1
|
van Bruggen FH, Diamond DM. Is Targeting LDL-C Levels Below 70 mg/dL Beneficial for Cardiovascular and Overall Health? A Critical Examination of the Evidence. J Clin Med 2025; 14:3569. [PMID: 40429563 PMCID: PMC12112069 DOI: 10.3390/jcm14103569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Revised: 05/17/2025] [Accepted: 05/18/2025] [Indexed: 05/29/2025] Open
Abstract
Over the past two decades, cardiovascular disease (CVD) prevention guidelines have progressively lowered LDL-C targets to <70 mg/dL for high-risk individuals based on the assumption of a linear relationship between LDL-C levels and CVD risk. However, the available evidence challenges this premise. Multiple studies demonstrate a weak or inconsistent association between LDL-C levels and atherosclerosis progression at the individual patient-level. Systematic reviews supporting the linearity assumption have notable limitations, including extrapolation beyond observed LDL-C ranges and potential ecological fallacy, as meta-regression analyses rely on study-level data, while patient-level data within the same trials often show no association between LDL-C reduction and CVD outcomes. Moreover, randomized controlled trials explicitly designed to assess LDL-C targets have yielded inconclusive and biased results. LDL-C itself is a heterogeneous marker, with particle size and composition influencing its atherogenicity. The cardiovascular benefits of lipid-lowering therapies may arise in part from pleiotropic effects unrelated to LDL-C lowering. Additionally, several studies indicate that higher LDL-C levels are paradoxically associated with longevity in elderly populations that is equal to or even greater than that of the general population. Collectively, this body of evidence raises questions about the validity of current LDL-C targets < 70 mg/dL in high-risk patients.
Collapse
Affiliation(s)
- Folkert H. van Bruggen
- Department of Primary and Long-Term Care, University Medical Centre Groningen, University of Groningen, P.O. Box 196, 9700 AD Groningen, The Netherlands
| | - David M. Diamond
- Department of Psychology, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
2
|
Corsini A, Ginsberg HN, Chapman MJ. Therapeutic PCSK9 targeting: Inside versus outside the hepatocyte? Pharmacol Ther 2025; 268:108812. [PMID: 39947256 DOI: 10.1016/j.pharmthera.2025.108812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
As a major regulator of LDL receptor (LDLR) activity and thus of LDL-cholesterol (LDL-C) levels, proprotein convertase subtilisin/kexin type 9 (PCSK9) represents an obvious therapeutic target for lipid lowering. The PCSK9 inhibitors, alirocumab and evolocumab, are human monoclonal antibodies (mAbs) that act outside the cell by complexing circulating PCSK9 and thus preventing its binding to the LDLR. In contrast, inclisiran, a small interfering RNA (siRNA), inhibits hepatic synthesis of PCSK9, thereby resulting in reduced amounts of the protein inside and outside the cell. Both approaches result in decreased plasma LDL-C concentrations and improved cardiovascular outcomes. Marginally superior LDL-C reduction (≈ 60 %) is achieved with mAbs as compared to the siRNA (≈ 50 %); head-to-head comparisons are required to confirm between-class differences in efficacy. Both drug classes have shown variability in LDL-C lowering response between individuals in waterfall analyses. Whereas mAb-mediated inhibition leads to a compensatory increase in plasma PCSK9 levels, siRNA treatment reduces them. These agents differ in their pharmacokinetic and pharmacodynamic features, which may translate into distinct clinical opportunities under acute (e.g. acute coronary syndromes) as compared to chronic conditions. Both drug classes provide additional reduction in LDL-C levels (up to 50 %) beyond those achieved with statin therapy, facilitating attainment of guideline-recommended LDL-C goals in high and very high-risk patients. Additional PCSK9 inhibitors, including an oral macrocyclic peptide, a small PCSK9 binding protein and a novel small molecule, plus hepatic gene editing of PCSK9, are under development. This review critically appraises pharmacological strategies to target PCSK9 either inside or outside the cell.
Collapse
Affiliation(s)
- Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy
| | - Henry N Ginsberg
- Irving Institute for Clinical and Translational Research, Columbia University, New York, USA
| | - M John Chapman
- Sorbonne University Medical Faculty, Lipidology and Cardiovascular Prevention Unit, Pitie-Salpetriere University Hospital, Paris, France.
| |
Collapse
|
3
|
Nicholls SJ, Nelson AJ. Can Vulnerable Plaque Imaging PREVENT Cardiovascular Events? JACC Cardiovasc Imaging 2025:S1936-878X(25)00038-5. [PMID: 40117397 DOI: 10.1016/j.jcmg.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 12/11/2024] [Indexed: 03/23/2025]
Affiliation(s)
- Stephen J Nicholls
- Victorian Heart Institute, Monash University, Clayton, Victoria, Australia.
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
4
|
Bucciarelli L, Andreini D, Stefanini G, Fiorina RM, Francone M, Catapano F, Lunati ME, Conte E, Marchetti D, Fiorina P. Pharmacological regression of atherosclerotic plaque in patients with type 2 diabetes. Pharmacol Res 2025; 213:107635. [PMID: 39921019 DOI: 10.1016/j.phrs.2025.107635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
Atherosclerosis of the coronary arteries continues to be one of the major global health burdens and acute coronary syndrome is responsible annually for at least 30 % of all deaths globally. Acute coronary syndrome may be the consequence of thrombus formation after erosion or rupture of obstructive or non-obstructive atherosclerotic plaque. The rupture of plaques is mostly caused by mechanical stress usually called cap fatigue. Vulnerable plaques are characterized by a softer atheromatous core and a thinner fibrous cap, with inflammation and hypercholesterolemia playing a crucial role in the atherothrombotic process. Based on animal studies that extend back to the 1920s, regression of atherosclerotic plaques in humans has just started to be considered and pursued. The idea that the human atherosclerotic plaques could regress at all met an important resistance over the decades; indeed, advanced plaques contain components, such as necrosis, calcification and fibrosis, which are hard to be removed. However, new animal models and imaging technics allowed a more complete and accurate quantitative assessment of plaque volume and are shedding new light on atherosclerosis regression. In this review, we are revisiting the existence of atherosclerosis regression in preclinical and clinical studies, with a focus on the latest mechanistic insights and on the newest pharmacological agents, particularly in patients with diabetes. Interestingly, we suggested that based on literature insights and preclinical studies, a combination of drugs to target hyperglycemia, dyslipidemia and inflammation may be desirable for a fast-track Pharmacological regression of atherosclerotic plaque in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Loredana Bucciarelli
- Pio Albergo Trivulzio, Italy; International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di scienze Biomediche e Cliniche, Università di Milano, Italy
| | - Daniele Andreini
- Division of University Cardiology, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Giulio Stefanini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy
| | - Roberta Maria Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di scienze Biomediche e Cliniche, Università di Milano, Italy
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy
| | - Federica Catapano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy
| | | | - Edoardo Conte
- Division of University Cardiology, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy
| | - Davide Marchetti
- Division of University Cardiology, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di scienze Biomediche e Cliniche, Università di Milano, Italy; Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy; Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Nicholls SJ, Nelson AJ. Achieving More Optimal Lipid Control with Non-Statin Lipid Lowering Therapy. Curr Atheroscler Rep 2025; 27:32. [PMID: 39954169 PMCID: PMC11829850 DOI: 10.1007/s11883-025-01280-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE OF REVIEW The use of statins has transformed approaches to the prevention of cardiovascular disease. However, many patients remain at a major risk of experiencing cardiovascular events, due to a range of factors including suboptimal control of low-density lipoprotein cholesterol (LDL-C). Accordingly, there is an ongoing need to develop additional strategies, beyond the use of statins, to achieve more effective reductions in cardiovascular risk. RECENT FINDINGS Genomic studies have implicated the causal role of LDL in atherosclerosis and identified that polymorphisms influencing factors involved in lipid metabolism influence both the level of LDL-C and cardiovascular risk. These findings have highlighted the potential for cardiovascular benefit from development of therapies targeting these factors and incremental benefit when used in combination with statins. Clinical trials have demonstrated that these new agents have favourable effects on both atherosclerotic plaque and cardiovascular events. Additional work has sought to improve intensification of statin therapy and adherence with lipid lowering therapy, to achieve more effective cardiovascular prevention via lipid lowering. Emerging therapies, beyond statins, have the potential to optimise lipid levels and play an effective role in the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Stephen J Nicholls
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne, Australia.
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne, Australia
| |
Collapse
|
6
|
Simony SB, Langsted A, Mortensen MB, Nordestgaard BG, Afzal S. Statin use is associated with less ST-elevation versus non-ST-elevation myocardial infarction in a nationwide study. Atherosclerosis 2024; 399:118625. [PMID: 39437595 DOI: 10.1016/j.atherosclerosis.2024.118625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/05/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND AIMS Statin therapy reduces myocardial infarction rate but whether it is associated with a shift of ST-elevation myocardial infarction (STEMI) towards non-ST-elevation myocardial infarction (non-STEMI) remains unknown. Thus, we tested the hypothesis that statin use is associated with less STEMI relative to non-STEMI in first-time myocardial infarction. METHODS In a nationwide study, including 66,896 patients with first-time myocardial infarction between 2010 and 2021, we obtained multivariable risk estimates for STEMI versus non-STEMI according to any statin use, cumulated statin use, and daily statin dose. Furthermore, we obtained hazard ratios for 60-day mortality (5545 deaths) following myocardial infarction according to type of infarction. RESULTS Odds ratios for STEMI versus non-STEMI were 0.81 (95 % CI:0.77-0.85) and 1.07 (1.01-1.13) in current and previous statin users compared to never statin users. Cumulated statin exposure yielded odds ratios of 0.96 (0.87-1.07) for <2 statin-years, 0.87 (0.79-0.95) for 2-4.9 statin-years, 0.80 (0.74-0.87) for 5-10 statin-years, and 0.75 (0.70-0.80) for >10 statin-years compared to never users. Corresponding odds ratios for statin dose intensity were 0.89 (0.84-0.95) for low-intensity, 0.77 (0.73-0.82) for moderate-intensity, and 0.70 (0.63-0.77) for high-intensity. Results were similar in multiple sensitivity analyses and using a cohort design. The hazard ratio for 60-day mortality after first-time STEMI versus non-STEMI was 2.24 (2.13-2.37). CONCLUSIONS In this nationwide study, prior statin use is associated with less STEMI relative to non-STEMI in a dose dependent manner. This indicates that statin therapy, in addition to reducing myocardial infarction event rates, also result in a less severe presentation of myocardial infarctions.
Collapse
Affiliation(s)
- Sofie B Simony
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 73, Entrance 7 4th floor N5, DK-2730 Herlev, Denmark; Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anne Langsted
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 73, Entrance 7 4th floor N5, DK-2730 Herlev, Denmark; Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Martin B Mortensen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 73, Entrance 7 4th floor N5, DK-2730 Herlev, Denmark; Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Shoaib Afzal
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 73, Entrance 7 4th floor N5, DK-2730 Herlev, Denmark; Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
7
|
Zhang X, Feng H, Han Y, Yuan X, Jiang M, Wang W, Gao L. Plaque Stabilization and Regression, from Mechanisms to Surveillance and Clinical Strategies. Rev Cardiovasc Med 2024; 25:459. [PMID: 39742242 PMCID: PMC11683705 DOI: 10.31083/j.rcm2512459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/13/2024] [Accepted: 08/02/2024] [Indexed: 01/03/2025] Open
Abstract
With advances in therapies to reduce cardiovascular events and improvements in coronary imaging, an increasing number of clinical trials have demonstrated that treatments to reduce cardiovascular events in coronary artery disease are associated with favorable effects on atherosclerotic plaque size and characteristics. It has been observed that various drugs may induce plaque regression and enhance plaque stability after plaque formation. Numerous clinical trials have been conducted to verify the occurrence of plaque stabilization and regression and their beneficial effects on cardiovascular events. Using invasive imaging techniques such as intravascular ultrasound (IVUS) and optical coherence tomography (OCT), researchers have been able to gather evidence supporting the existence of coronary plaque stabilization and regression. In this review, we explore the possible mechanisms of plaque stabilization and regression, summarize the imaging features of plaque stabilization and regression, and assemble the evidence from clinical studies that have used different features as observational endpoints.
Collapse
Affiliation(s)
- Xi Zhang
- Senior Department of Cardiology, Sixth Medical Center of Chinese PLA General Hospital, 100853 Beijing, China
- Medical School of Chinese PLA, 100853 Beijing, China
| | - Huanhuan Feng
- Medical School of Chinese PLA, 100853 Beijing, China
- Emergency Department, First Medical Center of Chinese PLA General Hospital, 100853 Beijing, China
| | - Yan Han
- Senior Department of Cardiology, Sixth Medical Center of Chinese PLA General Hospital, 100853 Beijing, China
- Medical School of Chinese PLA, 100853 Beijing, China
| | - Xiaohang Yuan
- Senior Department of Cardiology, Sixth Medical Center of Chinese PLA General Hospital, 100853 Beijing, China
- Medical School of Chinese PLA, 100853 Beijing, China
| | - Mengting Jiang
- Senior Department of Cardiology, Sixth Medical Center of Chinese PLA General Hospital, 100853 Beijing, China
- Medical School of Chinese PLA, 100853 Beijing, China
| | - Wei Wang
- Senior Department of Cardiology, Sixth Medical Center of Chinese PLA General Hospital, 100853 Beijing, China
| | - Lei Gao
- Senior Department of Cardiology, Sixth Medical Center of Chinese PLA General Hospital, 100853 Beijing, China
| |
Collapse
|
8
|
Davidson MH, Hsieh A, Dicklin MR, Maki KC. The Imperative to Enhance Cost-Effectiveness for Cardiovascular Therapeutic Development. JACC Basic Transl Sci 2024; 9:1029-1040. [PMID: 39297137 PMCID: PMC11405807 DOI: 10.1016/j.jacbts.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/06/2023] [Accepted: 12/14/2023] [Indexed: 09/21/2024]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide. Therapeutic agents, such as those that lower low-density lipoprotein cholesterol, have been a critical factor in mitigating CVD event risk and demonstrate the important role that drug discovery plays in reducing morbidity and mortality. However, rapidly rising development costs, diminishing returns, and an increasingly challenging regulatory environment have all contributed to a declining number of cardiovascular (CV) therapeutic agents entering the health care marketplace. For pharmaceutical companies, a traditional cardiovascular outcomes trial (CVOT) can be a major financial burden and impediment to CV agent development. They can take as long as a decade to conduct, delaying potential investment return while carrying risk of failure. For patients, lengthy CVOTs delay drug accessibility. Without cost-effective CVOTs, drug innovation may be compromised, with CV patients bearing the consequences. This paper reviews potential approaches for making CV drug development more cost-effective.
Collapse
Affiliation(s)
- Michael H Davidson
- University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
- NewAmsterdam Pharma, Naarden, the Netherlands
| | | | - Mary R Dicklin
- Midwest Biomedical Research, Addison, Illinois, and Boca Raton, Florida, USA
| | - Kevin C Maki
- Midwest Biomedical Research, Addison, Illinois, and Boca Raton, Florida, USA
- Indiana University School of Public Health, Bloomington, Indiana, USA
| |
Collapse
|
9
|
Bruoha S, Galli M, Sabouret P, Yosefy C, Taha L, Gragnano F, Savage MP, Shuvy M, Biondi-Zoccai G, Glikson M, Asher E. Atherosclerotic Plaque Erosion: Mechanisms, Clinical Implications, and Potential Therapeutic Strategies-A Review. J Cardiovasc Pharmacol 2024; 83:547-556. [PMID: 38421206 DOI: 10.1097/fjc.0000000000001554] [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: 11/27/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
Atherosclerosis is an insidious and progressive inflammatory disease characterized by the formation of lipid-laden plaques within the intima of arterial walls with potentially devastating consequences. While rupture of vulnerable plaques has been extensively studied, a distinct mechanism known as plaque erosion (PE) has gained recognition and attention in recent years. PE, characterized by the loss of endothelial cell lining in the presence of intact fibrous cap, contributes to a significant and growing proportion of acute coronary events. However, despite a heterogeneous substrate underlying coronary thrombosis, treatment remains identical. This article provides an overview of atherosclerotic PE characteristics and its underlying mechanisms, highlights its clinical implications, and discusses potential therapeutic strategies.
Collapse
Affiliation(s)
- Sharon Bruoha
- Department of Cardiology, Barzilai Medical Center, the Ben-Gurion University of the Negev, Israel
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Pierre Sabouret
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
- National College of French Cardiologists, 13 rue Niepce, 75014 Paris, France
| | - Chaim Yosefy
- Department of Cardiology, Barzilai Medical Center, the Ben-Gurion University of the Negev, Israel
| | - Louay Taha
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy
| | - Michael P Savage
- Division of Cardiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mony Shuvy
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy ; and
- Mediterranea Cardiocentro, Naples, Italy
| | - Michael Glikson
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Elad Asher
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| |
Collapse
|
10
|
Bryniarski KL, den Dekker W, Legutko J, Gasior P, Tahon J, Diletti R, Wilschut JM, Nuis RJ, Daemen J, Kleczynski P, Van Mieghem NM, Jang IK. Role of Lipid-Lowering and Anti-Inflammatory Therapies on Plaque Stabilization. J Clin Med 2024; 13:3096. [PMID: 38892807 PMCID: PMC11172633 DOI: 10.3390/jcm13113096] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Atherosclerosis is the predominant underlying etiopathology of coronary artery disease. Changes in plaque phenotype from stable to high risk may spur future major adverse cardiac events (MACE). Different pharmacological therapies have been implemented to mitigate this risk. Over the last two decades, intravascular imaging modalities have emerged in clinical studies to clarify how these therapies may affect the composition and burden of coronary plaques. Lipid-lowering agents, such as statins, ezetimibe, and proprotein convertase subtilisin/kexin type 9 inhibitors, were shown not only to reduce low-density lipoprotein levels and MACE but also to directly affect features of coronary plaque vulnerability. Studies have demonstrated that lipid-lowering therapy reduces the percentage of atheroma volume and number of macrophages and increases fibrous cap thickness. Future studies should answer the question of whether pharmacological plaque stabilization may be sufficient to mitigate the risk of MACE for selected groups of patients with atherosclerotic coronary disease.
Collapse
Affiliation(s)
- Krzysztof L. Bryniarski
- Department of interventional Cardiology, Thoraxcenter, Cardiovascular Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Interventional Cardiology, Jagiellonian University Medical College, Institute of Cardiology, St. John Paul II Hospital, 31-202 Krakow, Poland
| | - Wijnand den Dekker
- Department of interventional Cardiology, Thoraxcenter, Cardiovascular Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Jacek Legutko
- Department of Interventional Cardiology, Jagiellonian University Medical College, Institute of Cardiology, St. John Paul II Hospital, 31-202 Krakow, Poland
| | - Pawel Gasior
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Jeroen Tahon
- Department of interventional Cardiology, Thoraxcenter, Cardiovascular Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Interventional Cardiology, Imelda Hospital, 2820 Bonheiden, Belgium
| | - Roberto Diletti
- Department of interventional Cardiology, Thoraxcenter, Cardiovascular Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Jeroen M. Wilschut
- Department of interventional Cardiology, Thoraxcenter, Cardiovascular Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Rutger-Jan Nuis
- Department of interventional Cardiology, Thoraxcenter, Cardiovascular Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Joost Daemen
- Department of interventional Cardiology, Thoraxcenter, Cardiovascular Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Pawel Kleczynski
- Department of Interventional Cardiology, Jagiellonian University Medical College, Institute of Cardiology, St. John Paul II Hospital, 31-202 Krakow, Poland
| | - Nicolas M. Van Mieghem
- Department of interventional Cardiology, Thoraxcenter, Cardiovascular Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
11
|
Nicholls SJ, Nelson AJ. New targets and mechanisms of action for lipid-lowering and anti-inflammatory therapies in atherosclerosis: where does the field stand? Expert Opin Ther Targets 2024; 28:375-384. [PMID: 38815057 DOI: 10.1080/14728222.2024.2362644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/29/2024] [Indexed: 06/01/2024]
Abstract
INTRODUCTION Atherosclerotic cardiovascular disease remains a leading cause of morbidity and mortality worldwide, despite widespread use of statins. There is a need to develop additional therapeutic strategies that will complement statins to achieve more effective reductions in cardiovascular risk. AREAS COVERED This review provides a comprehensive summary of current areas of therapeutic development targeting both lipid and inflammatory factors implicated in the pathogenesis of atherosclerosis. In addition to develop of novel approaches that will produce more effective lowering of low-density lipoprotein cholesterol, clinical trials are currently evaluating the potential to target other atherogenic lipid parameters such as triglyceride-rich lipoproteins and Lp(a), in addition to promoting the biological properties of high-density lipoproteins. Targeting inflammation within the vascular wall has emerged as a new frontier in cardiovascular prevention, with early evidence that use of anti-inflammatory agents have the potential to reduce cardiovascular risk. EXPERT OPINION Clinical practice has an increasing array of therapeutic tools to achieve more effective lowering of low-density lipoprotein cholesterol for high-risk patients. In addition, clinical trials have the potential to deliver a range of additional agents to the clinic, that target alternative lipid and inflammatory mediators. This will permit the potential to personalize cardiovascular prevention.
Collapse
Affiliation(s)
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, Melbourne, Australia
| |
Collapse
|
12
|
Sabbour H, Bhatt DL, Elhenawi Y, Aljaberi A, Bennani L, Fiad T, Hasan K, Hashmani S, Hijazi RA, Khan Z, Shantouf R. A Practical Approach to the Management of Residual Cardiovascular Risk: United Arab Emirates Expert Consensus Panel on the Evidence for Icosapent Ethyl and Omega-3 Fatty Acids. Cardiovasc Drugs Ther 2024:10.1007/s10557-023-07519-z. [PMID: 38363478 DOI: 10.1007/s10557-023-07519-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/22/2023] [Indexed: 02/17/2024]
Abstract
PURPOSE Patients with hyperlipidemia treated with statins remain at a residual cardiovascular (CV) risk. Omega-3 polyunsaturated fatty acids hold the potential to mitigate the residual CV risk in statin-treated patients, with persistently elevated triglyceride (TG) levels. METHOD We reviewed the current evidence on the use of icosapent ethyl (IPE), an omega-3 fatty acid yielding a pure form of eicosapentaenoic acid. RESULTS REDUCE-IT reported a significant 25% reduction in CV events, including the need for coronary revascularization, the risk of fatal/nonfatal myocardial infarction, stroke, hospitalization for unstable angina, and CV death in patients on IPE, unseen with other omega-3 fatty acids treatments. IPE was effective in all patients regardless of baseline CV risk enhancers (TG levels, type-2 diabetes status, weight status, prior revascularization, or renal function). Adverse events (atrial fibrillation/flutter) related to IPE have occurred mostly in patients with prior atrial fibrillation. Yet, the net clinical benefit largely exceeded potential risks. The combination with other omega-3 polyunsaturated fatty acids, in particular DHA, eliminated the effect of EPA alone, as reported in the STRENGTH and OMEMI trials. Adding IPE to statin treatment seems to be cost-effective, especially in the context of secondary prevention of CVD, decreasing CV event frequency and subsequently the use of healthcare resources. CONCLUSION Importantly, IPE has been endorsed by 20 international medical societies as a statin add-on treatment in patients with dyslipidemia and high CV risk. Robust medical evidence supports IPE as a pillar in the management of dyslipidemia.
Collapse
Affiliation(s)
- Hani Sabbour
- Warren Alpert School of Medicine, Brown University, RI USA, Mediclinic Hospital, Abu Dhabi, United Arab Emirates.
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Yaser Elhenawi
- Heart And Vascular Institute, Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | - Asma Aljaberi
- Endocrine Division, Department of Medicine, Tawam Hospital, Abu Dhabi, United Arab Emirates
| | - Layal Bennani
- Medical Affairs, Biologix, Dubai, United Arab Emirates
| | - Tarek Fiad
- Centre Abu Dhabi, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Khwaja Hasan
- Packer Hospital Guthrie, Sayre, Pennsylvania, USA
| | - Shahrukh Hashmani
- Heart And Vascular Institute, Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | - Rabih A Hijazi
- Department of Endocrinology, Diabetes and Metabolism, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Zafar Khan
- Department of Cardiology, Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| | - Ronney Shantouf
- Heart And Vascular Institute, Cleveland Clinic, Abu Dhabi, United Arab Emirates
| |
Collapse
|
13
|
Pérez de Isla L, Díaz-Díaz JL, Romero MJ, Muñiz-Grijalvo O, Mediavilla JD, Argüeso R, de Andrés R, Fuentes F, Sánchez Muñoz-Torrero JF, Rubio P, Álvarez-Baños P, Mañas D, Suárez Gutierrez L, Saltijeral Cerezo A, Mata P. Characteristics of Coronary Atherosclerosis Related to Plaque Burden Regression During Treatment With Alirocumab: The ARCHITECT Study. Circ Cardiovasc Imaging 2024; 17:e016206. [PMID: 38205656 DOI: 10.1161/circimaging.123.016206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Intensive lipid-lowering therapy may induce coronary atherosclerosis regression. Nevertheless, the factors underlying the effect of lipid-lowering therapy on disease regression remain poorly characterized. Our aim was to determine which characteristics of atherosclerotic plaque are associated with a greater reduction in coronary plaque burden (PB) after treatment with alirocumab in patients with familial hypercholesterolemia. METHODS The ARCHITECT study (Effect of Alirocumab on Atherosclerotic Plaque Volume, Architecture and Composition) is a phase IV, open-label, multicenter, single-arm clinical trial to assess the effect of the treatment with alirocumab for 78 weeks on the coronary atherosclerotic PB and its characteristics in subjects with familial hypercholesterolemia without clinical atherosclerotic cardiovascular disease. Participants underwent a coronary computed tomographic angiography at baseline and a final one at 78 weeks. Every patient received alirocumab 150 mg subcutaneously every 14 days in addition to high-intensity statin therapy. RESULTS One hundred and four patients were enrolled. Median age was 53.3 (46.2-59.4) years and 54 were women (51.9%). The global coronary PB changed from 34.6% (32.5%-36.8%) at entry to 30.4% (27.4%-33.4%) at follow-up, which is -4.6% (-7.7% to -1.9%; P<0.001) reduction. A decrease in the percentage of unstable core (fibro-fatty+necrotic plaque; from 14.1 [7.9-22.3] to 8.0 [6.4-10.6]; -6.6%; P<0.001) was found. A greater PB (β, 0.36 [0.13-0.59]; P=0.002) and a higher proportion of unstable core (β, 0.15 [0.08-0.22]; P<0.001) were significantly related to PB regression. CONCLUSIONS Treatment with alirocumab in addition to high-intensity statin therapy might produce a greater PB regression in patients with familial hypercholesterolemia with higher baseline PB and in those with larger unstable core. Further studies are needed to corroborate the hypothesis raised by these results. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT05465278.
Collapse
Affiliation(s)
| | - Jose L Díaz-Díaz
- Internal Medicine Department, Hospital Abente y Lago, A Coruña, Spain (J.L.D.-D.)
| | - Manuel J Romero
- Internal Medicine Department, Hospital Infanta Elena, Huelva, Spain (M.J.R.)
| | | | - Juan D Mediavilla
- Internal Medicine Department, Hospital Universitario Virgen de las Nieves, Granada, Spain (J.D.M.)
| | - Rosa Argüeso
- Endocrinology Department, Hospital Universitario Lucus Augusti, Lugo, Spain (R.A.)
| | - Raimundo de Andrés
- Internal Medicine Department, Fundación Jiménez Díaz, Madrid, Spain (R.d.A.)
| | - Francisco Fuentes
- Lipid and Atherosclerosis Unit, CIBERObn, IMBIC, Hospital Universitario Reina Sofia, Córdoba, Spain (F.F.)
| | | | - Patricia Rubio
- Internal Medicine Department, Hospital Universitario Jerez de la Frontera, Spain (P.R.)
| | | | - Dolores Mañas
- Internal Medicine Department, Hospital General Universitario de Ciudad Real, Spain (D.M.)
| | | | | | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain (P.M.)
| |
Collapse
|
14
|
Abrahams T, Fujino M, Nelson AJ, Nicholls SJ. Impact of PCSK9 inhibitors on coronary atheroma phenotype following myocardial infarction: insights from intravascular imaging. Curr Opin Cardiol 2023; 38:504-508. [PMID: 37751373 DOI: 10.1097/hco.0000000000001080] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
PURPOSE OF REVIEW The aim of this study was to review the impact of combination lipid lowering with statins and proprotein convertase subtilisin kexin type 9 (PCSK9) inhibitors on coronary atherosclerosis using serial intravascular imaging. RECENT FINDINGS Early studies using intravascular ultrasound established the ability of increasingly intensive lipid lowering to both slow progression and ultimately promote regression of coronary disease. More recent clinical trials that have employed serial imaging with optical coherence tomography have permitted the ability to evaluate the impact of intensive lipid lowering on compositional features associated with plaque vulnerability. In particular, the combination of intensive statin and PCSK9 inhibitor therapy promotes plaque stability in patients following an acute coronary syndrome. SUMMARY More intensive lipid lowering using the combination of statins and PCSK9 inhibitors promote plaque regression in addition to promoting calcification, fibrous cap thickening and reductions in plaque lipid. These plaque-stabilizing effects underscore the benefits of combination therapy on cardiovascular events and highlight the importance of combination lipid-lowering therapy.
Collapse
Affiliation(s)
- Timothy Abrahams
- Victorian Heart Institute, Monash University, Clayton, Victoria, Australia
| | | | | | | |
Collapse
|
15
|
Iatan I, Guan M, Humphries KH, Yeoh E, Mancini GBJ. Atherosclerotic Coronary Plaque Regression and Risk of Adverse Cardiovascular Events: A Systematic Review and Updated Meta-Regression Analysis. JAMA Cardiol 2023; 8:937-945. [PMID: 37647074 PMCID: PMC10469293 DOI: 10.1001/jamacardio.2023.2731] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 07/08/2023] [Indexed: 09/01/2023]
Abstract
Importance The association between changes in atherosclerotic plaque induced by lipid-lowering therapies (LLTs) and reduction in major adverse cardiovascular events (MACEs) remains controversial. Objective To evaluate the association between coronary plaque regression assessed by intravascular ultrasound (IVUS) and MACEs. Data Sources A comprehensive, systematic search of publications in PubMed, Embase, Cochrane Central Register of Controlled Trials, and Web of Science was performed. Study Selection Clinical prospective studies of LLTs reporting change in percent atheroma volume (PAV) assessed by IVUS and describing MACE components were selected. Data Extraction and Synthesis Reporting was performed in compliance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. The association between mean change in PAV and MACEs was analyzed by meta-regression using mixed-effects, 2-level binomial logistic regression models, unadjusted and adjusted for clinical covariates, including mean age, baseline PAV, baseline low-density lipoprotein cholesterol level, and study duration. Main Outcome and Measures Mean PAV change and MACE in intervention and comparator arms were assessed in an updated systematic review and meta-regression analysis of IVUS trials of LLTs that also reported MACEs. Results This meta-analysis included 23 studies published between July 2001 and July 2022, including 7407 patients and trial durations ranging from 11 to 104 weeks. Mean (SD) patient age ranged from 55.8 (9.8) to 70.2 (7.6) years, and the number of male patients from 245 of 507 (48.3%) to 24 of 26 (92.3%). Change in PAV across 46 study arms ranged from -5.6% to 3.1%. The number of MACEs ranged from 0 to 72 per study arm (17 groups [37%] reported no events, 9 [20%] reported 1-2 events, and 20 [43%] reported ≥3 events). In unadjusted analysis, a 1% decrease in mean PAV was associated with 17% reduced odds of MACEs (unadjusted OR, 0.83; 95% CI, 0.71-0.98; P = .03), and with a 14% reduction in MACEs in adjusted analysis (adjusted OR, 0.86; 95% CI, 0.75-1.00; P = .050). Further adjustment for cardiovascular risk factors showed a 19% reduced risk (adjusted OR, 0.81; 95% CI, 0.68-0.96; P = .01) per 1% decrease in PAV. A 1% reduction of PAV change between intervention and comparator arms within studies was also associated with a significant 25% reduction in MACEs (OR, 0.75; 95% CI, 0.56-1.00; P = .046). Conclusions and Relevance In this meta-analysis, regression of atherosclerotic plaque by 1% was associated with a 25% reduction in the odds of MACEs. These findings suggest that change in PAV could be a surrogate marker for MACEs, but given the heterogeneity in the outcomes, additional data are needed.
Collapse
Affiliation(s)
- Iulia Iatan
- Centre for Heart Lung Innovation, Department of Medicine, St Paul’s Hospital, Providence Health Care, University of British Columbia, Vancouver, British Columbia, Canada
| | - Meijiao Guan
- British Columbia Centre for Improved Cardiovascular Health, Vancouver, British Columbia, Canada
| | - Karin H. Humphries
- British Columbia Centre for Improved Cardiovascular Health, Vancouver, British Columbia, Canada
| | - Eunice Yeoh
- Division of Cardiology, Centre for Cardiovascular Innovation, Cardiovascular Imaging Research Core Laboratory, University of British Columbia, Vancouver, British Columbia, Canada
| | - G. B. John Mancini
- Division of Cardiology, Centre for Cardiovascular Innovation, Cardiovascular Imaging Research Core Laboratory, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
16
|
Kononov S, Azarova I, Klyosova E, Bykanova M, Churnosov M, Solodilova M, Polonikov A. Lipid-Associated GWAS Loci Predict Antiatherogenic Effects of Rosuvastatin in Patients with Coronary Artery Disease. Genes (Basel) 2023; 14:1259. [PMID: 37372439 PMCID: PMC10298211 DOI: 10.3390/genes14061259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
We have shown that lipid-associated loci discovered by genome-wide association studies (GWAS) have pleiotropic effects on lipid metabolism, carotid intima-media thickness (CIMT), and CAD risk. Here, we investigated the impact of lipid-associated GWAS loci on the efficacy of rosuvastatin therapy in terms of changes in plasma lipid levels and CIMT. The study comprised 116 CAD patients with hypercholesterolemia. CIMT, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and triglycerides (TG) were measured at baseline and after 6 and 12 months of follow-up, respectively. Genotyping of fifteen lipid-associated GWAS loci was performed by the MassArray-4 System. Linear regression analysis adjusted for sex, age, body mass index, and rosuvastatin dose was used to estimate the phenotypic effects of polymorphisms, and p-values were calculated through adaptive permutation tests by the PLINK software, v1.9. Over one-year rosuvastatin therapy, a decrease in CIMT was linked to rs1689800, rs4846914, rs12328675, rs55730499, rs9987289, rs11220463, rs16942887, and rs881844 polymorphisms (Pperm < 0.05). TC change was associated with rs55730499, rs11220463, and rs6065906; LDL-C change was linked to the rs55730499, rs1689800, and rs16942887 polymorphisms; and TG change was linked to polymorphisms rs838880 and rs1883025 (Pperm < 0.05). In conclusion, polymorphisms rs1689800, rs55730499, rs11220463, and rs16942887 were found to be predictive markers for multiple antiatherogenic effects of rosuvastatin in CAD patients.
Collapse
Affiliation(s)
- Stanislav Kononov
- Department of Internal Medicine No. 2, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
| | - Iuliia Azarova
- Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| | - Elena Klyosova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
| | - Marina Bykanova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State University, 85 Pobedy Street, 308015 Belgorod, Russia
| | - Maria Solodilova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
| | - Alexey Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| |
Collapse
|
17
|
Yao Y, Zhang P. Novel ultrasound techniques in the identification of vulnerable plaques-an updated review of the literature. Front Cardiovasc Med 2023; 10:1069745. [PMID: 37293284 PMCID: PMC10244552 DOI: 10.3389/fcvm.2023.1069745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
Atherosclerosis is an inflammatory disease partly mediated by lipoproteins. The rupture of vulnerable atherosclerotic plaques and thrombosis are major contributors to the development of acute cardiovascular events. Despite various advances in the treatment of atherosclerosis, there has been no satisfaction in the prevention and assessment of atherosclerotic vascular disease. The identification and classification of vulnerable plaques at an early stage as well as research of new treatments remain a challenge and the ultimate goal in the management of atherosclerosis and cardiovascular disease. The specific morphological features of vulnerable plaques, including intraplaque hemorrhage, large lipid necrotic cores, thin fibrous caps, inflammation, and neovascularisation, make it possible to identify and characterize plaques with a variety of invasive and non-invasive imaging techniques. Notably, the development of novel ultrasound techniques has introduced the traditional assessment of plaque echogenicity and luminal stenosis to a deeper assessment of plaque composition and the molecular field. This review will discuss the advantages and limitations of five currently available ultrasound imaging modalities for assessing plaque vulnerability, based on the biological characteristics of the vulnerable plaque, and their value in terms of clinical diagnosis, prognosis, and treatment efficacy assessment.
Collapse
|
18
|
Pérez de Isla L, Díaz-Díaz JL, Romero MJ, Muñiz-Grijalvo O, Mediavilla JD, Argüeso R, Sánchez Muñoz-Torrero JF, Rubio P, Álvarez-Baños P, Ponte P, Mañas D, Suárez Gutierrez L, Cepeda JM, Casañas M, Fuentes F, Guijarro C, Ángel Barba M, Saltijeral Cerezo A, Padró T, Mata P. Alirocumab and Coronary Atherosclerosis in Asymptomatic Patients with Familial Hypercholesterolemia: The ARCHITECT Study. Circulation 2023; 147:1436-1443. [PMID: 37009731 PMCID: PMC10158600 DOI: 10.1161/circulationaha.122.062557] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 03/01/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND The effect of alirocumab, a PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitor, on coronary plaque burden in patients with familial hypercholesterolemia has not been addressed. Our aim was to assess changes in coronary plaque burden and its characteristics after treatment with alirocumab by quantification and characterization of atherosclerotic plaque throughout the coronary tree on the basis of a noninvasive analysis of coronary computed tomographic angiography in asymptomatic subjects with familial hypercholesterolemia receiving optimized and stable treatment with maximum tolerated statin dose with or without ezetimibe. METHODS This study is a phase IV, open-label, multicenter, single-arm clinical trial to assess changes in coronary plaque burden and its characteristics after 78 weeks of treatment with alirocumab in patients with familial hypercholesterolemia without clinical atherosclerotic cardiovascular disease. Participants underwent an initial coronary computed tomographic angiography at baseline and another at 78 weeks. Every patient received 150 mg of alirocumab subcutaneiously every 14 days in addition to high-intensity statin therapy. The main outcome was the change on coronary plaque burden and its characteristics by quantification and characterization of atherosclerotic plaque throughout the coronary tree on the basis of analysis of coronary computed tomographic angiography. RESULTS The study was completed by 104 patients. The median age was 53.3 (46.2-59.4) years. Of these patients, 54 were women (51.9%). Median low-density lipoprotein cholesterol was 138.9 (117.5-175.3) mg/dL at entry and 45.0 (36.0-65.0) mg/dL at follow-up (P<0.001). Coronary plaque burden changed from 34.6% (32.5%-36.8%) at entry to 30.4% (27.4%-33.4%) at follow-up (P<0.001). A significant change in the characteristics of the coronary atherosclerosis was also found: an increase in the proportion of calcified (+0.3%; P<0.001) and mainly fibrous (+6.2%; P<0.001) plaque, accompanied by a decrease in the percentage of fibro-fatty (-3.9%; P<0.001) and necrotic plaque (-0.6%; P<0.001). CONCLUSIONS Treatment with alirocumab in addition to high-intensity statin therapy resulted in significant regression of coronary plaque burden and plaque stabilization on coronary computed tomographic angiography over 78 weeks in these groups of patients with familial hypercholesterolemia without clinical atherosclerotic cardiovascular disease. ARCHITECT (Effect of Alirocumab on Atherosclerotic Plaque Volume, Architecture and Composition) could link and explain ODYSSEY OUTCOMES (Evaluation of Cardiovascular Outcomes After an Acute Coronary Syndrome During Treatment With Alirocumab) results. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT05465278.
Collapse
Affiliation(s)
| | - Jose L. Díaz-Díaz
- Internal Medicine Department, Hospital Abente y Lago, A Coruña, Spain (J.L.E.-D.)
| | - Manuel J. Romero
- Internal Medicine Department, Hospital Infanta Elena, Huelva, Spain M.J.R.)
| | | | - Juan D. Mediavilla
- Internal Medicine Department, Hospital Universitario Virgen de las Nieves, Granada, Spain (J.D.M.)
| | - Rosa Argüeso
- Endocrinology Department, Hospital Universitario Lucus Augusti, Lugo, Spain (R.A.)
| | | | - Patricia Rubio
- Internal Medicine Department, Hospital Universitario Jerez de la Frontera, Spain (P.R.)
| | | | - Paola Ponte
- Internal Medicine Department, Hospital Santa Creu i Sant Pau, Barcelona, Spain (P.P.)
| | - Dolores Mañas
- Internal Medicine Department, Hospital General Universitario de Ciudad Real, Spain (D.M.)
| | | | - José María Cepeda
- Internal Medicine Department, Hospital Comarcal Vega Baja, Orihuela, Alicante, Spain (J.M.C.)
| | - Marta Casañas
- Internal Medicine Department, Hospital San Pedro, Logroño, Spain (M.C.)
| | - Francisco Fuentes
- Lipid and Atherosclerosis Unit, CIBERObn, IMBIC. Hospital Universitario Reina Sofia, Córdoba, Spain (F.F.)
| | - Carlos Guijarro
- Internal Medicine Department, Hospital Universitario Fundación Alcorcón-Universidad Rey Juan Carlos, Madrid, Spain (C.G.)
| | - Miguel Ángel Barba
- Internal Medicine Department, Complejo Hospitalario Universitario, Albacete, Spain (M.A.B.)
| | | | - Teresa Padró
- Programa-ICCC Cardiovascular, Institut de Recerca Hospital Santa Creu i Sant Pau, IIB-Sant Pau, CIBERCV, Barcelona, Spain (T.P.)
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain (P.M.)
| |
Collapse
|
19
|
Tufaro V, Serruys PW, Räber L, Bennett MR, Torii R, Gu SZ, Onuma Y, Mathur A, Baumbach A, Bourantas CV. Intravascular imaging assessment of pharmacotherapies targeting atherosclerosis: advantages and limitations in predicting their prognostic implications. Cardiovasc Res 2023; 119:121-135. [PMID: 35394014 DOI: 10.1093/cvr/cvac051] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 02/17/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Intravascular imaging has been often used over the recent years to examine the efficacy of emerging therapies targeting plaque evolution. Serial intravascular ultrasound, optical coherence tomography, or near-infrared spectroscopy-intravascular ultrasound studies have allowed us to evaluate the effects of different therapies on plaque burden and morphology, providing unique mechanistic insights about the mode of action of these treatments. Plaque burden reduction, a decrease in necrotic core component or macrophage accumulation-which has been associated with inflammation-and an increase in fibrous cap thickness over fibroatheromas have been used as surrogate endpoints to assess the value of several drugs in inhibiting plaque evolution and improving clinical outcomes. However, some reports have demonstrated weak associations between the effects of novel treatments on coronary atheroma and composition and their prognostic implications. This review examines the value of invasive imaging in assessing pharmacotherapies targeting atherosclerosis. It summarizes the findings of serial intravascular imaging studies assessing the effects of different drugs on atheroma burden and morphology and compares them with the results of large-scale trials evaluating their impact on clinical outcome. Furthermore, it highlights the limited efficacy of established intravascular imaging surrogate endpoints in predicting the prognostic value of these pharmacotherapies and introduces alternative imaging endpoints based on multimodality/hybrid intravascular imaging that may enable more accurate assessment of the athero-protective and prognostic effects of emerging therapies.
Collapse
Affiliation(s)
- Vincenzo Tufaro
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | | - Lorenz Räber
- Department of Cardiology, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Ryo Torii
- Department of Mechanical Engineering, University College London, London, UK
| | - Sophie Zhaotao Gu
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Yoshinobu Onuma
- Department of Cardiology, National University of Ireland, Galway, Ireland
| | - Anthony Mathur
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Andreas Baumbach
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Yale University School of Medicine, New Haven, CT, USA
| | - Christos Vasileios Bourantas
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Institute of Cardiovascular Sciences, University College London, London, UK
| |
Collapse
|
20
|
Gallo G, Sarzani R, Cicero AFG, Genovese S, Pirro M, Gallelli L, Faggiano A, Volpe M. An Expert Opinion on the Role of the Rosuvastatin/Amlodipine Single Pill Fixed Dose Combination in Cardiovascular Prevention. High Blood Press Cardiovasc Prev 2023; 30:83-91. [PMID: 37020154 PMCID: PMC10089988 DOI: 10.1007/s40292-023-00570-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/13/2023] [Indexed: 04/07/2023] Open
Abstract
Current cardiovascular disease prevention strategies are based on the management of cardiovascular risk as a continuum, redefining the therapeutic goals for each individual based on the estimated global risk profile. Given the frequent clustering of the principal cardiovascular risk factors, such as hypertension, diabetes and dyslipidaemia, in the same individual, patients are required to take multiple drugs to achieve therapeutic targets. The adoption of single pill fixed dose combinations may contribute to achieve better control of blood pressure and cholesterol compared to the separate administration of the individual drugs, mostly due to better adherence related to therapeutic simplicities. This paper reports the outcomes of an Expert multidisciplinary Roundtable. In particular, the rational and potential clinical use of the single pill fixed dose combination "Rosuvastatin-Amlodipine" for the management of concomitant hypertension/hypercholesterolemia in different clinical fields are discussed. This Expert Opinion also illustrates the importance of an early and effective management of total cardiovascular risk, highlights the substantial benefits of combining blood pressure and lipid-lowering treatments in a single-pill fixed dose combination and attempts to identify and overcome the barriers to the implementation in clinical practice of the fixed dose combinations with dual targets. This Expert Panel identifies and proposes the categories of patients who may benefit the most from this fixed dose combination.
Collapse
Affiliation(s)
- Giovanna Gallo
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Riccardo Sarzani
- Internal Medicine and Geriatrics, 'Hypertension Excellence Centre' of the European Society of Hypertension, IRCCS INRCA, Ancona, Italy
- Department of Clinical and Molecular Sciences, University 'Politecnica delle Marche', Ancona, Italy
| | - Arrigo Francesco Giuseppe Cicero
- Hypertension and Cardiovascular Risk Research Unit, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, Bologna, Italy
- IRCCS AOU Policlinico di S. Orsola, Bologna, Italy
| | - Stefano Genovese
- Endocrine and Metabolic Diseases Unit IRCCS Centro Cardiologico Monzino, 20138, Milan, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luca Gallelli
- Department of Health Sciences, Campus "Salvatore Venuta", University of Catanzaro "Magna Græcia", 88100, Catanzaro, Italy
| | - Andrea Faggiano
- Cardiovascular Unit, Internal Medicine Department, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico di Milano, University of Milan, Milan, Italy
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.
- IRCCS San Raffaele Roma, Rome, Italy.
| |
Collapse
|
21
|
Di Giovanni G, Kataoka Y, Bubb K, Nelson AJ, Nicholls SJ. Impact of lipid lowering on coronary atherosclerosis moving from the lumen to the artery wall. Atherosclerosis 2023; 367:8-14. [PMID: 36716526 DOI: 10.1016/j.atherosclerosis.2023.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Randomized clinical trials have demonstrated that increasingly intensive lowering of low-density lipoprotein cholesterol (LDL-C) reduces the rate of cardiovascular events in the primary and secondary prevention setting. Integration of serial coronary imaging within clinical trials has enabled evaluation of medical therapies on the natural history of coronary disease. These studies have extended from early investigation of coronary obstruction with angiography to more contemporary evaluation of plaque burden and composition with imaging modalities that directly visualize the artery wall. The findings of these trials have demonstrated that intensive lipid lowering promotes plaque regression and stabilization. The lessons of this body of research provide a biological rationale underscoring the ability of intensive lipid lowering to reduce cardiovascular risk and have the potential to promote greater uptake in clinical practice.
Collapse
Affiliation(s)
- Giuseppe Di Giovanni
- South Australian Health and Medical Research Institute, Adelaide, Australia; Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Yu Kataoka
- National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Kristen Bubb
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | | |
Collapse
|
22
|
ANGPTL4 stabilizes atherosclerotic plaques and modulates the phenotypic transition of vascular smooth muscle cells through KLF4 downregulation. Exp Mol Med 2023; 55:426-442. [PMID: 36782020 PMCID: PMC9981608 DOI: 10.1038/s12276-023-00937-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/29/2022] [Accepted: 12/08/2022] [Indexed: 02/15/2023] Open
Abstract
Atherosclerosis, the leading cause of death, is a vascular disease of chronic inflammation. We recently showed that angiopoietin-like 4 (ANGPTL4) promotes cardiac repair by suppressing pathological inflammation. Given the fundamental contribution of inflammation to atherosclerosis, we assessed the role of ANGPTL4 in the development of atherosclerosis and determined whether ANGPTL4 regulates atherosclerotic plaque stability. We injected ANGPTL4 protein twice a week into atherosclerotic Apoe-/- mice and analyzed the atherosclerotic lesion size, inflammation, and plaque stability. In atherosclerotic mice, ANGPTL4 reduced atherosclerotic plaque size and vascular inflammation. In the atherosclerotic lesions and fibrous caps, the number of α-SMA(+), SM22α(+), and SM-MHC(+) cells was higher, while the number of CD68(+) and Mac2(+) cells was lower in the ANGPTL4 group. Most importantly, the fibrous cap was significantly thicker in the ANGPTL4 group than in the control group. Smooth muscle cells (SMCs) isolated from atherosclerotic aortas showed significantly increased expression of CD68 and Krüppel-like factor 4 (KLF4), a modulator of the vascular SMC phenotype, along with downregulation of α-SMA, and these changes were attenuated by ANGPTL4 treatment. Furthermore, ANGPTL4 reduced TNFα-induced NADPH oxidase 1 (NOX1), a major source of reactive oxygen species, resulting in the attenuation of KLF4-mediated SMC phenotypic changes. We showed that acute myocardial infarction (AMI) patients with higher levels of ANGPTL4 had fewer vascular events than AMI patients with lower levels of ANGPTL4 (p < 0.05). Our results reveal that ANGPTL4 treatment inhibits atherogenesis and suggest that targeting vascular stability and inflammation may serve as a novel therapeutic strategy to prevent and treat atherosclerosis. Even more importantly, ANGPTL4 treatment inhibited the phenotypic changes of SMCs into macrophage-like cells by downregulating NOX1 activation of KLF4, leading to the formation of more stable plaques.
Collapse
|
23
|
Legutko J, Bryniarski KL, Kaluza GL, Roleder T, Pociask E, Kedhi E, Wojakowski W, Jang IK, Kleczynski P. Intracoronary Imaging of Vulnerable Plaque-From Clinical Research to Everyday Practice. J Clin Med 2022; 11:jcm11226639. [PMID: 36431116 PMCID: PMC9699515 DOI: 10.3390/jcm11226639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 10/30/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
The introduction into clinical practice of intravascular imaging, including intravascular ultrasound (IVUS), optical coherence tomography (OCT) and their derivatives, allowed for the in vivo assessment of coronary atherosclerosis in humans, including insights into plaque evolution and progression process. Intravascular ultrasound, the most commonly used intravascular modality in many countries, due to its low resolution cannot assess many features of vulnerable plaque such as lipid plaque or thin-cap fibroatheroma. Thus, novel methods were introduced to facilitate this problem including virtual histology intravascular ultrasound and later on near-infrared spectroscopy and OCT. Howbeit, none of the currently used modalities can assess all known characteristics of plaque vulnerability; hence, the idea of combining different intravascular imaging methods has emerged including NIRS-IVUS or OCT-IVUS imaging. All of those described methods may allow us to identify the most vulnerable plaques, which are prone to cause acute coronary syndrome, and thus they may allow us to introduce proper treatment before plaque destabilization.
Collapse
Affiliation(s)
- Jacek Legutko
- Department of Interventional Cardiology, Faculty of Medicine, Institute of Cardiology, Jagiellonian University Medical College, 31-202 Kraków, Poland
- Clinical Department of Interventional Cardiology, John Paul II Hospital, 31-202 Kraków, Poland
| | - Krzysztof L. Bryniarski
- Department of Interventional Cardiology, Faculty of Medicine, Institute of Cardiology, Jagiellonian University Medical College, 31-202 Kraków, Poland
- Clinical Department of Interventional Cardiology, John Paul II Hospital, 31-202 Kraków, Poland
| | - Grzegorz L. Kaluza
- Skirball Center for Innovation, Cardiovascular Research Foundation, Orangeburg, NY 10019, USA
| | - Tomasz Roleder
- Department of Cardiology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Elzbieta Pociask
- Department of Biocybernetics and Biomedical Engineering, AGH University of Science and Technology, 30-059 Kraków, Poland
| | - Elvin Kedhi
- Clinique Hopitaliere Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Wojciech Wojakowski
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, 40-635 Katowice, Poland
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, GRB 800, Boston, MA 02115, USA
- Division of Cardiology, Kyung Hee University Hospital, Seoul 02447, Korea
| | - Pawel Kleczynski
- Department of Interventional Cardiology, Faculty of Medicine, Institute of Cardiology, Jagiellonian University Medical College, 31-202 Kraków, Poland
- Clinical Department of Interventional Cardiology, John Paul II Hospital, 31-202 Kraków, Poland
- Correspondence: ; Tel.: +48-12-614-35-01
| |
Collapse
|
24
|
Intensive lipid lowering agents and coronary atherosclerosis: Insights from intravascular imaging. Am J Prev Cardiol 2022; 11:100366. [PMID: 35856069 PMCID: PMC9287145 DOI: 10.1016/j.ajpc.2022.100366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 01/23/2023] Open
Abstract
Advances in intravascular imaging have permitted comprehensive evaluation of coronary atherosclerotic plaque from the perspective of its burden and individual components. These advances have been integrated in clinical trials that have evaluated the impact of intensive lipid lowering regimens. These trials have demonstrated that intensive lipid lowering, using high dose statins as monotherapy and in combination with new lipid lowering agents, produce favorable effects on coronary atheroma, resulting in regression and stabilization. These findings provide important biological insights to understand how intensive lipid lowering may reduce cardiovascular risk. This review aims to provide the reader with a contemporary overview of the findings of these studies and to propose the potential clinical implications for management of higher risk patients with atherosclerotic coronary artery disease.
Collapse
|
25
|
Bélanger AM, Akioyamen LE, Ruel I, Hales L, Genest J. Aortic stenosis in homozygous familial hypercholesterolaemia: a paradigm shift over a century. Eur Heart J 2022; 43:3227-3239. [PMID: 35776569 DOI: 10.1093/eurheartj/ehac339] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/13/2022] [Accepted: 06/09/2022] [Indexed: 01/31/2023] Open
Abstract
AIMS Homozygous familial hypercholesterolaemia (HoFH) is an orphan disease defined by extreme elevations in low-density lipoprotein cholesterol, cutaneous xanthomas, and pre-mature atherosclerotic cardiovascular disease. Survival has more than doubled over the past three decades. Aortic stenosis (AS) [supravalvular aortic stenosis (SVAS) or valvular aortic stenosis (VAS)] is commonly encountered. There are no medical treatments available and complex high-risk surgeries represent the only available option in severe cases. A systematic review was performed to summarize the current evidence on AS in HoFH and to determine whether pharmacological treatment (statins) have had an impact on clinical presentation, phenotype and clinical course over the past nine decades (PROSPERO CRD42021250565). METHODS AND RESULTS MEDLINE, Embase Classic + Embase, Cochrane Central Register of Controlled Trials, PubMed, AfricaWide, and Scopus were searched from inception to 10 November 2021. Searches identified 381 publications, of which 19 were retained; they were cross-sectional or retrospective studies. Separately, 108 individual case reports were described. Within the 424 HoFH cases, AS was identified in 57% of patients in the pre-statin era vs. 35% in patients reported more recently (>2000, long-term statin period). With an increase in longevity due to statins and lipoprotein apheresis, a change in the proportion of patients with SVAS and VAS with a SVAS:VAS ratio of 47:53 and 10:90 for HoFH patients not on statin and on long-term statin, respectively, was noted. CONCLUSION These data suggest that SVAS and VAS are frequent in HoFH and that the phenotype has shifted towards calcific VAS as statins and lipoprotein apheresis improve survival in these patients.
Collapse
Affiliation(s)
- Alexandre M Bélanger
- Research Institute of the McGill University Health Centre, 1001, Boul. Décarie, Office EM1.2212, Montréal, Québec, Canada
| | - Leo E Akioyamen
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Isabelle Ruel
- Research Institute of the McGill University Health Centre, 1001, Boul. Décarie, Office EM1.2212, Montréal, Québec, Canada
| | - Lindsay Hales
- McGill University Health Centre Medical Libraries, Montréal, Québec, Canada
| | - Jacques Genest
- Research Institute of the McGill University Health Centre, 1001, Boul. Décarie, Office EM1.2212, Montréal, Québec, Canada
| |
Collapse
|
26
|
Zhang W, Mintz GS, Cao Y, Matsumura M, Lee T, Hoshino M, Usui E, Kanaji Y, Murai T, Yonetsu T, Kakuta T, Maehara A. Clinical determinants of coronary artery disease burden and vulnerability using optical coherence tomography co-registered with intravascular ultrasound. Coron Artery Dis 2022; 33:114-124. [PMID: 34411011 DOI: 10.1097/mca.0000000000001088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES We investigated clinical determinants of disease burden and vulnerability using optical coherence tomography (OCT) co-registered with intravascular ultrasound (IVUS) in a large cohort of patients. METHODS A total of 704 patients [44.5% with acute coronary syndromes (ACS)] underwent coronary intervention. IVUS plaque burden and OCT lipid, macrophage and calcium indices and the presence of thrombus, plaque rupture and thin-cap fibroatheroma (TCFA) were analyzed. RESULTS Median patient age was 66 years with 81.8% men, 34.4% with diabetes mellitus and 15.5% with preadmission statins. Median lesion length was 25.7 mm, and 33.0% had a TCFA. Adjusted models indicated (1) older patient age was related to more calcium, but fewer macrophages; (2) men were related to more thrombus with plaque rupture while women had more thrombus without plaque rupture; (3) ACS presentation was related to morphological acute thrombotic events (more thrombus with/without rupture) and plaque vulnerability (more TCFA, more lipid and macrophages and larger plaque burden); (4) diabetes mellitus was related to a greater atherosclerotic disease burden (more lipid and calcium and larger plaque burden) and more thrombus without rupture; (5) hypertension was related to more macrophages; (6) current smoking was related to less calcium; and (7) renal insufficiency and preadmission statin therapy were not independently associated with IVUS or OCT plaque morphology. CONCLUSION Patient characteristics, especially diabetes mellitus and aging, affect underlying atherosclerotic burden, among which a greater lipidic burden along with sex differences influence local thrombotic morphology that affects clinical presentation.
Collapse
Affiliation(s)
- Wenbin Zhang
- Clinical Trials Center, Cardiovascular Research Foundation
- Division of Cardiology, NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, USA
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou
| | - Gary S Mintz
- Clinical Trials Center, Cardiovascular Research Foundation
| | - Yang Cao
- Clinical Trials Center, Cardiovascular Research Foundation
- Division of Cardiology, NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, USA
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | - Tetsumin Lee
- Clinical Trials Center, Cardiovascular Research Foundation
- Division of Cardiology, NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, USA
| | - Masahiro Hoshino
- Cardiovascular Medicine, Tsuchiura Kyodo Hospital, Ibaraki, Japan
| | - Eisuke Usui
- Clinical Trials Center, Cardiovascular Research Foundation
- Cardiovascular Medicine, Tsuchiura Kyodo Hospital, Ibaraki, Japan
| | - Yoshihisa Kanaji
- Cardiovascular Medicine, Tsuchiura Kyodo Hospital, Ibaraki, Japan
| | - Tadashi Murai
- Cardiovascular Medicine, Tsuchiura Kyodo Hospital, Ibaraki, Japan
| | - Taishi Yonetsu
- Cardiovascular Medicine, Tsuchiura Kyodo Hospital, Ibaraki, Japan
| | - Tsunekazu Kakuta
- Cardiovascular Medicine, Tsuchiura Kyodo Hospital, Ibaraki, Japan
| | - Akiko Maehara
- Clinical Trials Center, Cardiovascular Research Foundation
- Division of Cardiology, NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
27
|
What Is Hidden Behind Yellow Pixels: from Pathology to Intravascular Imaging of Atherosclerotic Plaque. Curr Atheroscler Rep 2022; 24:97-108. [PMID: 35107762 DOI: 10.1007/s11883-022-00990-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2021] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Intravascular imaging systems can identify lipid-rich and vulnerable plaques and help in treatment guidance. The comparability of different intracoronary imaging methods remains unclear. In this paper, we review atherosclerotic plaque pathology, plaque-stabilising effects of different lipid-lowering therapies and usage of intravascular imaging modalities. We present the results of our study in which we evaluated the correlation of the intravascular ultrasound iMAP system (iMAP-IVUS) and near-infrared spectroscopy (NIRS) in the diagnosis of vulnerable coronary plaques. RECENT FINDINGS Lipids have an essential contribution to plaque evolution and vulnerability. Increase in plaque vulnerability alone even without increase in plaque burden defines progression of atherosclerosis. Lipidic tissue has a significant diagnostic value in patient risk stratification and can serve as a treatment target. Different vulnerable plaque parameters can be visualised with iMAP-IVUS and NIRS. Intravascular imaging systems can differ with regard to their sensitivity, specificity and limitations. Lipid-lowering therapy is crucial in plaque stabilisation.
Collapse
|
28
|
Kononov S, Mal G, Azarova I, Klyosova E, Bykanova M, Churnosov M, Polonikov A. Pharmacogenetic loci for rosuvastatin are associated with intima-media thickness change and coronary artery disease risk. Pharmacogenomics 2022; 23:15-34. [PMID: 34905955 DOI: 10.2217/pgs-2021-0097] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 11/09/2021] [Indexed: 11/21/2022] Open
Abstract
Aim: Polymorphisms at LPA, LDLR, APOE, APOC1, MYLIP and ABCG2 are attractive targets for assessment of their impact on lipid-lowering therapy with rosuvastatin. The present study investigated whether polymorphisms at these genes are associated with the risk of coronary artery disease (CAD) development, and reduction of atherogenic lipids and carotid intima-media thickness (CIMT) in CAD patients, taking rosuvastatin. Materials & methods: 190 CAD patients and 1697 subjects were enrolled in pharmacogenetic and genetic association study, respectively. SNP genotyping was done using the MassARRAY-4 system. Results:MYLIP rs6924995, rs3757354, APOC1 rs445925, LDLR rs6511720, APOE rs7412, ABCG2 rs2199936, rs1481012 variants were significantly associated with CAD susceptibility (p = 0.016, 0.0003, <0.0001, <0.0001, 0.013, 0.016, 0.0035, respectively), as well as with CIMT regression (except ABCG2 variants; p = 0.05, 0.039, 0.039, 0.016, 0.0065), and changes in plasma lipids during rosuvastatin therapy. Conclusion: The studied polymorphisms possess pleiotropic effects on plasma lipids and CIMT, CAD susceptibility, and determine lipid-lowering response to rosuvastatin.
Collapse
Affiliation(s)
- Stanislav Kononov
- Department of Internal Medicine N 2, Kursk State Medical University, 14 Pirogova St., Kursk 305035, Russian Federation
| | - Galina Mal
- Department of Pharmacology, Kursk State Medical University, 3 Karl Marx St., Kursk 305041, Russian Federation
| | - Iuliia Azarova
- Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx St., Kursk 305041, Russian Federation
- Laboratory of Biochemical Genetics & Metabolomics, Research Institute for Genetic & Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation
| | - Elena Klyosova
- Laboratory of Biochemical Genetics & Metabolomics, Research Institute for Genetic & Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation
- Department of Biology, Medical Genetics & Ecology, Kursk State Medical University, 3 Karl Marx St., Kursk 305041, Russian Federation
| | - Marina Bykanova
- Department of Biology, Medical Genetics & Ecology, Kursk State Medical University, 3 Karl Marx St., Kursk 305041, Russian Federation
- Laboratory of Genomic Research, Research Institute for Genetic & Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State University, 85 Pobeda St., Belgorod 308015, Russian Federation
| | - Alexey Polonikov
- Department of Biology, Medical Genetics & Ecology, Kursk State Medical University, 3 Karl Marx St., Kursk 305041, Russian Federation
- Laboratory of Statistical Genetics & Bioinformatics, Research Institute for Genetic & Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation
| |
Collapse
|
29
|
Pulipati VP, Alenghat FJ. The impact of lipid-lowering medications on coronary artery plaque characteristics. Am J Prev Cardiol 2021; 8:100294. [PMID: 34877559 PMCID: PMC8627965 DOI: 10.1016/j.ajpc.2021.100294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/29/2021] [Accepted: 11/06/2021] [Indexed: 11/28/2022] Open
Abstract
Atherosclerosis is the predominant cause of coronary artery disease. The last several decades have witnessed significant advances in lipid-lowering therapies, which comprise a central component of atherosclerotic cardiovascular disease prevention. In addition to cardiovascular risk reduction with dyslipidemia management, some lipid-based therapies show promise at the level of the atherosclerotic plaque itself through mechanisms governing lipid accumulation, plaque stability, local inflammation, endothelial dysfunction, and thrombogenicity. The capacity of lipid-lowering therapies to modify atherosclerotic plaque burden, size, composition, and vulnerability should correlate with their ability to reduce disease progression. This review discusses plaque characteristics, diagnostic modalities to evaluate these characteristics, and how they are altered by current and emerging lipid-lowering therapies, all in human coronary artery disease.
Collapse
Affiliation(s)
- Vishnu Priya Pulipati
- Section of Cardiology, University of Chicago Medicine, 5841 S. Maryland Avenue, MC 6080, Chicago, IL 60637, United States
| | - Francis J. Alenghat
- Section of Cardiology, University of Chicago Medicine, 5841 S. Maryland Avenue, MC 6080, Chicago, IL 60637, United States
- Pritzker School of Medicine, University of Chicago, Chicago, United States
| |
Collapse
|
30
|
Libby P. Inflammation during the life cycle of the atherosclerotic plaque. Cardiovasc Res 2021; 117:2525-2536. [PMID: 34550337 PMCID: PMC8783385 DOI: 10.1093/cvr/cvab303] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation orchestrates each stage of the life cycle of atherosclerotic plaques. Indeed, inflammatory mediators likely link many traditional and emerging risk factors with atherogenesis. Atheroma initiation involves endothelial activation with recruitment of leucocytes to the arterial intima, where they interact with lipoproteins or their derivatives that have accumulated in this layer. The prolonged and usually clinically silent progression of atherosclerosis involves periods of smouldering inflammation, punctuated by episodes of acute activation that may arise from inflammatory mediators released from sites of extravascular injury or infection or from subclinical disruptions of the plaque. Smooth muscle cells and infiltrating leucocytes can proliferate but also undergo various forms of cell death that typically lead to formation of a lipid-rich 'necrotic' core within the evolving intimal lesion. Extracellular matrix synthesized by smooth muscle cells can form a fibrous cap that overlies the lesion's core. Thus, during progression of atheroma, cells not only procreate but perish. Inflammatory mediators participate in both processes. The ultimate clinical complication of atherosclerotic plaques involves disruption that provokes thrombosis, either by fracture of the plaque's fibrous cap or superficial erosion. The consequent clots can cause acute ischaemic syndromes if they embarrass perfusion. Incorporation of the thrombi can promote plaque healing and progressive intimal thickening that can aggravate stenosis and further limit downstream blood flow. Inflammatory mediators regulate many aspects of both plaque disruption and healing process. Thus, inflammatory processes contribute to all phases of the life cycle of atherosclerotic plaques, and represent ripe targets for mitigating the disease.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston, MA, USA
| |
Collapse
|
31
|
Buono MF, Slenders L, Wesseling M, Hartman RJG, Monaco C, den Ruijter HM, Pasterkamp G, Mokry M. The changing landscape of the vulnerable plaque: a call for fine-tuning of preclinical models. Vascul Pharmacol 2021; 141:106924. [PMID: 34607015 DOI: 10.1016/j.vph.2021.106924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/08/2021] [Accepted: 09/28/2021] [Indexed: 11/17/2022]
Abstract
For decades, the pathological definition of the vulnerable plaque led to invaluable insights into the mechanisms that underlie myocardial infarction and stroke. Beyond plaque rupture, other mechanisms, such as erosion, may elicit thrombotic events underlining the complexity and diversity of the atherosclerotic disease. Novel insights, based on single-cell transcriptomics and other "omics" methods, provide tremendous opportunities in the ongoing search for cell-specific determinants that will fine-tune the description of the thrombosis prone lesion. It coincides with an increasing awareness that knowledge on lesion characteristics, cell plasticity and clinical presentation of ischemic cardiovascular events have shifted over the past decades. This shift correlates with an observed changes of cell composition towards phenotypical stabilizing of human plaques. These stabilization features and mechanisms are directly mediated by the cells present in plaques and can be mimicked in vitro via primary plaque cells derived from human atherosclerotic tissues. In addition, the rapidly evolving of sequencing technologies identify many candidate genes and molecular mechanisms that may influence the risk of developing an atherosclerotic thrombotic event - which bring the next challenge in sharp focus: how to translate these cell-specific insights into tangible functional and translational discoveries?
Collapse
Affiliation(s)
- Michele F Buono
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Lotte Slenders
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marian Wesseling
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Robin J G Hartman
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michal Mokry
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands; Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
32
|
Kefer J, Chenu P, Gurné O, Maes F, Tamakloé T, Beauloye C. Reduction of Lipid-Core Burden Index in Nonculprit Lesions at Follow-Up after ST-Elevation Myocardial Infarction: A Randomized Study of Bioresorbable Vascular Scaffold versus Optimal Medical Therapy. J Interv Cardiol 2021; 2021:5590093. [PMID: 34290573 PMCID: PMC8266479 DOI: 10.1155/2021/5590093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/24/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Non-flow-limiting nonculprit lesions (NCL) that contain a large lipid-rich necrotic core (nonculprit lipid-rich plaques (NC-LRP)) are most likely to cause recurrent acute coronary syndrome after ST-elevation myocardial infarction (STEMI). Near-infrared spectroscopy (NIRS) detects LRPs using the maximum 4 mm lipid-core burden index (maxLCBI4 mm). Few data are available regarding NIRS-guided therapy of these NC-LRPs, which are a potential target for preventive stenting. Bioresorbable vascular scaffold (BVS) provides local drug delivery and could facilitate plaque passivation after resorption. This study sought to assess the safety of BVS implantation in NC-LRPs and its efficacy in reducing maxLCBI4 mm at 2-year follow-up after STEMI. METHODS AND RESULTS In total, 33 non-flow-limiting NCLs from 29 STEMI patients were included in this study. Of these, 15 were LRPs and were randomly assigned to either the BVS + optimal medical therapy (OMT) arm (group 1; N = 7) or the OMT arm (group 2; N = 8). At baseline, there were no differences in plaque characteristics between groups (fractional flow reserve: 0.85 ± 0.04 vs. 0.89 ± 0.06; diameter stenosis (DS): 43.4 ± 8 vs. 40.1 ± 10.7%; plaque burden 54.98 ± 5.8 vs. 49.76 ± 8.31%; and maxLCBI4 mm 402 [348; 564] vs. 373 [298; 516]; p=NS for all comparisons between groups 1 and 2, respectively). Seven BVSs were implanted 3 ± 1 days after STEMI in six patients, without complications. At angiographic follow-up (712 [657; 740] days), a significant and similar reduction of maxLCBI4 mm was observed in both groups, with a median change of 306 [257; 377] in group 1 vs. 300 [278; 346] in group 2 (p=0.44). DS was significantly lower in group 1 vs. group 2 (19.8 ± 7 vs. 41.7 ± 13%, p=0.003), while plaque burden remained unchanged in both groups. Overall survival was 100%, target lesion failure was 13%, and stent thrombosis was 0%. CONCLUSIONS BVS + OMT and OMT appear as similarly safe and effective in reducing maxLCBI4mm in NC-LRPs at 2-year follow-up after STEMI.
Collapse
Affiliation(s)
- Joelle Kefer
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Patrick Chenu
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Olivier Gurné
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Frederic Maes
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Théophile Tamakloé
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Christophe Beauloye
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
33
|
Borén J, Chapman MJ, Krauss RM, Packard CJ, Bentzon JF, Binder CJ, Daemen MJ, Demer LL, Hegele RA, Nicholls SJ, Nordestgaard BG, Watts GF, Bruckert E, Fazio S, Ference BA, Graham I, Horton JD, Landmesser U, Laufs U, Masana L, Pasterkamp G, Raal FJ, Ray KK, Schunkert H, Taskinen MR, van de Sluis B, Wiklund O, Tokgozoglu L, Catapano AL, Ginsberg HN. Low-density lipoproteins cause atherosclerotic cardiovascular disease: pathophysiological, genetic, and therapeutic insights: a consensus statement from the European Atherosclerosis Society Consensus Panel. Eur Heart J 2021; 41:2313-2330. [PMID: 32052833 PMCID: PMC7308544 DOI: 10.1093/eurheartj/ehz962] [Citation(s) in RCA: 879] [Impact Index Per Article: 219.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/10/2019] [Accepted: 01/08/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - M John Chapman
- Endocrinology-Metabolism Division, Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, France.,National Institute for Health and Medical Research (INSERM), Paris, France
| | - Ronald M Krauss
- Department of Atherosclerosis Research, Children's Hospital Oakland Research Institute and UCSF, Oakland, CA 94609, USA
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Jacob F Bentzon
- Department of Clinical Medicine, Heart Diseases, Aarhus University, Aarhus, Denmark.,Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Mat J Daemen
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Linda L Demer
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Physiology, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert A Hegele
- Department of Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Australia
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, University of Copenhagen, Denmark
| | - Gerald F Watts
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia.,Department of Cardiology, Lipid Disorders Clinic, Royal Perth Hospital, Perth, Australia
| | - Eric Bruckert
- INSERM UMRS1166, Department of Endocrinology-Metabolism, ICAN - Institute of CardioMetabolism and Nutrition, AP-HP, Hopital de la Pitie, Paris, France
| | - Sergio Fazio
- Departments of Medicine, Physiology and Pharmacology, Knight Cardiovascular Institute, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Brian A Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, UK.,Institute for Advanced Studies, University of Bristol, Bristol, UK.,MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - Jay D Horton
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ulf Landmesser
- Department of Cardiology, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Liebigstraße 20, Leipzig, Germany
| | - Luis Masana
- Research Unit of Lipids and Atherosclerosis, IISPV, CIBERDEM, University Rovira i Virgili, C. Sant Llorenç 21, Reus 43201, Spain
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frederick J Raal
- Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Kausik K Ray
- Department of Primary Care and Public Health, Imperial Centre for Cardiovascular Disease Prevention, Imperial College London, London, UK
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Faculty of Medicine, Technische Universität München, Lazarettstr, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Marja-Riitta Taskinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Olov Wiklund
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lale Tokgozoglu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, and IRCCS MultiMedica, Milan, Italy
| | - Henry N Ginsberg
- Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA
| |
Collapse
|
34
|
Affiliation(s)
- Eric Boersma
- Erasmus MC, Department of Cardiology, Rotterdam, The Netherlands
| | - Isabella Kardys
- Erasmus MC, Department of Cardiology, Rotterdam, The Netherlands
| |
Collapse
|
35
|
Lorenzatti AJ. Anti-inflammatory Treatment and Cardiovascular Outcomes: Results of Clinical Trials. Eur Cardiol 2021; 16:e15. [PMID: 33976710 PMCID: PMC8086421 DOI: 10.15420/ecr.2020.51] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/20/2021] [Indexed: 01/09/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder of the vasculature where cholesterol accumulates in the arterial wall stimulating infiltration of immune cells. This plays an important role in plaque formation, as well as complications caused by its build up. Pro-inflammatory cytokines and chemokines are implicated throughout the progression of the disease and different therapies that aim to resolve this chronic inflammation, reduce cardiovascular (CV) events and improve clinical outcomes have been tested. The results from the pivotal CANTOS trial show that targeting the pro-inflammatory cytokine IL-1β successfully reduces the incidence of secondary CV events. This review briefly assesses the role of inflammation in atherosclerosis, providing a picture of the multiple players involved in the process and offering a perspective on targeting inflammation to prevent atherosclerotic CV events, as well as focusing on the results of the latest Phase III clinical trials.
Collapse
|
36
|
Cismaru G, Serban T, Tirpe A. Ultrasound Methods in the Evaluation of Atherosclerosis: From Pathophysiology to Clinic. Biomedicines 2021; 9:418. [PMID: 33924492 PMCID: PMC8070406 DOI: 10.3390/biomedicines9040418] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/09/2021] [Accepted: 04/10/2021] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is a key pathological process that causes a plethora of pathologies, including coronary artery disease, peripheral artery disease, and ischemic stroke. The silent progression of the atherosclerotic disease prompts for new surveillance tools that can visualize, characterize, and provide a risk evaluation of the atherosclerotic plaque. Conventional ultrasound methods-bright (B)-mode US plus Doppler mode-provide a rapid, cost-efficient way to visualize an established plaque and give a rapid risk stratification of the patient through the Gray-Weale standardization-echolucent plaques with ≥50% stenosis have a significantly greater risk of ipsilateral stroke. Although rather disputed, the measurement of carotid intima-media thickness (C-IMT) may prove useful in identifying subclinical atherosclerosis. In addition, contrast-enhanced ultrasonography (CEUS) allows for a better image resolution and the visualization and quantification of plaque neovascularization, which has been correlated with future cardiovascular events. Newly emerging elastography techniques such as strain elastography and shear-wave elastography add a new dimension to this evaluation-the biomechanics of the arterial wall, which is altered in atherosclerosis. The invasive counterpart, intravascular ultrasound (IVUS), enables an individualized assessment of the anti-atherosclerotic therapies, as well as a direct risk assessment of these lesions through virtual histology IVUS.
Collapse
Affiliation(s)
- Gabriel Cismaru
- Fifth Department of Internal Medicine, Cardiology-Rehabilitation, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Teodora Serban
- Medical Imaging Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania;
| | - Alexandru Tirpe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| |
Collapse
|
37
|
Bajaj R, Garcia-Garcia HM, Courtney BK, Ramasamy A, Tufaro V, Erdogan E, Khan AH, Alves N, Rathod KS, Onuma Y, Serruys PW, Mathur A, Baumbach A, Bourantas C. Multi-modality intravascular imaging for guiding coronary intervention and assessing coronary atheroma: the Novasight Hybrid IVUS-OCT system. Minerva Cardiol Angiol 2021; 69:655-670. [PMID: 33703857 DOI: 10.23736/s2724-5683.21.05532-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Intravascular imaging has evolved alongside interventional cardiology as an adjunctive tool for assessing plaque pathology and for guiding and optimising percutaneous coronary intervention (PCI) in challenging lesions. The two modalities which have dominated the field are intravascular ultrasound (IVUS), which relies on sound waves and optical coherence tomography (OCT), relying on light waves. These approaches however have limited efficacy in assessing plaque morphology and vulnerability that are essential for guiding PCI in complex lesions and identifying patient at risk that will benefit from emerging therapies targeting plaque evolution. These limitations are complementary and, in this context, it has been recognised and demonstrated in multi-modality studies that the concurrent use of IVUS and OCT can help overcome these deficits enabling a more complete and accurate plaque assessment. The Conavi Novasight Hybrid IVUS-OCT catheter is the first commercially available device that is capable of invasive clinical coronary assessment with simultaneously acquired and co-registered IVUS and OCT imaging. It represents a significant evolution in the field and is expected to have broad application in clinical practice and research. In this review article we present the limitations of standalone intravascular imaging techniques, summarise the data supporting the value of multimodality imaging in clinical practice and research, describe the Novasight Hybrid IVUS-OCT system and highlight the potential utility of this technology in coronary intervention and in the study of atherosclerosis.
Collapse
Affiliation(s)
- Retesh Bajaj
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Brian K Courtney
- Sunnybrook Research Institute, Schulich Heart Program, University of Toronto, Toronto, ON, Canada.,Conavi Medical, North York, ON, Canada
| | - Anantharaman Ramasamy
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Vincenzo Tufaro
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Emrah Erdogan
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Ameer H Khan
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Natasha Alves
- Sunnybrook Research Institute, Schulich Heart Program, University of Toronto, Toronto, ON, Canada
| | - Krishnaraj S Rathod
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Yoshinobu Onuma
- School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Patrick W Serruys
- School of Medicine, National University of Ireland Galway, Galway, Ireland.,National Heart & Lung Institute, Imperial College London, London, UK
| | - Anthony Mathur
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Andreas Baumbach
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK.,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Christos Bourantas
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK - .,Cardiovascular Devices Hub, Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|
38
|
van 't Klooster CC, van der Graaf Y, Nathoe HM, Bots ML, de Borst GJ, Visseren FLJ, Leiner T. Added value of cardiovascular calcifications for prediction of recurrent cardiovascular events and cardiovascular interventions in patients with established cardiovascular disease. Int J Cardiovasc Imaging 2021; 37:2051-2061. [PMID: 33580447 PMCID: PMC8255266 DOI: 10.1007/s10554-021-02164-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/15/2021] [Indexed: 12/23/2022]
Abstract
The purpose is to investigate the added prognostic value of coronary artery calcium (CAC), thoracic aortic calcium (TAC), and heart valve calcium scores for prediction of a combined endpoint of recurrent major cardiovascular events and cardiovascular interventions (MACE +) in patients with established cardiovascular disease (CVD). In total, 567 patients with established CVD enrolled in a substudy of the UCC-SMART cohort, entailing cardiovascular CT imaging and calcium scoring, were studied. Five Cox proportional hazards models for prediction of 4-year risk of MACE + were developed; traditional CVD risk predictors only (model I), with addition of CAC (model II), TAC (model III), heart valve calcium (model IV), and all calcium scores (model V). Bootstrapping was performed to account for optimism. During a median follow-up of 3.43 years (IQR 2.28–4.74) 77 events occurred (MACE+). Calibration of predicted versus observed 4-year risk for model I without calcium scores was good, and the c-statistic was 0.65 (95%CI 0.59–0.72). Calibration for models II–V was similar to model I, and c-statistics were 0.67, 0.65, 0.65, and 0.68 for model II, III, IV, and V, respectively. NRIs showed improvement in risk classification by model II (NRI 15.24% (95%CI 0.59–29.39)) and model V (NRI 20.00% (95%CI 5.59–34.92)), but no improvement for models III and IV. In patients with established CVD, addition of the CAC score improved performance of a risk prediction model with classical risk factors for the prediction of the combined endpoint MACE+ . Addition of the TAC or heart valve score did not improve risk predictions.
Collapse
Affiliation(s)
- Cilie C van 't Klooster
- Department of Vascular Medicine, University Medical Center Utrecht (UMCU), Utrecht University, PO Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Yolanda van der Graaf
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
| | - Hendrik M Nathoe
- Department of Cardiology, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
| | - Michiel L Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
| | - Gert J de Borst
- Department of Vascular Surgery, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
| | - Frank L J Visseren
- Department of Vascular Medicine, University Medical Center Utrecht (UMCU), Utrecht University, PO Box 85500, 3508 GA, Utrecht, The Netherlands.
| | - Tim Leiner
- Department of Radiology, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
39
|
Abstract
The development of potent cholesterol-reducing medications in the last decade of the twentieth century has altered the approach to prevention and treatment of cardiovascular disease (CVD). Initial experience with statins, and more recently with the addition of PCSK9 inhibitors, has proven that human CVD, like that in animal models, can be halted and regressed. Available clinical data show that the lower the achieved level of low-density lipoprotein cholesterol, the greater the regression of disease. Investigative studies are now aimed to understand those factors that both accelerate and impede this healing process. Some of these are likely to be modifiable, and the future of atherosclerotic CVD treatment is likely to be early screening, use of measures to repair atherosclerotic arteries, and prevention of most CVD events.
Collapse
Affiliation(s)
- Ira J Goldberg
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA;
| | - Gaurav Sharma
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA;
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA;
| |
Collapse
|
40
|
Pérez de Isla L, Alonso R, Gómez de Diego JJ, Muñiz-Grijalvo O, Díaz-Díaz JL, Zambón D, Miramontes JP, Fuentes F, de Andrés R, Werenitzky J, Padró T, Saltijeral A, Mata P. Coronary plaque burden, plaque characterization and their prognostic implications in familial hypercholesterolemia: A computed tomographic angiography study. Atherosclerosis 2020; 317:52-58. [PMID: 33261814 DOI: 10.1016/j.atherosclerosis.2020.11.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/20/2020] [Accepted: 11/12/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS Heterozygous familial hypercholesterolemia (FH) is associated with premature atherosclerotic cardiovascular disease. Semi-automated plaque characterization (SAPC) by coronary computed tomographic angiography (CTA) provides information regarding coronary plaque burden and plaque characterization. Our aim was to quantify and characterize the coronary plaque burden of patients with FH using SAPC analysis and to identify which factors are related to plaque burden and plaque characteristics. A second aim was to analyse the prognostic implications of these parameters. METHODS Two hundred and fifty-nine asymptomatic individuals with molecularly determined FH were enrolled in this follow-up cohort study and underwent a coronary CTA analysed with SAPC. RESULTS Mean follow-up time after coronary CTA was 3.9 ± 2 years. Mean age was 46.9 (10.7) years (130 women, 50.2%). Median plaque burden was 25.0% (19.0-29.0), non-calcified plaque burden 22.83% (17.94-26.88), calcified plaque-burden 1.12% (0.31-2.86) and CCS 8.9 (0-93). Five-year risk was independently related to plaque burden, non-calcified plaque burden, calcified plaque burden and coronary calcium score (B:3.75, 95%CI:2.92-4.58; p < 0.001, B:2.9, 95%CI:2.15-3.66; p < 0.001, B:0.75, 95%CI 0.4-1.1; p < 0.001 and B:82.2, 95%CI:49.28-115.16; p < 0.001 respectively). During follow-up, there were 15 (5.81%) nonfatal events and 1 (0.4%) fatal event. Plaque burden was significantly related to event-free survival during follow-up (HR:1.11; 95%CI:1.05-1.18; p < 0.001). CONCLUSIONS Coronary atherosclerosis and its qualitative components may be quantified by means of SAPC in patients with FH. Plaque burden, calcified plaque burden and non-calcified plaque burden were independently related to the estimated cardiovascular risk. Plaque burden was also related to prognosis.
Collapse
Affiliation(s)
- Leopoldo Pérez de Isla
- Cardiology Department, Hospital Clínico San Carlos, IDISSC, Facultad de Medicina Universidad Complutense, Madrid, Spain; Fundación Hipercolesterolemia Familiar, Madrid, Spain.
| | - Rodrigo Alonso
- Fundación Hipercolesterolemia Familiar, Madrid, Spain; Center for Advanced Metabolic Medicine and Nutrition, Santiago de Chile, Chile
| | - José Juan Gómez de Diego
- Cardiology Department, Hospital Clínico San Carlos, IDISSC, Facultad de Medicina Universidad Complutense, Madrid, Spain
| | | | | | - Daniel Zambón
- Lipids Clinic. Department of Endocrinology, Hospital Clinic, (IDIBAPS) Institut D'Investigacions Biomèdiques August Pi I Sunyer University of Barcelona, Barcelona, Spain
| | | | - Francisco Fuentes
- Lipids and Atherosclerosis Unit, CIBERObn, IMIBIC/Reina Sofia University Hospital/University of Cordoba, Spain
| | | | - José Werenitzky
- Cardiology Department, Hospital Clínico San Carlos, IDISSC, Facultad de Medicina Universidad Complutense, Madrid, Spain
| | - Teresa Padró
- Programa-ICCC Cardiovascular, Institut de Recerca Del Hospital Santa Creu I Sant Pau, IIB Santa Pau, Barcelona, Spain
| | - Adriana Saltijeral
- Cardiology Department, Hospital Del Tajo, Aranjuez, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain.
| | | |
Collapse
|
41
|
Kilic ID, Fabris E, Kedhi E, Ghilencea LN, Caiazzo G, Sherif SA, Di Mario C. Intra-coronary Imaging for the Evaluation of Plaque Modifications Induced by Drug Therapies for Secondary Prevention. Curr Atheroscler Rep 2020; 22:76. [PMID: 33025069 PMCID: PMC7538414 DOI: 10.1007/s11883-020-00890-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Patients diagnosed with coronary artery disease are at a high risk of subsequent cardiovascular events; therefore, secondary prevention in the form of therapeutic lifestyle changes, and drug therapies is vital. This article aims to review potential application of intra-coronary imaging for the evaluation of plaque modifications, induced by medications for secondary prevention for CAD. RECENT FINDINGS Intra-coronary imaging provides detailed information on the atherosclerotic plaque which is the primary pathological substrate for the recurrent ischemic cardiovascular events. These modalities can detect features associated with high risk and allow serial in vivo imaging of lesions. Therefore, intravascular imaging tools have been used in landmark studies and played a role in improving our understanding of the disease processes. Changes in size and plaque composition over time can be evaluated by these tools and may help understanding the impact of a treatment. Moreover, surrogate imaging end points can be used when testing new drugs for secondary prevention.
Collapse
Affiliation(s)
- Ismail Dogu Kilic
- Department of Cardiology, Pamukkale University Hospitals, Denizli, Turkey
| | - Enrico Fabris
- Cardiovascular Department, University of Trieste, Trieste, Italy
| | - Elvin Kedhi
- Department of Cardiology, Isala Heart Center, Zwolle, the Netherlands
| | | | | | | | - Carlo Di Mario
- Cardio-toraco-vascular Department, Careggi University Hospital, Florence, Italy
| |
Collapse
|
42
|
C-reactive protein levels and plaque regression with evolocumab: Insights from GLAGOV. Am J Prev Cardiol 2020; 3:100091. [PMID: 34327467 PMCID: PMC8315612 DOI: 10.1016/j.ajpc.2020.100091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Objective On-treatment levels of high sensitivity C-reactive protein (hsCRP) in statin-treated patients predict plaque progression and the prospective risk of atherosclerotic cardiovascular events. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors produce additional LDL-C lowering, reduce plaque burden and improve cardiovascular outcomes in statin-treated patients. It is unknown whether residual systemic inflammation attenuates their favorable effects on plaque burden. Methods GLAGOV compared the effects of treatment for 78 weeks with evolocumab or placebo on progression of coronary atherosclerosis in statin-treated patients with coronary artery disease. Clinical demographics, biochemistry and changes in both the burden (percentage atheroma volume (PAV), total atheroma volume (TAV), n = 413) and composition (n = 162) of coronary plaque were evaluated in evolocumab-treated patients according to baseline hsCRP strata (<1, 1–3, >3 mg/L). Results The study cohort comprised 413 evolocumab-treated patients (32% low [<1 mg/L], 41% intermediate [1–3 mg/L] and 27% high [>3 mg/L] baseline hsCRP levels). Patients in the highest hsCRP stratum were more likely to be female and had a higher prevalence of diabetes, hypertension, and the metabolic syndrome. LDL-C levels were similar across the groups, however participants with higher hsCRP levels had higher triglyceride and lower HDL-C levels at baseline. At follow-up, the change in PAV from baseline (−0.87% [low] vs. −0.84% [intermediate] vs. −1.22% [high], p = 0.46) and the proportion of patients experiencing any degree of regression (65.9% vs. 63.5% vs. 63.1%, p = 0.88) was similar across hsCRP strata and when evaluated by levels of achieved LDL-C. There were no serial differences in plaque composition by hsCRP strata. Conclusion The ability of evolocumab to induce regression in statin-treated patients is not attenuated by the presence of enhanced systemic inflammation. This underscores the potential benefits of intensive lipid lowering, even in the presence of heightened inflammatory states.
Collapse
|
43
|
Kitahara S, Kataoka Y, Sugane H, Otsuka F, Asaumi Y, Noguchi T, Yasuda S. In vivo imaging of vulnerable plaque with intravascular modalities: its advantages and limitations. Cardiovasc Diagn Ther 2020; 10:1461-1479. [PMID: 33224768 DOI: 10.21037/cdt-20-238] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In vivo imaging of plaque instability has been considered to have a great potential to predict future coronary events and evaluate the stabilization effect of novel anti-atherosclerotic medical therapies. Currently, there are several intravascular imaging modalities which enable to visualize plaque components associated with its vulnerability. These include virtual histology intravascular ultrasound (VH-IVUS), integrated backscatter IVUS (IB-IVUS), optical coherence tomography (OCT), near-infrared spectroscopy and coronary angioscopy. Recent studies have shown that these tools are applicable for risk stratification of cardiovascular events as well as drug efficacy assessment. However, several limitation exists in each modality. The current review paper will outline advantages and limitation of VH-IVUS, IB-IVUS, OCT, NIRS and coronary angioscopy imaging.
Collapse
Affiliation(s)
- Satoshi Kitahara
- Department of Cardiovascular Medicine, National Cerebral & Cardiovascular Center, Suita, Osaka, Japan
| | - Yu Kataoka
- Department of Cardiovascular Medicine, National Cerebral & Cardiovascular Center, Suita, Osaka, Japan
| | - Hiroki Sugane
- Department of Cardiovascular Medicine, Chikamori Hospital, Kochi, Japan
| | - Fumiyuki Otsuka
- Department of Cardiovascular Medicine, National Cerebral & Cardiovascular Center, Suita, Osaka, Japan
| | - Yasuhide Asaumi
- Department of Cardiovascular Medicine, National Cerebral & Cardiovascular Center, Suita, Osaka, Japan
| | - Teruo Noguchi
- Department of Cardiovascular Medicine, National Cerebral & Cardiovascular Center, Suita, Osaka, Japan
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, National Cerebral & Cardiovascular Center, Suita, Osaka, Japan
| |
Collapse
|
44
|
Alvarez C, Keh E, Li Y, Siu H. Significant Difference in Acute Coronary Lesions in Patients on Therapeutic Statin Therapy Compared to Those Without. Cureus 2020; 12:e10673. [PMID: 33133839 PMCID: PMC7592525 DOI: 10.7759/cureus.10673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/16/2020] [Indexed: 11/05/2022] Open
Abstract
Background Statin therapy has been shown to alter coronary plaque via decreasing lipid-rich pools, substituting with fibrocalcific plaque. We propose that acute coronary lesions in patients on therapeutic statin therapy, compared with patients not on statin therapy with elevated low-density lipoprotein (LDL) levels, will have features typically not associated with lipid-rich disease, such as lengthy fibrocalcific lesions, stent failure (in-stent restenosis and stent thrombosis), and/or bypass graft degeneration. Methods Charts and coronary angiograms of 143 consecutive patients from May 2016 to December 2018 with Type 1 Myocardial Infarction Fourth Universal Definition were retrospectively reviewed. Patients were divided into two groups: group 1, those on statin therapy with an LDL < 100 mg/dL, and group 2, those not on statin therapy with an LDL ≥ 100 mg/dL at the time of an acute coronary syndrome. Acute lesion characteristics and angioplasty techniques/equipment were recorded. Results There were 56 patients in group 1 and 39 patients in group 2. Group 1 patients, compared with group 2, were more likely to have moderate-severe vessel calcification (55.4% vs 10.3%; p < 0.01), lesion length greater than 20 mm (51.8% vs 23.1%; p = 0.019), stent failure (39.3% vs 7.7%; p < 0.01), and bypass graft failure (23.2% vs 7.7%; p = 0.04). Coronary interventions in group 1, compared with group 2, more often required adjunctive angioplasty techniques, including the need for multiple coronary wires, guide extender, thrombectomy, and circulatory support (51.8% vs 7.7%; p < 0.01). There was no significant difference between the number of culprit vessels, including the specific coronary culprit vessel identified between both groups. Conclusion Acute coronary lesions in patients on therapeutic statin therapy, compared to those not on statin therapy with an elevated LDL, tend to be longer, more calcific with increased vessel tortuosity and angulation, as well as from a stent or bypass graft failure mechanism. Furthermore, there is a signal for heightened procedural complexity in the arm with statin therapy. These clinical findings are likely the result of the altered natural history of plaque pathophysiology seen with statin therapy.
Collapse
Affiliation(s)
- Chikezie Alvarez
- Cardiology, Hartford Hospital/University of Connecticut, Hartford, USA
| | - Ernestine Keh
- Internal Medicine, St. Francis Medical Center, Trenton, USA
| | - Yiting Li
- Internal Medicine, St. Francis Medical Center, Trenton, USA
| | - Henry Siu
- Interventional Cardiology, Chinatown Cardiology, P.C., New York, USA
| |
Collapse
|
45
|
Bonaterra GA, Bender K, Wilhelm B, Schwarzbach H, Metz S, Kelber O, Weiser D, Metz J, Kinscherf R. Effect of cholesterol re-supplementation and atorvastatin on plaque composition in the thoracic aorta of New Zealand white rabbits. BMC Cardiovasc Disord 2020; 20:420. [PMID: 32942987 PMCID: PMC7499881 DOI: 10.1186/s12872-020-01703-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/10/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Effects of re-supplementation of a cholesterol-enriched diet (CEDrs) on size, cholesterol content and morphology of already existing plaques are not known to date. METHODS A group of rabbits received standard chow (SC) for 6 weeks ("negative control"; for plasma lipid measurements only). Group I-IV received 2% CED (induction) for 6 weeks; thereafter, groups II-IV have been fed a SC (= cholesterol withdrawal) for 68 weeks. Afterwards, feeding of groups II-IV was continued as follows: Group II - 10 weeks SC, group III - 4 weeks 0.5% CED (~re-supplementation), afterwards 6 weeks SC (~withdrawal again); group IV - 4 weeks 0.5% CED (re-supplementation) + atorvastatin (2.5 mg/kg body weight/day), afterwards 6 weeks SC (~withdrawal again) + atorvastatin. Plasma lipids, but also plaque size, morphology and cholesterol contents of thoracic aortas were quantified. RESULTS After CEDrs, plasma cholesterol levels were increased. However, after withdrawal of CEDrs, plasma cholesterol levels decreased, whereas the cholesterol content of the thoracic aorta was increased in comparison with the group without CEDrs. Plaque size remained unaffected. Atorvastatin application did not change plasma cholesterol level, cholesterol content of the thoracic aorta and plaque size in comparison with the group without drug treatment. However, atorvastatin treatment increased the density of macrophages (MΦ) compared with the group without treatment, with a significant correlation between densities of MΦ (Mac-1+) and apoptotic (TUNEL+; TP53+), antigen-presenting (HLA-DR+) or oxidatively stressed (SOD2+) cells. CONCLUSIONS In rabbits with already existing plaques, CEDrs affects plaque morphology and cellular composition, but not plaque size. Despite missing effects on plasma cholesterol levels, cholesterol content of the thoracic aorta and size of already existing atherosclerotic plaques, atorvastatin treatment transforms the already existing lesions to a more active form, which may accelerate the remodelling to a more stable plaque.
Collapse
Affiliation(s)
- G A Bonaterra
- Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany.
| | - K Bender
- Department of Anatomy and Cell Biology III, University of Heidelberg, 69120, Heidelberg, Germany
| | - B Wilhelm
- Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| | - H Schwarzbach
- Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| | - S Metz
- Department of Radiology, Technical University, 81675, Munich, Germany
| | - O Kelber
- Steigerwald Arzneimittelwerk, 64295, Darmstadt, Germany
| | - D Weiser
- Steigerwald Arzneimittelwerk, 64295, Darmstadt, Germany
| | - J Metz
- Department of Anatomy and Cell Biology III, University of Heidelberg, 69120, Heidelberg, Germany
| | - R Kinscherf
- Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| |
Collapse
|
46
|
Tsai S, Vega GL. Coronary and peripheral artery plaques: do differences in plaque characteristics translate to differences in lipid management? J Investig Med 2020; 68:1141-1151. [DOI: 10.1136/jim-2019-001252] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2020] [Indexed: 12/22/2022]
Abstract
Optimal medical management of patients with peripheral arterial disease (PAD) includes statin therapy, which has been shown to decrease the risk of major cardiovascular events. However, the relationship between low-density lipoprotein (LDL) lowering, PAD progression and limb outcomes remains controversial. Although prevention of coronary and cerebrovascular events is a priority, limb outcomes are still important determinants of quality of life and healthcare spending. This review will highlight differences between coronary artery disease (CAD) and PAD, and in particular, the more prevalent role of lipids and LDL cholesterol in CAD versus calcification in PAD. This difference may contribute to the differential impact of LDL cholesterol levels on coronary events and outcomes versus limb outcomes. Beyond LDL lowering, immune modulators have emerged as another agent to treat atherosclerosis in CAD, however similar data in PAD are lacking. Small studies have suggested that other lipids besides LDL cholesterol, such as triglycerides or small dense LDL, may have a greater impact on limb outcomes in patients with PAD. Although statin therapy is central in the management of patients with PAD, current understanding of the distinctions between PAD and CAD suggest that there may be other non-LDL targets for risk reduction that require further study.
Collapse
|
47
|
Bruckert E, Parhofer KG, Gonzalez-Juanatey JR, Nordestgaard B, Arca M, Giovas P, Ray K. Proportion of High-Risk/Very High-Risk Patients in Europe with Low-Density Lipoprotein Cholesterol at Target According to European Guidelines: A Systematic Review. Adv Ther 2020; 37:1724-1736. [PMID: 32200537 PMCID: PMC7467492 DOI: 10.1007/s12325-020-01285-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Assess achievement of low-density lipoprotein cholesterol (LDL-C) targets in European Society of Cardiology (ESC)/European Atherosclerosis Society (EAS) guidelines. DESIGN Systematic literature review. DATA SOURCES Medline, EMBASE, Cumulated Index to Nursing and Allied Health Literature. ELIGIBILITY CRITERIA Observational studies reporting LDL-C levels/target attainment, measured between 1 August 2006 to 31 August 2017, in European adults with established cardiovascular disease (CVD), diabetes with target organ damage, familial hypercholesterolaemia (FH) or 10-year risk of fatal CVD ≥ 5% (assessed by Systematic Coronary Risk Evaluation [SCORE]). DATA EXTRACTION AND SYNTHESIS Two reviewers independently extracted relevant studies and assessed study quality using the Risk of Bias for Non-Randomised Studies-Interventions (ROBINS-I) tool. Primary outcome was the proportion of patients achieving LDL-C targets in the 2011/2016 ESC/EAS guidelines. Where available, patient characteristics were presented as means weighted by sample size. The proportions of patients achieving LDL-C targets in the 5 years before and after publication of the 2011 guidelines were compared using a chi-square test. RESULTS Across 81 eligible studies (303,534 patients), achievement of LDL-C < 1.8 mmol/L was poor among patients with established CVD (16%; range 9-56%) and at very high risk of CVD (SCORE ≥ 10% [18%; 14-25%]). In individuals with FH, SCORE 5-10%, or diabetes and target organ damage, LDL-C < 2.5 mmol/L was achieved by 15% (9-22%), 46% (21-55%) and 13% (6-34%), respectively. Comparing the 5 years before/after publication of the 2011 guidelines, target achievement increased significantly over time but remained suboptimal (LDL-C < 1.8, 22% versus 15%; LDL-C < 2.5, 68% versus 61%; both p < 0.001; established CVD group only). CONCLUSIONS These data show suboptimal LDL-C control among European patients at high risk of CVD. Those at greatest overall risk (clinically established CVD or at least a 10% 10-year risk of fatal CVD) had the lowest achievement of 2011/2016 EAS/ESC LDL-C targets. With lower LDL-C targets advocated in 2019 ESC/EAS guidelines, this unmet need will increase. PROTOCOL REGISTRATION PROSPERO registration number; CRD77844.
Collapse
Affiliation(s)
- Eric Bruckert
- Endocrinologie Métabolisme et Prévention Cardiovasculaire, Institut E3M et IHU Cardiométabolique (ICAN), Hôpital Pitié Salpêtrière, Paris, France.
| | - Klaus Georg Parhofer
- Medizinische Klinik IV-Grosshadern, Klinikum der Universität München, Marchioninistr. 15, 81377, Munich, Germany
| | | | - Børge Nordestgaard
- Department of Clinical Biochemistry, Faculty of Health and Medical Sciences, Herlev and Gentofte Hospital, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Marcello Arca
- Department of Internal Medicine and Medical Specialties, UOS Atherosclerosis Center, La Sapienza University of Rome, Rome, Italy
| | | | - Kausik Ray
- Department of Public Health and Primary Care, Imperial Centre for Cardiovascular Disease Prevention, Imperial College London, London, UK
| |
Collapse
|
48
|
Shishikura D, Kataoka Y, Di Giovanni G, Takata K, Scherer DJ, Andrews J, Psaltis PJ, Puri R, Wolski K, Nissen SE, Nicholls SJ. Progression of ultrasound plaque attenuation and low echogenicity associates with major adverse cardiovascular events. Eur Heart J 2020; 41:2965-2973. [DOI: 10.1093/eurheartj/ehaa173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/05/2019] [Accepted: 03/04/2020] [Indexed: 02/02/2023] Open
Abstract
Abstract
Aims
Intravascular ultrasound (IVUS) imaging can visualize vulnerable plaque features including attenuation (AP) and echolucency (ELP). While IVUS-derived vulnerable plaque features associate with microvascular obstruction during percutaneous coronary intervention, the relationship between these plaque features and clinical outcomes has not been established. This analysis aimed to evaluate the association of AP/ELP with cardiovascular events.
Methods and results
Serial IVUS imaging was reviewed in 1497 patients, followed for 18–24 months, with coronary artery disease from two clinical trials. Attenuated plaque and ELP were identified to measure each characteristics (AP arc, ELP area, and lengths), which permitted calculation of an AP index (API) and ELP volume. Attenuated plaque/ELP progression was defined as patients with any increase of API or ELP volume on serial imaging. The major cardiovascular events (MACEs) were defined as death, myocardial infarction, stroke, and coronary revascularization. AP or ELP was identified in 282 patients (18.8%) at baseline and 160 (10.7%) patients demonstrated an increase in AP or ELP at follow-up. The incidence of MACE was higher in patients with baseline AP/ELP than those without (8.2% vs. 3.9%, P = 0.002). Patients with AP/ELP progression were more likely to be acute coronary syndrome (41.9 vs. 33.2%, P = 0.03) and have greater baseline percent atheroma volume (40.0% vs. 35.8%, P < 0.001) than those without. On multivariable analysis, AP/ELP progression was more strongly associated with MACE [baseline AP/ELP: hazard ratio (HR) 1.76, 95% confidence interval (CI) 1.05–2.97, AP/ELP progression: HR 2.19, 95% CI 1.24–3.86].
Conclusion
Attenuation/ELP progression was associated with a higher prevalence of cardiovascular events, supporting a potential role for the identification of high-risk vulnerable plaques in patients with coronary artery disease.
Collapse
Affiliation(s)
- Daisuke Shishikura
- Heart Health Them, South Australian Health & Medical Research Institute, University of Adelaide, North Terrace, Adelaide, SA 5001, Australia
| | - Yu Kataoka
- Heart Health Them, South Australian Health & Medical Research Institute, University of Adelaide, North Terrace, Adelaide, SA 5001, Australia
| | - Giuseppe Di Giovanni
- Heart Health Them, South Australian Health & Medical Research Institute, University of Adelaide, North Terrace, Adelaide, SA 5001, Australia
| | - Kohei Takata
- Heart Health Them, South Australian Health & Medical Research Institute, University of Adelaide, North Terrace, Adelaide, SA 5001, Australia
| | - Daniel J Scherer
- Heart Health Them, South Australian Health & Medical Research Institute, University of Adelaide, North Terrace, Adelaide, SA 5001, Australia
| | - Jordan Andrews
- Heart Health Them, South Australian Health & Medical Research Institute, University of Adelaide, North Terrace, Adelaide, SA 5001, Australia
| | - Peter J Psaltis
- Heart Health Them, South Australian Health & Medical Research Institute, University of Adelaide, North Terrace, Adelaide, SA 5001, Australia
| | - Rishi Puri
- Department of Cardiovascular Medicine, Cleveland Clinic Coordinating Centre for Clinical Research, Euclid Avenue, Cleveland, OH 44195, USA
| | - Kathy Wolski
- Department of Cardiovascular Medicine, Cleveland Clinic Coordinating Centre for Clinical Research, Euclid Avenue, Cleveland, OH 44195, USA
| | - Steven E Nissen
- Department of Cardiovascular Medicine, Cleveland Clinic Coordinating Centre for Clinical Research, Euclid Avenue, Cleveland, OH 44195, USA
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, 246 Clayton Rd, Clayton, Victoria 3168, Australia
| |
Collapse
|
49
|
Tajbakhsh A, Kovanen PT, Rezaee M, Banach M, Sahebkar A. Ca 2+ Flux: Searching for a Role in Efferocytosis of Apoptotic Cells in Atherosclerosis. J Clin Med 2019; 8:2047. [PMID: 31766552 PMCID: PMC6947386 DOI: 10.3390/jcm8122047] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/09/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
In atherosclerosis, macrophages in the arterial wall ingest plasma lipoprotein-derived lipids and become lipid-filled foam cells with a limited lifespan. Thus, efficient removal of apoptotic foam cells by efferocytic macrophages is vital to preventing the dying foam cells from forming a large necrotic lipid core, which, otherwise, would render the atherosclerotic plaque vulnerable to rupture and would cause clinical complications. Ca2+ plays a role in macrophage migration, survival, and foam cell generation. Importantly, in efferocytic macrophages, Ca2+ induces actin polymerization, thereby promoting the formation of a phagocytic cup necessary for efferocytosis. Moreover, in the efferocytic macrophages, Ca2+ enhances the secretion of anti-inflammatory cytokines. Various Ca2+ antagonists have been seminal for the demonstration of the role of Ca2+ in the multiple steps of efferocytosis by macrophages. Moreover, in vitro and in vivo experiments and clinical investigations have revealed the capability of Ca2+ antagonists in attenuating the development of atherosclerotic plaques by interfering with the deposition of lipids in macrophages and by reducing plaque calcification. However, the regulation of cellular Ca2+ fluxes in the processes of efferocytic clearance of apoptotic foam cells and in the extracellular calcification in atherosclerosis remains unknown. Here, we attempted to unravel the molecular links between Ca2+ and efferocytosis in atherosclerosis and to evaluate cellular Ca2+ fluxes as potential treatment targets in atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Halal Research Center of IRI, FDA, Tehran, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mahdi Rezaee
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948, Iran
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland
- Polish Mother’s Memorial Hospital Research Institute (PMMHRI), 93-338 Lodz, Poland
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948, Iran
| |
Collapse
|
50
|
Jaminon A, Reesink K, Kroon A, Schurgers L. The Role of Vascular Smooth Muscle Cells in Arterial Remodeling: Focus on Calcification-Related Processes. Int J Mol Sci 2019; 20:E5694. [PMID: 31739395 PMCID: PMC6888164 DOI: 10.3390/ijms20225694] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022] Open
Abstract
Arterial remodeling refers to the structural and functional changes of the vessel wall that occur in response to disease, injury, or aging. Vascular smooth muscle cells (VSMC) play a pivotal role in regulating the remodeling processes of the vessel wall. Phenotypic switching of VSMC involves oxidative stress-induced extracellular vesicle release, driving calcification processes. The VSMC phenotype is relevant to plaque initiation, development and stability, whereas, in the media, the VSMC phenotype is important in maintaining tissue elasticity, wall stress homeostasis and vessel stiffness. Clinically, assessment of arterial remodeling is a challenge; particularly distinguishing intimal and medial involvement, and their contributions to vessel wall remodeling. The limitations pertain to imaging resolution and sensitivity, so methodological development is focused on improving those. Moreover, the integration of data across the microscopic (i.e., cell-tissue) and macroscopic (i.e., vessel-system) scale for correct interpretation is innately challenging, because of the multiple biophysical and biochemical factors involved. In the present review, we describe the arterial remodeling processes that govern arterial stiffening, atherosclerosis and calcification, with a particular focus on VSMC phenotypic switching. Additionally, we review clinically applicable methodologies to assess arterial remodeling and the latest developments in these, seeking to unravel the ubiquitous corroborator of vascular pathology that calcification appears to be.
Collapse
Affiliation(s)
- Armand Jaminon
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Koen Reesink
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Abraham Kroon
- Department of Internal Medicine, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands;
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|