal risk factor for atherosclerotic cardiovascular disease (ASCVD). Proprotein convertase subtilisin/kexin-9 monoclonal antibodies (PCSK9mAbs) can lower Lp(a) levels in clinical trials, but their effects in patients with elevated Lp(a) in clinical practice remain unclear. AIMS To investigate the effectiveness and safety of PCSK9mAbs in lowering plasma Lp(a) in patients with elevated Lp(a) concentrations in a lipid clinic. METHODS This was an open-label study of 53 adult patients with elevated Lp(a) concentration (≥0.5 g/L). Clinical, biochemical, and safety data were collected before and on treatment with evolocumab or alirocumab over a mean period of 11 months. RESULTS Treatment with a PCSK9mAb resulted in a significant reduction of 0.29 g/L (-22%) in plasma Lp(a) concentration (p<.001). There were also significant reductions in low-density lipoprotein-cholesterol (LDL-C) (-53%), remnant-cholesterol (-12%) and apolipoprotein B (-43%) concentrations. The change in Lp(a) concentration was significantly different from a comparable group of 35 patients with elevated Lp(a) who were not treated with a PCSK9mAb (-22% vs. -2%, p<.001). The reduction in Lp(a) concentration was not associated with the corresponding changes in LDL-C, remnant-cholesterol, and apolipoprotein B (p>.05 in all). 7.5% and 47% of the patients attained a target concentration of Lp(a) <0.5 g/L and LDL-C <1.8 mmol/L, respectively. PCSK9mAbs were well tolerated, the common adverse effects being pharyngitis (9.4%), nasal congestion (7.6%), myalgia (9.4%), diarrhoea (7.6%), arthralgia (9.4%) and injection site reactions (11%). CONCLUSION PCSK9mAbs can effectively and safely lower plasma Lp(a) concentrations in patients with elevated Lp(a) in clinical practice; the impact of the fall in Lp(a) on ASCVD outcomes requires further investigation.
Collapse
Affiliation(s)
- Anindita Chakraborty
- School of Medicine, Faculty of Health and Medical SciencesUniversity of Western AustraliaPerthAustralia
| | - Jing Pang
- School of Medicine, Faculty of Health and Medical SciencesUniversity of Western AustraliaPerthAustralia
| | - Dick C. Chan
- School of Medicine, Faculty of Health and Medical SciencesUniversity of Western AustraliaPerthAustralia
| | - Wendy Barnett
- Lipid Disorders Clinic, Cardiometabolic Services, Department of CardiologyRoyal Perth HospitalPerthAustralia
| | - Ann Marie Woodward
- Lipid Disorders Clinic, Cardiometabolic Services, Department of CardiologyRoyal Perth HospitalPerthAustralia
| | - Mary Vorster
- Lipid Disorders Clinic, Cardiometabolic Services, Department of CardiologyRoyal Perth HospitalPerthAustralia
| | - Gerald F. Watts
- School of Medicine, Faculty of Health and Medical SciencesUniversity of Western AustraliaPerthAustralia
- Lipid Disorders Clinic, Cardiometabolic Services, Department of CardiologyRoyal Perth HospitalPerthAustralia
| |
Collapse
|
12
|
Xu J, Shapiro MD. Current Evidence and Future Directions of PCSK9 Inhibition. US CARDIOLOGY REVIEW 2021; 15:e01. [PMID: 39720497 PMCID: PMC11664773 DOI: 10.15420/usc.2020.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/10/2020] [Indexed: 11/04/2022] Open
Abstract
Recent scientific and therapeutic advances in proprotein convertase subtilisin kexin type 9 (PCSK9) inhibition have opened a chapter in the management of hypercholesterolemia, especially in patients who are inadequately controlled on or intolerant to statins. The two PCSK9 monoclonal antibodies, evolocumab and alirocumab, reduce LDL cholesterol by 60% and improve cardiovascular outcomes when taken in addition to statin therapy. More recently, inclisiran, a silencing RNA (siRNA) that inhibits translation of PCSK9 mRNA, demonstrated LDL cholesterol reduction by 45-50% with the advantage of dramatically reduced dose frequency. Other modes of PCSK9 inhibition include small molecule antagonists, vaccines, CRISPR gene editing, and antagonism at various steps of translation, and post-translational processing.
Collapse
Affiliation(s)
- Jiaqian Xu
- Center for the Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Wake Forest University Baptist Medical Center Winston Salem, NC
| | - Michael D Shapiro
- Center for the Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Wake Forest University Baptist Medical Center Winston Salem, NC
| |
Collapse
|
13
|
Zanetti D, Gustafsson S, Assimes TL, Ingelsson E. Comprehensive Investigation of Circulating Biomarkers and Their Causal Role in Atherosclerosis-Related Risk Factors and Clinical Events. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2020; 13:e002996. [PMID: 33125266 DOI: 10.1161/circgen.120.002996] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Circulating biomarkers have been previously associated with atherosclerosis-related risk factors, but the nature of these associations is incompletely understood. METHODS We performed multivariable-adjusted regressions and 2-sample Mendelian randomization analyses to assess observational and causal associations of 27 circulating biomarkers with 7 cardiovascular traits in up to 451 933 participants of the UK Biobank. RESULTS After multiple-testing correction (alpha=1.3×10-4), we found a total of 15, 9, 21, 22, 26, 24, and 26 biomarkers strongly associated with coronary artery disease, ischemic stroke, atrial fibrillation, type 2 diabetes, systolic blood pressure, body mass index, and waist-to-hip ratio; respectively. The Mendelian randomization analyses confirmed strong evidence of previously suggested causal associations for several glucose- and lipid-related biomarkers with type 2 diabetes and coronary artery disease. Particularly interesting findings included a protective role of IGF-1 (insulin-like growth factor 1) in systolic blood pressure, and the strong causal association of lipoprotein(a) in coronary artery disease development (β, -0.13; per SD change in exposure and outcome and odds ratio, 1.28; P=2.6×10-4 and P=7.4×10-35, respectively). In addition, our results indicated a causal role of increased ALT (alanine aminotransferase) in the development of type 2 diabetes and hypertension (odds ratio, 1.59 and β, 0.06, per SD change in exposure and outcome; P=4.8×10-11 and P=6.0×10-5). Our results suggest that it is unlikely that CRP (C-reactive protein) and vitamin D play causal roles of any meaningful magnitude in development of cardiometabolic disease. CONCLUSIONS We confirmed and extended known associations and reported several novel causal associations providing important insights about the cause of these diseases, which can help accelerate new prevention strategies.
Collapse
Affiliation(s)
- Daniela Zanetti
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, CA (D.Z., T.L.A., E.I.)
- Stanford Cardiovascular Institute (D.Z., T.L.A., E.I.), Stanford University, CA
- Stanford Diabetes Research Center (D.Z., E.I.), Stanford University, CA
| | - Stefan Gustafsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Sweden (S.G.)
| | - Themistocles L Assimes
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, CA (D.Z., T.L.A., E.I.)
- Stanford Cardiovascular Institute (D.Z., T.L.A., E.I.), Stanford University, CA
| | - Erik Ingelsson
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, CA (D.Z., T.L.A., E.I.)
- Stanford Cardiovascular Institute (D.Z., T.L.A., E.I.), Stanford University, CA
- Stanford Diabetes Research Center (D.Z., E.I.), Stanford University, CA
| |
Collapse
|
14
|
Aggressive Treatment for Severe Forms of Familial Hypercholesterolemia. J Am Coll Cardiol 2020; 75:575-577. [PMID: 32057370 DOI: 10.1016/j.jacc.2020.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/03/2020] [Indexed: 11/23/2022]
|
15
|
Cho KH, Hong YJ. Proprotein convertase subtilisin/kexin type 9 inhibition in cardiovascular disease: current status and future perspectives. Korean J Intern Med 2020; 35:1045-1058. [PMID: 32921006 PMCID: PMC7487297 DOI: 10.3904/kjim.2020.140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/20/2020] [Indexed: 01/14/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) targets the degradation of low-density lipoprotein (LDL) receptors; it has been proved that its inhibition improves cardiovascular outcomes in patients with established atherosclerotic cardiovascular disease (ASCVD). Herein, we review the current status of PCSK9 inhibitors in clinical practice and the future scope of PCSK9 inhibition. The results of two recent large clinical trials reveal that two PCSK9 monoclonal antibodies evolocumab and alirocumab reduce the risk of a cardiovascular event on top of background statin therapy in patients with stable ASCVD and those with recent acute coronary syndrome, respectively. However, there are several ongoing concerns regarding the efficacy in reducing mortality, cost-effectiveness, and long-term safety of extremely low LDL cholesterol levels with PCSK9 inhibition. The results of ongoing cardiovascular outcomes trials with PCSK9 monoclonal antibodies for primary prevention and with small interfering RNA to PCSK9 for secondary prevention may help to shape the use of this new therapeutic class.
Collapse
Affiliation(s)
- Kyung Hoon Cho
- Division of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| | - Young Joon Hong
- Division of Cardiology, Chonnam National University Medical School, Gwangju, Korea
- Correspondence to Young Joon Hong, M.D. Division of Cardiology, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 61469, Korea Tel: +82-62-220-5778 Fax: +82-62-223-3105 E-mail:
| |
Collapse
|
16
|
Lipoprotein(a) and Atherosclerotic Cardiovascular Disease: Current Understanding and Future Perspectives. Cardiovasc Drugs Ther 2020; 33:739-748. [PMID: 31655942 DOI: 10.1007/s10557-019-06906-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE To review current knowledge of elevated lipoprotein(a) [Lp(a)] levels in relation to atherosclerotic cardiovascular disease (ASCVD) and discuss their potential use as biomarkers and therapeutic approaches in clinical practice. METHODS We summarized the current understanding and recent advances in the structure, metabolism, atherogenic mechanisms, standardized laboratory measurement, recommended screening populations, and prognostic value of Lp(a), with a special focus on the current potential treatment approaches for hyperlipoprotein(a)emia in patients with ASCVD. RESULTS Lp(a) is composed of LDL-like particle and characteristic apolipoprotein(a) [apo(a)] connected by a disulfide bond. Substantial evidence shows that elevated plasma Lp(a) level is a heritable, independent, and possibly causal risk factor for ASCVD through its proatherogenic, proinflammatory, and potentially prothrombotic properties. Current guidelines recommend Lp(a) measurement for patients with an intermediate-high risk of ASCVD, familial hypercholesterolemia, a family history of early ASCVD or elevated Lp(a), and progressive ASCVD despite receiving optimal therapy. Traditional Lp(a)-lowering approaches such as niacin, PCSK9 inhibitors, mipomersen, lomitapide, and lipoprotein apheresis were associated with a non-specific and limited reduction of Lp(a), intolerable side effects, invasive procedure, and high expense. The phase 2 randomized controlled trial of antisense oligonucleotide against the apo(a) encoding gene LPA mRNA showed that IONIS-APO(a)-LRX could specifically reduce the level of Lp(a) by 90% with good tolerance, which may become a promising candidate for the prevention and treatment of ASCVD in the future. CONCLUSIONS It is reasonable to measure Lp(a) levels to reclassify ASCVD risk and manage individuals with elevated Lp(a) to further reduce the residual risk of ASCVD, especially with IONIS-APO(a)-LRX.
Collapse
|
17
|
Shapiro MD, Fazio S. Lipoprotein(a). JACC Basic Transl Sci 2020; 5:558-560. [PMID: 32614936 PMCID: PMC7315180 DOI: 10.1016/j.jacbts.2020.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
18
|
Julius U, Tselmin S, Schatz U, Fischer S, Birkenfeld AL, Bornstein SR. Actual situation of lipoprotein apheresis in patients with elevated lipoprotein(a) levels. ATHEROSCLEROSIS SUPP 2020; 40:1-7. [PMID: 31818437 DOI: 10.1016/j.atherosclerosissup.2019.08.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
An elevation of lipoprotein(a) (Lp(a)) is an internationally recognized atherogenic risk factor, documented in epidemiological studies, in studies with Mendelian randomization and in genome-wide association studies (GWAS). At present, no drug is available to effectively reduce its concentration. In Germany, an elevation of Lp(a) associated with progressive cardiovascular diseases is officially recognized as an indication for a lipoprotein apheresis (LA). The number of patients who were treated with LA with this abnormality was steadily increasing in the years 2013-2016 - the official data are reported. In all new patients, who started to be treated at our LA center in 2017 (n = 20) the increased Lp(a) was a main indication for extracorporeal therapy, though some of them also showed clearly elevated LDL cholesterol (LDL-C) concentrations despite being treated with a maximal tolerated lipid-lowering drug therapy. A diabetes mellitus was seen in 5 patients. The higher was the Lp(a) level before the first LA session, the higher was the cardiovascular risk. Lp(a) concentrations measured before LA sessions were usually about 20% lower than those before the start of the LA therapy. Acutely, Lp(a) levels were reduced by about 70%. Following LA sessions the Lp(a) levels increased and in the majority reach pre-session concentrations after one week. Thus a weekly interval is best for the patients, but a few may need two sessions per week to stop the progress of atherosclerosis. The interval mean values were about 39% lower than previous levels. Several papers had been published showing a higher efficiency of LA therapy on the incidence of cardiovascular events in patients with high Lp(a) values when comparing with hypercholesterolemic patients with normal Lp(a) concentrations. Russian specific anti-Lp(a) columns positively affected coronary atherosclerosis. PCSK9 inhibitors reduce Lp(a) concentrations in many patients and in this way have a positive impact on cardiovascular outcomes. In the future, an antisense oligonucleotide against apolipoprotein(a) may be an alternative therapeutic option, provided a clear-cut reduction of cardiovascular events will be demonstrated.
Collapse
Affiliation(s)
- Ulrich Julius
- Lipidology and Center for Extracorporeal Treatment, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Sergey Tselmin
- Lipidology and Center for Extracorporeal Treatment, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany, Fetscherstr. 74, 01307, Dresden, Germany
| | - Ulrike Schatz
- Lipidology and Center for Extracorporeal Treatment, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany, Fetscherstr. 74, 01307, Dresden, Germany
| | - Sabine Fischer
- Lipidology and Center for Extracorporeal Treatment, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany, Fetscherstr. 74, 01307, Dresden, Germany
| | - Andreas L Birkenfeld
- Lipidology and Center for Extracorporeal Treatment, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany, Fetscherstr. 74, 01307, Dresden, Germany
| | - Stefan R Bornstein
- Lipidology and Center for Extracorporeal Treatment, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany, Fetscherstr. 74, 01307, Dresden, Germany
| |
Collapse
|
19
|
Liberopoulos E. Lipoprotein(a) reduction with proprotein convertase subtilisin/kexin type 9 inhibitors: An unsolved mystery. Eur J Prev Cardiol 2020; 28:813-815. [PMID: 33611488 DOI: 10.1177/2047487320926777] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
20
|
Unusual responses to PCSK9 inhibitors in a clinical cohort utilizing a structured follow-up protocol. Am J Prev Cardiol 2020; 1:100012. [PMID: 34327453 PMCID: PMC8315390 DOI: 10.1016/j.ajpc.2020.100012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 12/02/2022] Open
Abstract
Objective To characterize unusual responses to PCSK9 inhibitor (PCSK9i) therapy in a real-world setting, given their extremely low prevalence in clinical trials. Methods A retrospective study of patients seen in a structured academic PCSK9i clinic who had LDL-C measurements before and after initiation of PCSK9i (up to 12 months). Unusual response was defined as: (1) no response: no changes in LDL-C level at all time points; (2) delayed response: <30% LDL-C reduction by the third dose, but achieving this threshold at a later time; (3) reduced response: <30% LDL-C reduction at all time points; and (4) lost response: ≥30% LDL-C reduction by the third dose, but displaying <30% reduction at a later time. Results Of the 411 patients meeting inclusion criteria, 54 were initially classified as unusual responders. After excluding those not adherent to prescribed interventions, 31 patients (7.5%) were classified as true unusual responders. These included: 2 with no response, 12 with delayed response, 3 with reduced response, 6 with delayed or reduced response, 4 with lost response, and 4 with delayed and lost response. Response to PCSK9i therapy at all time points revealed higher on-treatment LDL-C values (94–100 vs. 47–51 mg/dL, p < 0.001) and lower degree of percent reduction in LDL-C (23.3–34% vs. 61.1–64.5%, p < 0.001) in the unusual versus usual responders. Lipoprotein (a) (Lp[a]) values were consistently higher in the unusual responders (81–92.5 vs. 28.5–52 mg/dL, p < 0.01). Fold change in post-versus pre-treatment PCSK9 plasma results was similar between the two cohorts (p > 0.05), suggesting that unusual responses were not due to insufficient plasma PCSK9 blockade. Multiple logistic regression analysis identified clinical FH (OR 2.9, 95% CI 1.27-7.24) and no ezetimibe therapy (OR 0.334, 95% CI 0.150-0.728) as factors related to true unusual response. Conclusions Unusual responses to PCSK9i in a clinical cohort are more common than reported in clinical trials. Of the suspected unusual responders, nearly half were the result of adherence issues, and thus careful medication reconciliation should be the first step in diagnosing an unusual response.
Collapse
|
21
|
Shapiro MD, Minnier J, Tavori H, Kassahun H, Flower A, Somaratne R, Fazio S. Relationship Between Low-Density Lipoprotein Cholesterol and Lipoprotein(a) Lowering in Response to PCSK9 Inhibition With Evolocumab. J Am Heart Assoc 2020; 8:e010932. [PMID: 30755061 PMCID: PMC6405654 DOI: 10.1161/jaha.118.010932] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Beyond their potent LDL (low‐density lipoprotein) cholesterol (LDL‐C)–lowering efficacy (50–60%), PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors also reduce Lp(a) (lipoprotein[a]) levels by 25% to 30%, suggesting a 2:1 response ratio. We aimed to characterize the relationship between LDL‐C and Lp(a) lowering by evolocumab, a PCSK9 inhibitor, in a large clinical trial population and to determine the prevalence of concordant/discordant LDL‐C and Lp(a) responses to PCSK9 inhibition. Methods and Results Data were analyzed from 4 randomized, 12‐week, multicenter, phase 3 evolocumab trials. Patients with familial hypercholesterolemia, nonfamilial hypercholesterolemia, or statin intolerance participated in the trials. The main measure was the degree of concordance or discordance of LDL‐C and Lp(a) in response to PCSK9 inhibition; concordant response was defined as LDL‐C reduction >35% and Lp(a) reduction >10%. The study cohort comprised 895 patients (438 female; median age: 59.0 years [interquartile range: 51–66 years]). Baseline mean level of LDL‐C was 133.6 mg/dL (SE: 1.7) and median Lp(a) level was 46.4 mg/dL (interquartile range: 18.4–82.4 mg/dL). A discordant response was observed in 165 (19.7%) patients. With these cutoffs, the prevalence of discordance was higher when considering baseline Lp(a) concentrations >30 mg/dL (26.5%) or >50 mg/dL (28.6%). Conclusions We demonstrate high prevalence of discordance in LDL‐C and Lp(a) reduction in response to evolocumab, particularly when considering higher baseline Lp(a) concentrations, indicating the possibility of alternative pathways beyond LDLR (LDL receptor)–mediated clearance involved in Lp(a) reduction by evolocumab. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifiers: NCT01763827, NCT01763866, NCT01763905, NCT01763918. See Editorial by Nestel
Collapse
Affiliation(s)
- Michael D. Shapiro
- Knight Cardiovascular InstituteCenter for Preventive CardiologyOregon Health & Science UniversityPortlandOR
| | - Jessica Minnier
- Knight Cardiovascular InstituteCenter for Preventive CardiologyOregon Health & Science UniversityPortlandOR
- OHSU‐PSU School of Public HealthOregon Health & Science UniversityPortlandOR
| | - Hagai Tavori
- Knight Cardiovascular InstituteCenter for Preventive CardiologyOregon Health & Science UniversityPortlandOR
| | | | | | | | - Sergio Fazio
- Knight Cardiovascular InstituteCenter for Preventive CardiologyOregon Health & Science UniversityPortlandOR
| |
Collapse
|
22
|
Croyal M, Blanchard V, Ouguerram K, Chétiveaux M, Cabioch L, Moyon T, Billon-Crossouard S, Aguesse A, Bernardeau K, Le May C, Flet L, Lambert G, Hadjadj S, Cariou B, Krempf M, Nobécourt-Dupuy E. VLDL (Very-Low-Density Lipoprotein)-Apo E (Apolipoprotein E) May Influence Lp(a) (Lipoprotein [a]) Synthesis or Assembly. Arterioscler Thromb Vasc Biol 2020; 40:819-829. [DOI: 10.1161/atvbaha.119.313877] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Objective:
To clarify the association between PCSK9 (proprotein convertase subtilisin/kexin type 9) and Lp(a) (lipoprotein [a]), we studied Lp(a) kinetics in patients with loss-of-function and gain-of-function
PCSK9
mutations and in patients in whom extended-release niacin reduced Lp(a) and PCSK9 concentrations.
Approach and Results:
Six healthy controls, 9 heterozygous patients with familial hypercholesterolemia (5 with low-density lipoprotein receptor [
LDLR
] mutations and 4 with
PCSK9
gain-of-function mutations) and 3 patients with heterozygous dominant-negative
PCSK9
loss-of-function mutations were included in the preliminary study. Eight patients were enrolled in a second study assessing the effects of 2 g/day extended-release niacin. Apolipoprotein kinetics in VLDL (very-low-density lipoprotein), LDL (low-density lipoprotein), and Lp(a) were studied using stable isotope techniques. Plasma Lp(a) concentrations were increased in
PCSK9
-gain-of-function and familial hypercholesterolemia-
LDLR
groups compared with controls and
PCSK9
-loss-of-function groups (14±12 versus 5±4 mg/dL;
P
=0.04), but no change was observed in Lp(a) fractional catabolic rate. Subjects with
PCSK9
-loss-of-function mutations displayed reduced apoE (apolipoprotein E) concentrations associated with a VLDL-apoE absolute production rate reduction. Lp(a) and VLDL-apoE absolute production rates were correlated (
r
=0.50;
P
<0.05). ApoE-to-apolipoprotein (a) molar ratios in Lp(a) increased with plasma Lp(a) (
r
=0.96;
P
<0.001) but not with PCSK9 levels. Extended-release niacin-induced reductions in Lp(a) and VLDL-apoE absolute production rate were correlated (
r
=0.83;
P
=0.015). In contrast, PCSK9 reduction (−35%;
P
=0.008) was only correlated with that of VLDL-apoE absolute production rate (
r
=0.79;
P
=0.028).
Conclusions:
VLDL-apoE production could determine Lp(a) production and/or assembly. As PCSK9 inhibitors reduce plasma apoE and Lp(a) concentrations, apoE could be the link between PCSK9 and Lp(a).
Collapse
Affiliation(s)
- Mikaël Croyal
- From the NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, France (M. Croyal, K.O., S.B.-C., A.A., M.K.)
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
| | - Valentin Blanchard
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothrombose Réunion Océan Indien (DéTROI), Plateforme CYROI, Saint-Denis de La Réunion, France (V.B., G.L.)
| | - Khadija Ouguerram
- From the NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, France (M. Croyal, K.O., S.B.-C., A.A., M.K.)
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
| | - Maud Chétiveaux
- L’institut du thorax, INSERM, CNRS, University of Nantes, France (M. Chétiveaux, C.L.M.)
| | - Léa Cabioch
- Biogenouest-Corsaire platform, Saint Gilles, France (L.C.)
| | - Thomas Moyon
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
| | - Stéphanie Billon-Crossouard
- From the NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, France (M. Croyal, K.O., S.B.-C., A.A., M.K.)
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
| | - Audrey Aguesse
- From the NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, France (M. Croyal, K.O., S.B.-C., A.A., M.K.)
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
| | - Karine Bernardeau
- P2R «Production de protéines recombinantes», CRCINA, SFR-Santé, INSERM, CNRS, UNIV Nantes, CHU Nantes, France (K.B.)
| | - Cédric Le May
- L’institut du thorax, INSERM, CNRS, University of Nantes, France (M. Chétiveaux, C.L.M.)
| | - Laurent Flet
- Pharmacy Department, Nantes University Hospital, France (L.F.)
| | - Gilles Lambert
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothrombose Réunion Océan Indien (DéTROI), Plateforme CYROI, Saint-Denis de La Réunion, France (V.B., G.L.)
| | - Samy Hadjadj
- L’institut du thorax, INSERM, CNRS, University of Nantes, CHU Nantes, France (S.H., B.C.)
| | - Bertrand Cariou
- L’institut du thorax, INSERM, CNRS, University of Nantes, CHU Nantes, France (S.H., B.C.)
| | - Michel Krempf
- From the NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, France (M. Croyal, K.O., S.B.-C., A.A., M.K.)
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
- ELSAN, clinique Bretéché, Nantes, France (M.K.)
| | | |
Collapse
|
23
|
Khoury E, Brisson D, Gaudet D. Preclinical discovery and development of evolocumab for the treatment of hypercholesterolemia. Expert Opin Drug Discov 2020; 15:403-414. [DOI: 10.1080/17460441.2020.1704728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Etienne Khoury
- Clinical Lipidology and Rare Lipid Disorders Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Québec, Canada
| | - Diane Brisson
- Clinical Lipidology and Rare Lipid Disorders Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Québec, Canada
| | - Daniel Gaudet
- Clinical Lipidology and Rare Lipid Disorders Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Québec, Canada
- Lipid Clinic, Chicoutimi Hospital, Chicoutimi, Québec, Canada
| |
Collapse
|
24
|
Apolipoprotein(a) phenotype determines the correlations of lipoprotein(a) and proprotein convertase subtilisin/kexin type 9 levels in patients with potential familial hypercholesterolemia. Atherosclerosis 2019; 277:477-482. [PMID: 30270088 DOI: 10.1016/j.atherosclerosis.2018.08.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 06/01/2018] [Accepted: 08/17/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND AIMS The aim of this study is to investigate the relation between lipoprotein(a) [Lp(a)] and proprotein convertase subtilisin/kexin type 9 (PCSK9) concentrations, and their complex, in patients with potential familial hypercholesterolemia (FH), depending on apo(a) phenotype. METHODS The study included 205 patients with total cholesterol (TC) > 7.5 mmol/L and/or low density lipoprotein cholesterol (LDL-C)>4.9 mmol/L, 32 (15%) patients suffered from ischemic heart disease (IHD), 64 were taking statins. The diagnosis of FH was estimated according to the Dutch Lipid Clinics Network criteria. Lipid parameters, apoB-containing lipoprotein subfractions, Lp(a), PCSK9, Lp(a)-PCSK9 complex levels and apo(a) phenotype were determined. Depending on the apo(a) phenotype, all patients were divided into 2 groups: with high molecular weight (HMW) (n = 145) and low molecular weight (LMW) (n = 60) apo(a) phenotype. RESULTS The groups were comparable by all major clinical characteristics and biochemical parameters. In the whole group, PCSK9 concentration correlated with age, statins intake, Lp(a), TC and TG levels. Correlation between Lp(a) and PCSK9 levels was found only in the LMW apo(a) phenotype group independently of statins intake (r = 0.46, p < 0.001). Associations between Lp(a)-PCSK9 complex and large subfractions of intermediate (r = 0.30) and low-density lipoproteins (r = 0.30, p < 0.05 for both) were observed, with more significance in group 2 (r = 0.59, p < 0.005 and r = 0.40, p < 0.05, respectively). CONCLUSIONS In patients with potential familial hypercholesterolemia, positive correlations between concentrations of Lp(a) and PCSK9, as well as of Lp(a)-PCSK9 plasma complex with large subfractions of intermediate and low-density lipoproteins (IDL-1 and LDL-C), were determined by the LMW apo(a) phenotype.
Collapse
|
25
|
Tsimikas S, Fazio S, Ferdinand KC, Ginsberg HN, Koschinsky ML, Marcovina SM, Moriarty PM, Rader DJ, Remaley AT, Reyes-Soffer G, Santos RD, Thanassoulis G, Witztum JL, Danthi S, Olive M, Liu L. NHLBI Working Group Recommendations to Reduce Lipoprotein(a)-Mediated Risk of Cardiovascular Disease and Aortic Stenosis. J Am Coll Cardiol 2019; 71:177-192. [PMID: 29325642 DOI: 10.1016/j.jacc.2017.11.014] [Citation(s) in RCA: 347] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
Pathophysiological, epidemiological, and genetic studies provide strong evidence that lipoprotein(a) [Lp(a)] is a causal mediator of cardiovascular disease (CVD) and calcific aortic valve disease (CAVD). Specific therapies to address Lp(a)-mediated CVD and CAVD are in clinical development. Due to knowledge gaps, the National Heart, Lung, and Blood Institute organized a working group that identified challenges in fully understanding the role of Lp(a) in CVD/CAVD. These included the lack of research funding, inadequate experimental models, lack of globally standardized Lp(a) assays, and inadequate understanding of the mechanisms underlying current drug therapies on Lp(a) levels. Specific recommendations were provided to facilitate basic, mechanistic, preclinical, and clinical research on Lp(a); foster collaborative research and resource sharing; leverage expertise of different groups and centers with complementary skills; and use existing National Heart, Lung, and Blood Institute resources. Concerted efforts to understand Lp(a) pathophysiology, together with diagnostic and therapeutic advances, are required to reduce Lp(a)-mediated risk of CVD and CAVD.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Vascular Medicine Program, Sulpizio Cardiovascular Center, Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, California.
| | - Sergio Fazio
- Oregon Health & Science University, Portland, Oregon
| | | | - Henry N Ginsberg
- College of Physicians and Surgeons, Columbia University, New York, New York
| | - Marlys L Koschinsky
- Robarts Research Institute and Department of Physiology & Pharmacology Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | | | | | - Daniel J Rader
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alan T Remaley
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Raul D Santos
- Heart Institute (InCor) University of Sao Paulo Medical School Hospital and Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | | | - Joseph L Witztum
- Division of Endocrinology, Department of Medicine, University of California San Diego, La Jolla, California
| | - Simhan Danthi
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Michelle Olive
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lijuan Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
26
|
Warden BA, Minnier J, Watts GF, Fazio S, Shapiro MD. Impact of PCSK9 inhibitors on plasma lipoprotein(a) concentrations with or without a background of niacin therapy. J Clin Lipidol 2019; 13:580-585. [DOI: 10.1016/j.jacl.2019.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/18/2019] [Accepted: 04/20/2019] [Indexed: 12/24/2022]
|
27
|
Abstract
PURPOSE OF REVIEW The use of therapeutic monoclonal antibodies to target proprotein convertase subtilisin/kexin type 9 (PCSK9) represents a novel approach to the management of hypercholesteremia and prevention of atherosclerotic cardiovascular disease. We review the most recent literature relevant to PCSK9 inhibition with emphasis on how recent results and ongoing trials have and will continue to shape the use of this new therapeutic class in preventive cardiology. RECENT FINDINGS PCSK9 inhibitors reduce plasma lipoprotein(a) concentrations but a mechanistic understanding remains elusive. Evaluation of evolocumab for use in patients without prior myocardial infarction or stroke is underway (NCT03872401). Concerns regarding the cost-effectiveness of PCSK9 inhibitors have continued to thwart access to these drugs, though innovative models of care delivery and price reductions have improved this situation. Inclisiran, a small interfering ribonucleic acid (siRNA), reduces translation of PCSK9 and demonstrates more durable reductions in low-density lipoprotein-cholesterol (LDL-C). It is currently evaluated in the context of a phase III cardiovascular outcome trial in patients with established vascular disease (NCT03705234). SUMMARY The current scope of PCSK9 inhibitor therapy in preventive cardiology is limited to patients with familial hypercholesterolemia and/or established atherosclerotic cardiovascular disease. Future cardiovascular outcome trial results with PCSK9 blocking antibodies in primary prevention and with siRNA to PCSK9 in secondary prevention, improved understanding of the drivers of lipoprotein(a) reduction with PCSK9 inhibition, and cost-effectiveness will determine the future role of this therapeutic class.
Collapse
|
28
|
The PCSK9 revolution: Current status, controversies, and future directions. Trends Cardiovasc Med 2019; 30:179-185. [PMID: 31151804 DOI: 10.1016/j.tcm.2019.05.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 02/07/2023]
Abstract
Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) has revolutionized our understanding of cholesterol homeostasis and added to our arsenal against atherosclerotic cardiovascular disease (ASCVD). In a span of approximately 15 years, PCSK9 has morphed from an esoteric and rare cause of familial hypercholesterolemia (FH) into the most efficient cholesterol-lowering target ever known, with the completion of two large scale cardiovascular outcome trials showing positive results. Current Food and Drug Administration (FDA) approved modalities to inhibit PCSK9 are in the form of monoclonal antibodies which display an unparalleled degree of low-density lipoprotein cholesterol (LDL-C) lowering and expand upon the notion that lower LDL-C is better for ASCVD risk reduction. However, the accelerated pace of discovery and therapeutic development has left large gaps in our knowledge regarding the physiology and function of PCSK9. The aim of this review is to provide context to the discovery, history, treatment and current status of PCSK9 and its therapeutic inhibitors and highlight areas of controversy and future directions.
Collapse
|
29
|
O’Donoghue ML, Fazio S, Giugliano RP, Stroes ES, Kanevsky E, Gouni-Berthold I, Im K, Lira Pineda A, Wasserman SM, Češka R, Ezhov MV, Jukema JW, Jensen HK, Tokgözoğlu SL, Mach F, Huber K, Sever PS, Keech AC, Pedersen TR, Sabatine MS. Lipoprotein(a), PCSK9 Inhibition, and Cardiovascular Risk. Circulation 2019; 139:1483-1492. [DOI: 10.1161/circulationaha.118.037184] [Citation(s) in RCA: 354] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Michelle L. O’Donoghue
- TIMI Study Group, Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA (M.L.O., R.P.G., E.K., K.A.I., M.S.S.)
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland (S.F.)
| | - Robert P. Giugliano
- TIMI Study Group, Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA (M.L.O., R.P.G., E.K., K.A.I., M.S.S.)
| | - Erik S.G. Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands (E.S.G.S.)
| | - Estella Kanevsky
- TIMI Study Group, Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA (M.L.O., R.P.G., E.K., K.A.I., M.S.S.)
| | - Ioanna Gouni-Berthold
- Polyclinic for Endocrinology, Diabetes and Preventive Medicine, University of Cologne, Germany (I.G.-B.)
| | - KyungAh Im
- TIMI Study Group, Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA (M.L.O., R.P.G., E.K., K.A.I., M.S.S.)
| | | | | | - Richard Češka
- Center for Preventive Cardiology, 3rd Internal Medicine Clinic, University General Hospital and Charles University 1st Medical Faculty, Prague, Czech Republic (R.C.)
| | - Marat V. Ezhov
- National Cardiology Research Center, Moscow, Russia (M.V.E.)
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, The Netherlands (J.W.J.)
| | - Henrik K. Jensen
- Department of Cardiology, Aarhus University Hospital and Department of Clinical Medicine, Health, Aarhus University, Denmark (H.K.J.)
| | | | - François Mach
- Cardiology Division, University of Geneva, Switzerland (F.M.)
| | - Kurt Huber
- 3rd Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminenhospital and Sigmund Freud University, Medical Faculty, Vienna, Austria (K.H.)
| | | | - Anthony C. Keech
- Sydney Medical School, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Australia (A.C.K.)
| | - Terje R. Pedersen
- Oslo University Hospital, Ullevål and Medical Faculty, University of Oslo, Norway (T.R.P.)
| | - Marc S. Sabatine
- TIMI Study Group, Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA (M.L.O., R.P.G., E.K., K.A.I., M.S.S.)
| |
Collapse
|
30
|
Reiner Ž. Can Lp(a) Lowering Against Background Statin Therapy Really Reduce Cardiovascular Risk? Curr Atheroscler Rep 2019; 21:14. [PMID: 30847681 DOI: 10.1007/s11883-019-0773-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The association between elevated plasma levels of lipoprotein (a) [Lp(a)] and atherosclerotic cardiovascular disease (ASCVD) has been discussed for many years. Recent genetic findings have confirmed that elevated Lp(a) similar to elevated LDL-cholesterol (LDL-C) might be causally related to premature ASCVD. Lp(a) is relatively refractory to lifestyle interventions. The results of studies with statins and their possible effect on Lp(a) are conflicting. Specific Lp(a) apheresis is used as a treatment against background statin therapy and can decrease Lp(a). The purpose of this review is to discuss whether new drugs which decrease Lp(a) can prevent ASCVD and decrease ASCVD mortality when applied in addition to statins. RECENT FINDINGS Some new LDL-C-lowering drugs such as mipomersen and lomitapide decrease elevated Lp(a) in addition to statins but they have some unpleasant adverse effects. Recently, an antisense oligonucleotide against apo(a), AKCEA-APO(a)Rx, has been shown to selectively decrease Lp(a). The most recent advance in LDL-C lowering are PCSK9 inhibitors. Alirocumab and evolocumab do not only significantly reduce LDL-C on top of maximally tolerated statin therapy and prevent ASCVD events, but also further decrease Lp(a). There is no data to indicate whether mipomersen, lomitapide, or IONIS-APO(a)-LRx decrease ASCVD events and mortality. Conclusive evidence is still lacking as to whether the treatment with PCSK9 inhibitors against background statin therapy actually additionally reduces ASCVD risk due to the lowering of Lp(a) or simply due to lowering LDL-C to levels much lower than high-intensity statin treatment as monotherapy. Ongoing trials will probably provide an answer to these questions.
Collapse
Affiliation(s)
- Željko Reiner
- Department of Internal Medicine, University Hospital Centre Zagreb, School of Medicine, University of Zagreb, Kispaticeva 12, 10000, Zagreb, Croatia.
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Our primary objective is to review the most recent findings on the biology of PCSK9 and on two key aspects of PCSK9 inhibition beyond LDL control of great clinical relevance: the regulation of lipoprotein (a) circulating levels by PCSK9 inhibitors and the putative diabetogenic effects of these novel therapies. RECENT FINDINGS The reality of two distinct extracellular and intracellular pathways by which PCSK9 decreases the abundance of the LDLR at the surface of many cell types, most importantly hepatocytes, has recently been established. In contrast, the exact mechanisms by which PCSK9 inhibitors lower the circulating levels of lipoprotein (a) remain a point of major dispute. Despite strong indications from genetic studies that PCSK9 inhibition should increase diabetes risk, no such effect has been observed in clinical trials, and in-vitro and in-vivo studies do not clarify this issue. SUMMARY The trafficking pathways by which PCSK9 enhance LDLR degradation via the endolysosomal extracellular route or via the Golgi-lysosomal intracellular route remain to be fully elucidated. The mechanisms by which PCSK9 inhibitors reduce lipoprotein (a) also merit additional research efforts. The role of PCSK9 on glucose metabolism should likewise be studied in depth.
Collapse
|
32
|
Abstract
Clinical trials have unequivocally shown that inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) efficaciously and safely prevents cardiovascular events by lowering levels of LDL cholesterol. PCSK9 in the circulation is derived mainly from the liver, but the protein is also expressed in the pancreas, the kidney, the intestine and the central nervous system. Although PCSK9 modulates cholesterol metabolism by regulating LDL receptor expression in the liver, in vitro and in vivo studies have suggested that PCSK9 is involved in various other physiological processes. Although therapeutic PCSK9 inhibition could theoretically have undesired effects by interfering with these non-cholesterol-related processes, studies of individuals with genetically determined reduced PCSK9 function and clinical trials of PCSK9 inhibitors have not revealed clinically meaningful adverse consequences of almost completely eradicating PCSK9 from the circulation. The clinical implications of PCSK9 functions beyond lipid metabolism in terms of wanted or unwanted effects of therapeutic PCSK9 inhibition therefore appear to be limited. The objective of this Review is to describe the physiological role of PCSK9 beyond the LDL receptor to provide a rational basis for monitoring the effects of PCSK9 inhibition as these drugs gain traction in the clinic.
Collapse
Affiliation(s)
| | - Gilles Lambert
- Inserm UMR 1188 DéTROI, Université de La Réunion, Saint-Denis de La Réunion, France
| | - Bertrand Cariou
- L'institut du thorax, INSERM, CNRS, Université de Nantes, CHU Nantes, Nantes, France
| | - G Kees Hovingh
- Department of Vascular Medicine, Academisch Medisch Centrum, Amsterdam, Netherlands.
| |
Collapse
|
33
|
Sbrana F, Bigazzi F, Dal Pino B, Toma M, Ripoli A, Sampietro T. Elusive Therapeutic Effect of PCSK9 Inhibitors on Lipoprotein(a) Levels. Ther Apher Dial 2018; 23:385-386. [DOI: 10.1111/1744-9987.12766] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/03/2018] [Accepted: 10/09/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Francesco Sbrana
- Lipoapheresis Unit ‐ Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias Pisa Italy
| | - Federico Bigazzi
- Lipoapheresis Unit ‐ Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias Pisa Italy
| | - Beatrice Dal Pino
- Lipoapheresis Unit ‐ Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias Pisa Italy
| | - Maddalena Toma
- Lipoapheresis Unit ‐ Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias Pisa Italy
| | - Andrea Ripoli
- Bioengineering DepartmentFondazione Toscana “Gabriele Monasterio” Pisa Italy
| | - Tiziana Sampietro
- Lipoapheresis Unit ‐ Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias Pisa Italy
| |
Collapse
|
34
|
Ruscica M, Simonelli S, Botta M, Ossoli A, Lupo MG, Magni P, Corsini A, Arca M, Pisciotta L, Veglia F, Franceschini G, Ferri N, Calabresi L. Plasma PCSK9 levels and lipoprotein distribution are preserved in carriers of genetic HDL disorders. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:991-997. [DOI: 10.1016/j.bbalip.2018.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 04/18/2018] [Accepted: 05/27/2018] [Indexed: 01/27/2023]
|
35
|
Ma KL, Gong TK, Hu ZB, Zhang Y, Wang GH, Liu L, Chen PP, Lu J, Lu CC, Liu BC. Lipoprotein(a) accelerated the progression of atherosclerosis in patients with end-stage renal disease. BMC Nephrol 2018; 19:192. [PMID: 30071823 PMCID: PMC6090984 DOI: 10.1186/s12882-018-0986-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 07/19/2018] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Increased plasma level of lipoprotein(a) (Lpa) is a risk factor of cardiovascular diseases. This study aimed to explore the role of Lpa in the progression of atherosclerosis in patients with end-stage renal disease (ESRD) and to investigate whether its potential mechanism is mediated by CXC chemokine ligand 16 (CXCL16) and low-density lipoprotein receptor (LDLr). METHODS This is a retrospective clinical study. From January 2015 to April 2016, forty-six ESRD patients from Danyang First People's Hospital were investigated. The patients were grouped according to their plasma Lpa levels: control group (Lpa < 300 mg/l, n = 23) and high Lpa group (Lpa ≥ 300 mg/l, n = 23). ESRD Patients with acute infective diseases, cancer, and/or chronic active hepatitis were excluded. Biochemical indexes and lipid profiles of the patients were measured. Surgically removed tissues from the radial arteries of ESRD patients receiving arteriovenostomy were used for the preliminary evaluation of atherosclerosis. Haematoxylin-eosin (HE) and filipin staining were used to observe foam cell formation. Protein expression levels of Lpa, CXCL16, and LDLr were detected by immunohistochemistry staining and immunofluorescent staining. RESULTS There was more foam cell formation and cholesterol accumulation in the radial arteries of the high Lpa group than in those of the control group. Furthermore, the expression levels of Lpa, CXCL16, and LDLr were significantly increased in the radial arteries of the high Lpa group. Correlation analyses showed that the protein expression levels of Lpa (r = 0.72, P < 0.01), LDLr (r = 0.54, P < 0.01), and CXCL16 (r = 0.6, P < 0.01) in the radial arteries of ESRD patients were positively correlated with the plasma Lpa levels. Further analyses showed that the co-expression of Lpa with LDLr or CXCL16 was increased in the high Lpa group. CONCLUSIONS High plasma Lpa levels accelerated the progression of atherosclerosis in ESRD through inducing Lpa accumulation in the arteries, which was associated with LDLr and CXCL16. These two lipoproteins could both be major lipoprotein components that regulate the entry of Lpa into arterial cells.
Collapse
Affiliation(s)
- Kun Ling Ma
- Institute of Nephrology, Zhong Da Hospital, Medical School of Southeast University, NO. 87, Ding Jia Qiao Road, Nang Jing City, 210009 Jiang Su Province China
| | - Tie Kai Gong
- Institute of Nephrology, Zhong Da Hospital, Medical School of Southeast University, NO. 87, Ding Jia Qiao Road, Nang Jing City, 210009 Jiang Su Province China
- Renal Department, Danyang First People’s Hospital, Jiang Su Province, China
| | - Ze Bo Hu
- Institute of Nephrology, Zhong Da Hospital, Medical School of Southeast University, NO. 87, Ding Jia Qiao Road, Nang Jing City, 210009 Jiang Su Province China
| | - Yang Zhang
- Institute of Nephrology, Zhong Da Hospital, Medical School of Southeast University, NO. 87, Ding Jia Qiao Road, Nang Jing City, 210009 Jiang Su Province China
| | - Gui Hua Wang
- Institute of Nephrology, Zhong Da Hospital, Medical School of Southeast University, NO. 87, Ding Jia Qiao Road, Nang Jing City, 210009 Jiang Su Province China
| | - Liang Liu
- Institute of Nephrology, Zhong Da Hospital, Medical School of Southeast University, NO. 87, Ding Jia Qiao Road, Nang Jing City, 210009 Jiang Su Province China
| | - Pei Pei Chen
- Institute of Nephrology, Zhong Da Hospital, Medical School of Southeast University, NO. 87, Ding Jia Qiao Road, Nang Jing City, 210009 Jiang Su Province China
| | - Jian Lu
- Institute of Nephrology, Zhong Da Hospital, Medical School of Southeast University, NO. 87, Ding Jia Qiao Road, Nang Jing City, 210009 Jiang Su Province China
| | - Chen Chen Lu
- Institute of Nephrology, Zhong Da Hospital, Medical School of Southeast University, NO. 87, Ding Jia Qiao Road, Nang Jing City, 210009 Jiang Su Province China
| | - Bi Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Medical School of Southeast University, NO. 87, Ding Jia Qiao Road, Nang Jing City, 210009 Jiang Su Province China
| |
Collapse
|
36
|
Bhatnagar D, Shiraz A. Modulation of cardiovascular risk factors. Curr Opin Lipidol 2018; 29:269-270. [PMID: 29715242 DOI: 10.1097/mol.0000000000000516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Deepak Bhatnagar
- Diabetes Centre, Royal Oldham Hospital, Oldham
- Division of Diabetes, Endocrinology & Gastroenterology, Univesrity of Manchester, Manchester
- University of Salford, Salford, UK
| | | |
Collapse
|
37
|
Abstract
Unknown 15 years ago, PCSK9 (proprotein convertase subtilisin/kexin type 9) is now common parlance among scientists and clinicians interested in prevention and treatment of atherosclerotic cardiovascular disease. What makes this story so special is not its recent discovery nor the fact that it uncovered previously unknown biology but rather that these important scientific insights have been translated into an effective medical therapy in record time. Indeed, the translation of this discovery to novel therapeutic serves as one of the best examples of how genetic insights can be leveraged into intelligent target drug discovery. The PCSK9 saga is unfolding quickly but is far from complete. Here, we review major scientific understandings as they relate to the role of PCSK9 in lipoprotein metabolism and atherosclerotic cardiovascular disease and the impact that therapies designed to inhibit its action are having in the clinical setting.
Collapse
Affiliation(s)
- Michael D Shapiro
- From the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland
| | - Hagai Tavori
- From the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland
| | - Sergio Fazio
- From the Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland.
| |
Collapse
|
38
|
Wójcik C. Incorporation of PCSK9 inhibitors into prevention of atherosclerotic cardiovascular disease. Postgrad Med 2017; 129:801-810. [PMID: 28879791 DOI: 10.1080/00325481.2017.1376570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Primary and secondary prevention of atherosclerotic cardiovascular disease (ASCVD) has become recently more complex than ever, leaving the clinicians perplexed with outdated guidelines and emerging evidence about new LDL-C lowering therapies. 2013 American College of Cardiology (ACC)/American Heart Association (AHA) guidelines have focused on high intensity statin therapy for specific groups of patients, while abandoning long established LDL-C goals, a strategy which no longer seems valid. PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors have emerged as the add-on therapy on top of statins and/or ezetimibe for the treatment of hypercholesterolemia and ASCVD prevention. In several clinical trials, PCSK9 inhibitors have demonstrated their safety and robust LDL-C-lowering power. One completed cardiovascular (CV) outcomes trial (FOURIER; Further Cardiovascular Outcomes Research with PCSK9 Inhibitions in Subjects with Elevated Risk) has demonstrated that PCSK9 inhibition reduces rates of CV death as well as non-fatal stroke and MI, while another major CV outcome trial is under way (ODYSSEY-OUTCOMES). Several trials studying CV benefits of novel LDL-C-lowering therapies are also being conducted. Prompt revision of ACC/AHA guidelines is necessary. In the meantime, physicians need to use clinical judgment integrating the most recent evidence into their practice.
Collapse
Affiliation(s)
- Cezary Wójcik
- a Department of Family Medicine , Oregon Health and Science University , Portland , OR , USA
| |
Collapse
|