1
|
Bergmark BA, Marston NA, Prohaska TA, Alexander VJ, Zimerman A, Moura FA, Kang YM, Murphy SA, Zhang S, Lu MT, Karwatowska-Prokopczuk E, Tsimikas S, Giugliano RP, Sabatine MS. Olezarsen in patients with hypertriglyceridemia at high cardiovascular risk: Rationale and design of the Essence-TIMI 73b trial. Am Heart J 2025; 286:116-124. [PMID: 40081744 PMCID: PMC12065083 DOI: 10.1016/j.ahj.2025.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/30/2025] [Accepted: 02/18/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Elevated triglycerides are an important risk factor for atherosclerosis. However, the magnitude of triglyceride lowering with currently available therapies is modest and the impact of triglyceride-lowering on atherosclerosis remains undefined. Olezarsen is an antisense oligonucleotide (ASO) targeting mRNA for apolipoprotein C-III (apoC-III), an inhibitor of triglyceride clearance. METHODS The Essence-TIMI 73b trial (NCT05610280) is a randomized, double-blind, placebo-controlled phase 3 trial of olezarsen 50 mg or 80 mg every 4 weeks compared with placebo. The trial enrolled adults with either moderate hypertriglyceridemia (200-499 mg/dL) plus increased cardiovascular risk, or severe hypertriglyceridemia (≥500 mg/dL). The primary endpoint is the percent change in triglyceride levels from baseline to 6 months, reported as the difference between each olezarsen dose group and pooled placebo. A coronary computed tomography angiography (CTA) substudy will examine changes in noncalcified plaque volume from baseline to 12 months. RESULTS A total of 1,478 patients were randomized at 160 sites in North America and Europe. The median age is 63 (IQR 56-69) years, 39% are women, and 71% are non-Hispanic White. Overall, 60% of patients have diabetes, and 42% have atherosclerotic cardiovascular disease. At randomization, 97% were receiving lipid-lowering therapies, including 82% on a statin. The median baseline triglyceride level was 249 (195-339) mg/dL and 9% of patients had triglycerides ≥500 mg/dL at enrollment. Approximately 1000 patients completed a baseline CTA, of whom 555 (55%) had measurable noncalcified coronary plaque and continued in the substudy. DISCUSSION Targeting apoC-III to facilitate clearance of triglyceride-rich lipoproteins is a potential therapeutic strategy for lowering triglyceride levels, regressing atherosclerosis, and reducing cardiovascular risk. The phase 3 Essence-TIMI 73b trial, which has enrolled nearly 1,500 patients, including over 550 in a coronary CTA substudy, should provide key insights into the efficacy and safety of olezarsen in patients with largely moderate hypertriglyceridemia and elevated cardiovascular risk. TRIAL REGISTRATION Clinicaltrials.gov: NCT05610280.
Collapse
Affiliation(s)
- Brian A Bergmark
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA.
| | - Nicholas A Marston
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | | | | | - Andre Zimerman
- Clinical Trials Unit, Hospital Moinhos de Vento, Moinhos de Vento College of Health Sciences, Porto Alegre, Brazil
| | - Filipe A Moura
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT; VA Connecticut Healthcare System, West Haven, CT
| | - Yu Mi Kang
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA; Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Sabina A Murphy
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Shuanglu Zhang
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Michael T Lu
- Harvard Medical School, Boston, MA; Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA
| | | | - Sotirios Tsimikas
- Ionis Pharmaceuticals, Carlsbad, CA; Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA
| | - Robert P Giugliano
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Marc S Sabatine
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| |
Collapse
|
2
|
Adamopoulou E, Dimitriadis K, Kyriakoulis K, Pyrpyris N, Beneki E, Fragkoulis C, Konstantinidis D, Aznaouridis K, Tsioufis K. Defining "Vulnerable" in coronary artery disease: predisposing factors and preventive measures. Cardiovasc Pathol 2025; 77:107736. [PMID: 40228760 DOI: 10.1016/j.carpath.2025.107736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
The likelihood of a plaque to cause an acute coronary syndrome (ACS) depends on several factors, both lesion- and patient-related. One of the most investigated and established contributing factors is the presence of high-risk or "vulnerable plaque" characteristics, which have been correlated with increased incidence of major adverse cardiovascular events (MACE). The recognition, however, that a significant percentage of vulnerable plaques do not result in causing clinical events has led the scientific community towards the more multifaceted concept of "vulnerable patients". Incorporating the morphological features of an atherosclerotic plaque into its hemodynamic surroundings can better predict the chance of its disruption, as altered fluid dynamics play a significant role in plaque destabilization. The advances in coronary imaging and the field of computational fluid dynamics (CFD) can contribute to develop more accurate lesion- and patient-related ACS prediction models that take into account both the morphology of a plaque and the forces applied upon it. The aim of this review is to provide the latest data regarding the aforementioned predictive factors as well as relevant preventive measures.
Collapse
Affiliation(s)
- Eleni Adamopoulou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece.
| | - Konstantinos Kyriakoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Nikolaos Pyrpyris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Eirini Beneki
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Christos Fragkoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Dimitris Konstantinidis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Konstantinos Aznaouridis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| |
Collapse
|
3
|
Yao W, Chen H, Huang K, Peng W, Zhang X, Yang D, Teng Z, Shen J, Yang J, Cheng X, Han Y, Zhu W, Wang J, Du J, Liu X. Atherosclerotic plaque evolution predicts cerebral ischemic events in patients with intracranial atherosclerosis: a multicentre longitudinal study using high-resolution MRI. Eur Radiol 2025; 35:3238-3248. [PMID: 39702635 PMCID: PMC12081491 DOI: 10.1007/s00330-024-11248-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/20/2024] [Accepted: 10/28/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Intracranial atherosclerosis (ICAS) is the leading cause of ischemic stroke in Asians and the recurrent rate remains high despite the optimal medical treatment. This study aimed to confirm that follow-up high-resolution magnetic resonance imaging (hrMRI) provided essential values in predicting subsequent cerebral ischemic events in patients with ICAS. METHODS Patients with moderate to severe stenosis in the middle cerebral artery (MCA) defined by magnetic resonance (MRA) or computed tomography angiography (CTA) were recruited from three centers retrospectively. Detailed plaque composition was analyzed on baseline and follow-up hrMRI. Multivariate Cox proportional hazards regression analysis was used to identify the key risk factors for predicting subsequent ischemic events. RESULTS Among 152 patients, a total of 86 patients with MCA atherosclerotic stenosis underwent follow-up hrMRI exams and ipsilateral cerebral ischemic events occurred in 12 patients during a 1-year follow-up. Analyses showed the predictors of ischemic events were age (adjusted Hazard ratio (HR) = 0.942; 95% Confidence Interval (CI), [0.903, 0.983]; p = 0.006), progression of plaque burden (HR = 3.818; 95% CI [1.117, 13.051]; p = 0.033), vessel expansion (HR = 5.173; 95% CI [1.077, 24.838]; p = 0.040) and enhancement ratio progression (HR = 6.144; 95% CI [1.480, 25.511]; p = 0.012). The combined model achieved a concordance index of 0.804 (95% CI [0.658, 0.950]). CONCLUSION Longitudinal hrMRI evaluation improved the accuracy in identifying higher-risk patients with intracranial atherosclerosis. KEY POINTS Question Can longitude high-resolution magnetic resonance imaging (hrMRI) help clinicians observe intracranial plaque evolution? Findings Compared with the baseline exam, intracranial plaque evolution distinguished by follow-up hrMRI exam showed a higher accuracy in predicting subsequent ischemic events. Clinical relevance Longitudinal high-resolution magnetic resonance vessel wall imaging enables dynamic observation and evaluation of the changes in plaque characteristics among intracranial atherosclerosis patients. Atherosclerotic plaque evolution revealed by repeated exams can strengthen the risk stratification of patients with intracranial atherosclerosis.
Collapse
Affiliation(s)
- Weihe Yao
- Department of Neurology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hongbing Chen
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kangmo Huang
- Department of Neurology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenjia Peng
- Department of Radiology, Changhai Hospital, Shanghai, China
| | - Xuefeng Zhang
- Department of Radiology, Changhai Hospital, Shanghai, China
| | - Dahong Yang
- Department of Neurology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhongzhao Teng
- Department of Radiology, University of Cambridge, Cambridge, UK
- Nanjing Jingsan Medical Science and Technology Ltd., Jiangsu, China
| | - Jinhua Shen
- Nanjing Jingsan Medical Science and Technology Ltd., Jiangsu, China
| | - Jialuo Yang
- Department of Medical Imaging, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaoqing Cheng
- Department of Medical Imaging, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yunfei Han
- Department of Neurology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wusheng Zhu
- Department of Neurology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Junjun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Clinical Laboratory, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Juan Du
- Department of Neurology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
4
|
Poredoš P, Mangaroska AS, Poredoš P. Atherosclerotic plaque stabilization and regression. VASA 2025. [PMID: 40356553 DOI: 10.1024/0301-1526/a001202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Atherosclerotic plaques represent a typical deterioration of arterial wall in atherosclerotic process and are the source of cardiovascular events. Plaque progression and composition represent a major risk for cardiovascular events. Therefore, recently many studies have assessed changes in plaque characteristics and their response to various treatment modalities. In the last two decades, improvement in plaque imaging modalities that can assess plaque volumes and composition enable to follow plaque characteristics in a non-invasive way. Clinical trials utilizing arterial imaging modalities have shown that reducing LDL cholesterol to low levels can reduce atherosclerotic plaque burden and favourably modify plaque composition. These outcomes have been achieved with statin therapy and newer lipid-lowering strategies such as protein convertase subtilisin/kexin type 9 inhibitors. Also, some anti-inflammatory drugs and other anti-atherosclerotic medications can lead to significant reduction in plaque burden. However, the data assessing association of plaque regression to reduction of cardiovascular events are limited. Therefore, the aim of this narrative review is to elucidate the possibilities and the role of plaque assessment and if it might offer the potential to guide personalized management of patients at risk for cardiovascular events in the future.
Collapse
Affiliation(s)
- Pavel Poredoš
- Department of Vascular Disease, University Medical Centre Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Slovenia
| | | | - Peter Poredoš
- Faculty of Medicine, University of Ljubljana, Slovenia
- Department of Anaesthesiology and Surgical Intensive Care, University Medical Centre Ljubljana, Slovenia
| |
Collapse
|
5
|
Zhu J, Zhu X, Lv S, Guo D, Li H, Zhao Z. Incremental Value of Pericoronary Adipose Tissue Radiomics Models in Identifying Vulnerable Plaques. J Comput Assist Tomogr 2025; 49:422-430. [PMID: 39724572 DOI: 10.1097/rct.0000000000001704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Inflammatory characteristics in pericoronary adipose tissue (PCAT) may enhance the diagnostic capability of radiomics techniques for identifying vulnerable plaques. This study aimed to evaluate the incremental value of PCAT radiomics scores in identifying vulnerable plaques defined by intravascular ultrasound imaging (IVUS). METHODS In this retrospective study, a PCAT radiomics model was established and validated using IVUS as the reference standard. The dataset consisted of patients with coronary artery disease who underwent both coronary computed tomography angiography and IVUS examinations at a tertiary hospital between March 2023 and January 2024. The dataset was randomly assigned to the training and validation sets in a 7:3 ratio. The diagnostic performance of various models was evaluated on both sets using the area under the curve (AUC). RESULTS From 88 lesions in 79 patients, we selected 9 radiomics features (5 texture features, 1 shape feature, 1 gray matrix feature, and 2 first-order features) from the training cohort (n = 61) to build the PCAT model. The PCAT radiomics model demonstrated moderate to high AUCs (0.847 and 0.819) in both the training and test cohorts. Furthermore, the AUC of the PCAT radiomics model was significantly higher than that of the fat attenuation index model (0.847 vs 0.659, P < 0.05). The combined model had a higher AUC than the clinical model (0.925 vs 0.714, P < 0.01). CONCLUSIONS The PCAT radiomics signature of coronary CT angiography enabled the detection of vulnerable plaques defined by IVUS.
Collapse
Affiliation(s)
- Jinke Zhu
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, Shaoxing, Zhejiang, China
| | - Xiucong Zhu
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, Shaoxing, Zhejiang, China
| | - Sangying Lv
- Department of radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| | - Danling Guo
- Department of radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| | - Huaifeng Li
- Department of radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| | - Zhenhua Zhao
- Department of radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| |
Collapse
|
6
|
Zhang YS, Shi R, Jiang YN, Gao Y, Jiang Y, Wang J, Li WR, Li JK, Yang ZG, Li Y. The association between the triglyceride-glucose index and vulnerable plaques in patients with type 2 diabetes mellitus: insights from coronary computed tomography angiography. Cardiovasc Diabetol 2025; 24:169. [PMID: 40241069 PMCID: PMC12004695 DOI: 10.1186/s12933-025-02673-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/05/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND The triglyceride‒glucose index (TyG index) has been verified to be a useful predictor of insulin resistance (IR), and is associated with the occurrence of acute coronary syndrome (ACS). However, the effect of the TyG index on vulnerable plaques (VP), which were identified when at least two high-risk features are present within the same lesion, in type 2 diabetes mellitus (T2DM) patients is not fully understood. This study aimed to explore the association between the TyG index and the presence of VP. METHODS We retrospectively enrolled 2056 T2DM patients who underwent coronary computed tomography angiography (CCTA) examinations at West China Hospital from February 2017 to February 2022. These patients were divided into four groups on the basis of the quartiles of the TyG index. The high-risk coronary plaque features, vulnerable plaques, plaque type, coronary artery stenosis, segment involvement score (SIS), segment stenosis score (SSS) and multivessel disease (MVD) based on CCTA data were evaluated and compared among the four groups. RESULTS Patients with a higher TyG index had more noncalcified and mixed plaques, high-risk plaque features, vulnerable plaques and fewer calcified plaques (P < 0.05 for all). The proportion of patients with high-risk plaque features, including low-attenuation noncalcified plaques, positive remodeling and "napkin ring" sign was associated with the TyG index (P for trend < 0.05 for all). Multivariate analysis revealed that the TyG index was significantly associated with vulnerable plaques in T2DM patients [OR = 1.23 (95% CI 1.00-1.51), P = 0.046]. Subgroup analysis revealed that the association between the TyG index and vulnerable plaques varied with age and the prevalence of cardiovascular (CVD) symptoms, even after controlling for confounding factors (P for interaction < 0.05 for both). CONCLUSION The TyG index was independently associated with vulnerable plaques of the coronary artery among patients with T2DM. The TyG index could be regarded as a marker to reduce the incidence of cardiovascular events in the targeted population of T2DM patients.
Collapse
Affiliation(s)
- Yu-Shan Zhang
- Department of Radiology, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China
| | - Rui Shi
- Department of Radiology, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China
| | - Yi-Ning Jiang
- Department of Radiology, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China
| | - Yue Gao
- Department of Radiology, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China
| | - Yu Jiang
- Department of Radiology, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China
| | - Jin Wang
- Department of Radiology, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China
| | - Wen-Rong Li
- Department of Radiology, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China
| | - Jia-Ke Li
- Department of Radiology, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China
| | - Zhi-Gang Yang
- Department of Radiology, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China.
| | - Yuan Li
- Department of Radiology, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
7
|
Ji H, Dong Z, Yang Y, Cui W, Han J, Hu Y, Chen H, Qiao C, Li Q, Li H, Wu S. Neixiao-ruanmai decoction No 2 improves carotid atherosclerosis by modulating gut microbiota and inhibiting TLR4/NF-κB pathway activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156775. [PMID: 40286751 DOI: 10.1016/j.phymed.2025.156775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Carotid atherosclerosis(CAs) plaques are challenging to reverse. Neixiao-Ruanmai Decoction No 2(NXRMT No 2), a Traditional Chinese Medicine (TCM) decoction, has shown potential in treating CAs. However, while preliminary clinical trials have confirmed the efficacy of NXRMT No 2 in improving CAs, the comparative effectiveness of long-term versus short-term treatment courses remains unclear, and the underlying mechanisms of this decoction are not yet fully understood. METHODS We conducted clinical trials, animal studies, 16S rRNA sequencing, metabolomics and fecal microbiota transplantation. RESULT Clinical research results indicate that NXRMT No 2(24 weeks of treatment) reduced total plaque area by 22.02%, maximum plaque thickness by 7.91%, and maximum plaque area by 21.29%. NXRMT No 2 improves patients'serum inflammatory levels, with a 24-week treatment course demonstrated superior efficacy compared to the 12-week treatment. Animal experiments demonstrated that NXRMT No 2 improved CAs progression, modulated the gut microbiota, inhibited the intestinal Toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) pathway and activated the expression of intestinal tight junction proteins. CONCLUSION NXRMT No 2 significantly attenuates CAs progression, with its primary mechanism likely related to modulating the gut microbiota to counteract the TLR4/NF-κB pathway and protect the intestinal barrier. This study provides evidence-based support for the use of NXRMT No 2 in treating CAs, offers guidance on optimal treatment duration for patients, and contributes to the development of traditional Chinese medicine formulations that improve CAs by modulating the gut microbiota-a significant advance in the prevention and treatment of CAs.
Collapse
Affiliation(s)
- Hanrui Ji
- Beijing Huairou Hospital of Traditional Chinese Medicine, Beijing101400, PR China
| | - Zhizhi Dong
- Beijing Huairou Hospital of Traditional Chinese Medicine, Beijing101400, PR China
| | - Yanan Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Wenqiang Cui
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250014 Jinan, PR China
| | - Jingbo Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100010, PR China
| | - Yibin Hu
- Neurology Department of The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 250355, PR China
| | - Haonan Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100010, PR China
| | - Chongxuan Qiao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100010, PR China
| | - Qingxiao Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100010, PR China
| | - He Li
- Neurology Department of The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 250355, PR China.
| | - Shengxian Wu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100010, PR China.
| |
Collapse
|
8
|
Campbell DJ, Francis VCM, Young GR, Woodford NWF. Investigation of Myocardial Substrate for Sudden Arrhythmic Death in Coronary Artery Disease Without Acute Coronary Thrombosis or Myocardial Infarction. J Am Heart Assoc 2025; 14:e039624. [PMID: 40194965 DOI: 10.1161/jaha.124.039624] [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: 10/22/2024] [Accepted: 02/12/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND This cohort study aimed to evaluate the potential myocardial arrhythmic substrate in people with coronary artery disease who died from sudden arrhythmic death (SAD) without acute coronary thrombosis or myocardial infarction. METHODS AND RESULTS We performed histological analysis of the left ventricular free wall obtained at autopsy from decedents with ≥1 coronary artery and ≥75% area stenosis who died suddenly from either noncardiac causes (25 men, 23 women) or SAD (25 men, 25 women), matched for age and sex. Decedents with acute coronary thrombosis, myocardial infarction, or other myocardial abnormality were excluded. Decedents with either noncardiac death or SAD had similar height, weight, and heart weight. Decedents with SAD had higher cumulative area stenosis of coronary arteries (mean, 162% versus 134%; mean difference, 29% [95% CI, 1%-56%], P=0.042) and a higher proportion of decedents with SAD had diabetes (mean, 10% versus 0%; mean difference, 10% [95% CI, 2%-18%], P=0.025) and chronic, nonocclusive, organized coronary artery thrombus (mean, 16% versus 0%; mean difference, 16% [95% CI, 6%-26%], P=0.0040). Moreover, decedents with SAD had lower cardiomyocyte width (mean, 18.6 μm versus 19.6 μm; mean difference, 1.0 μm [95% CI, 0.2-1.8], P=0.014) and higher capillary length density (mean, 3618 mm/mm3 versus 3164 mm/mm3; mean difference, 453 mm/mm3 [95% CI, 210-697], P=0.0003) than decedents with noncardiac death. CONCLUSIONS SAD in people with coronary artery disease without acute coronary thrombosis or myocardial infarction was associated with greater coronary artery plaque burden and cardiomyocyte atrophy that may have contributed to myocardial substrate for arrhythmia.
Collapse
Affiliation(s)
- Duncan J Campbell
- St. Vincent's Institute of Medical Research Fitzroy Victoria Australia
- University of Melbourne Parkville Victoria Australia
- St. Vincent's Hospital Melbourne Victoria Australia
| | - Victoria C M Francis
- Department of Forensic Medicine, School of Public Health and Preventive Medicine Monash University Southbank Victoria Australia
- Victorian Institute of Forensic Medicine Southbank Victoria Australia
| | - Gregory R Young
- Department of Forensic Medicine, School of Public Health and Preventive Medicine Monash University Southbank Victoria Australia
- Victorian Institute of Forensic Medicine Southbank Victoria Australia
| | - Noel W F Woodford
- Department of Forensic Medicine, School of Public Health and Preventive Medicine Monash University Southbank Victoria Australia
- Victorian Institute of Forensic Medicine Southbank Victoria Australia
| |
Collapse
|
9
|
Boucher DM, Robichaud S, Lorant V, Leon JS, Suliman I, Rasheed A, Susser LI, Emerton C, Geoffrion M, De Jong E, Bowdish DM, Aikawa M, Aikawa E, Singh SA, Rayner KJ, Ouimet M. Age-Related Impairments in Immune Cell Efferocytosis and Autophagy Hinder Atherosclerosis Regression. Arterioscler Thromb Vasc Biol 2025; 45:481-495. [PMID: 39945065 PMCID: PMC11936474 DOI: 10.1161/atvbaha.124.321662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Aging is a well-established risk factor for the development and progression of atherosclerosis, but the molecular mechanisms underlying this relationship remain poorly defined, and its role in atherosclerosis regression is unknown. To uncover age-related alterations that may impair atherosclerosis regression, we investigated the response of young and old macrophages to atherogenic lipoproteins in vitro and in vivo. METHODS Metabolic and proteomic studies were performed in vitro using macrophages differentiated from the bone marrow of young or old mice. To test the role of immune cell aging in atherosclerosis regression, bone marrow from young and old donors was transplanted into irradiated young recipient mice expressing gain-of-function AAV-PCSK9 (adeno-associated virus-proprotein convertase subtilisin/kexin type 9). Following 14 weeks of Western diet feeding, atherosclerosis regression was induced by switching to a standard laboratory diet for 4 weeks. RESULTS Compared with young macrophages, old macrophages accumulated more lipid droplets upon lipid loading with the pro-atherogenic lipoprotein aggregated LDL (low-density lipoprotein), accompanied by a failure to proportionally induce autophagy and cholesterol efflux. Proteomic analysis of bone marrow-derived macrophages revealed that pathways related to endocytosis, engulfment, and phagocytosis were downregulated in old lipid-loaded macrophages. Functional studies confirmed a reduction in efferocytic capacity in old macrophages. In recipient mice transplanted with old bone marrow, atherosclerosis regression was impaired, as evidenced by inefficient resolution of circulating inflammatory cell levels, reduced activation of plaque autophagy and apoptotic cell clearance, and persistent plaque CD45+ and CD68+ content. CONCLUSIONS Aging impairs macrophage function through reduced efferocytosis and autophagy activation, limiting atherosclerosis regression. These results highlight the need to better define the mechanisms linking aging to atherosclerosis to develop targeted therapies for the aging population.
Collapse
Affiliation(s)
- Dominique M. Boucher
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Sabrina Robichaud
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Victoria Lorant
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Jonathan S. Leon
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Issraa Suliman
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Adil Rasheed
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Leah I. Susser
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Christina Emerton
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Michele Geoffrion
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Erica De Jong
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada (E.D.J., D.M.E.B.)
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, ON, Canada (E.D.J., D.M.E.B.)
| | - Dawn M.E. Bowdish
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada (E.D.J., D.M.E.B.)
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, ON, Canada (E.D.J., D.M.E.B.)
| | - Masanori Aikawa
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Center for Excellence in Vascular Biology (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Center for Excellence in Vascular Biology (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sasha A. Singh
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Center for Excellence in Vascular Biology (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Katey J. Rayner
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Mireille Ouimet
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| |
Collapse
|
10
|
Williams KJ. Inflammation in atherosclerosis: a Big Idea that has underperformed so far. Curr Opin Lipidol 2025; 36:78-87. [PMID: 39846349 PMCID: PMC11888836 DOI: 10.1097/mol.0000000000000973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
PURPOSE OF REVIEW For many years, inflammation has been a major concept in basic research on atherosclerosis and in the development of potential diagnostic tools and treatments. The purpose of this review is to assess the performance of this concept with an emphasis on recent clinical trials. In addition, contemporary literature may help identify new therapeutic targets, particularly in the context of the treatment of early, rather than end-stage, arterial disease. RECENT FINDINGS Newly reported clinical trials cast doubt on the efficacy of colchicine, the sole anti-inflammatory agent currently approved for use in patients with atherosclerotic cardiovascular disease (ASCVD). New analyses also challenge the hypothesis that residual ASCVD event risk after optimal management of lipids, blood pressure, and smoking arises primarily from residual inflammatory risk. Current clinical practice to initiate interventions so late in the course of atherosclerotic arterial disease may be a better explanation. Lipid-lowering therapy in early atherosclerosis, possibly combined with novel add-on agents to specifically accelerate resolution of maladaptive inflammation, may be more fruitful than the conventional approach of testing immunosuppressive strategies in end-stage arterial disease. Also discussed is the ongoing revolution in noninvasive technologies to image the arterial wall. These technologies are changing screening, diagnosis, and treatment of atherosclerosis, including early and possibly reversable disease. SUMMARY The burden of proof that the Big Idea of inflammation in atherosclerosis has clinical value remains the responsibility of its advocates. This responsibility requires convincing trial data but still seems largely unmet. Unfortunately, the focus on inflammation as the source of residual ASCVD event risk has distracted us from the need to screen and treat earlier.
Collapse
Affiliation(s)
- Kevin Jon Williams
- Department of Cardiovascular Sciences and Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Occhipinti G, Brugaletta S, Abbate A, Pedicino D, Del Buono MG, Vinci R, Biondi Zoccai G, Sabate M, Angiolillo D, Liuzzo G. Inflammation in coronary atherosclerosis: diagnosis and treatment. Heart 2025:heartjnl-2024-325408. [PMID: 40139681 DOI: 10.1136/heartjnl-2024-325408] [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: 11/14/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025] Open
Abstract
Coronary atherosclerosis is a chronic condition characterised by the development of an atherosclerotic plaque in the inner layer of the coronary artery, mainly associated with cholesterol accumulation and favoured by endothelial dysfunction related to other cardiovascular risk factors, such as smoking, diabetes and hypertension. A key actor in this process is the systemic inflammatory response, which can make plaques either grow slowly over the course of years (like a 'mountain'), obstructing coronary flow, and causing stable coronary artery disease, or make them explode (like a 'volcano') with subsequent abrupt thrombosis causing an acute coronary syndrome. This central role of inflammation in coronary atherosclerosis has led to its consideration as a modifiable cardiovascular risk factor and a therapeutic target. Classic anti-inflammatory drugs have been tested in clinical trials with some encouraging results, and new drugs specifically designed to tackle inflammation are currently being under investigation in ongoing trials. The objectives of this review are to (1) summarise the role of inflammatory biomarkers and imaging techniques to detect inflammation at each stage of plaque progression, and (2) explore currently available and upcoming anti-inflammatory therapies.
Collapse
Affiliation(s)
- Giovanni Occhipinti
- Cardiovascular Clinic Institute, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Hospital Clínic de Barcelona, Barcelona, Catalunya, Spain
| | - Salvatore Brugaletta
- Cardiovascular Clinic Institute, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Hospital Clínic de Barcelona, Barcelona, Catalunya, Spain
- Universitat de Barcelona Facultat de Medicina i Ciències de la Salut, Barcelona, Catalunya, Spain
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Daniela Pedicino
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ramona Vinci
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
| | - Giuseppe Biondi Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome La Sapienza, Latina, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Manel Sabate
- Cardiovascular Clinic Institute, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Hospital Clínic de Barcelona, Barcelona, Catalunya, Spain
- Universitat de Barcelona Facultat de Medicina i Ciències de la Salut, Barcelona, Catalunya, Spain
| | - Dominick Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Giovanna Liuzzo
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
| |
Collapse
|
12
|
Yu H, Wei D, Liao W, Shang X, Li D, Liu C, Deng Q, Huangfu H. Exosome-mediated effects of BRCA1 on cardiovascular artery disease. Cell Biol Toxicol 2025; 41:59. [PMID: 40080209 PMCID: PMC11906578 DOI: 10.1007/s10565-025-09996-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 01/28/2025] [Indexed: 03/15/2025]
Abstract
The progression of coronary artery disease atherosclerosis (CAD) is closely associated with cardiomyocyte apoptosis and inflammatory responses. This study focused on investigating the impact of BRCA1 in exosomes (Exo) derived from M1 macrophages on CAD. Through the analysis of single-cell RNA-seq datasets, significant communication between macrophages and cardiomyocytes in CAD patients was observed. BRCA1, identified as a significant apoptosis-related gene, was pinpointed through the assessment of differential gene expression and weighted gene co-expression network analysis (WGCNA). Experimental procedures involved BRCA1 lentivirus transfection of M1 macrophages, isolation of Exo for application to cardiomyocytes and smooth muscle cells, cell viability assessments, and characterization of Exo. The results showed that BRCA1-Exo from M1 macrophages induced cardiomyocyte apoptosis and affected smooth muscle cell behavior. In vivo studies further supported the exacerbating effects of BRCA1-Exo on CAD progression. Overall, the involvement of Exo carrying BRCA1 from M1 macrophages is evident in the induction of cardiomyocyte apoptosis and the regulation of smooth muscle cell behaviors, thereby contributing to CAD atherosclerosis progression. These findings unveil novel molecular targets that could have potential implications for CAD treatment strategies.
Collapse
Affiliation(s)
- Hairui Yu
- Department of Preventive Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Dong Wei
- Department of Preventive Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Weiqian Liao
- Department of Cardiology, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Xiaoming Shang
- Department of Cardiology, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Dandan Li
- Department of Cardiology, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Chunzhao Liu
- Department of Preventive Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Qimei Deng
- Department of Preventive Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China
| | - Haiquan Huangfu
- Department of Cardiology, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518000, China.
| |
Collapse
|
13
|
Denicolai M, Morello M, Golino M, Corna G, Del Buono MG, Agatiello CR, Van Tassell BW, Abbate A. Interleukin-1 Blockade in Patients With ST-Segment Elevation Myocardial Infarction Across the Spectrum of Coronary Artery Disease Complexity. J Cardiovasc Pharmacol 2025; 85:200-210. [PMID: 39531530 DOI: 10.1097/fjc.0000000000001652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
ABSTRACT Patients with ST-segment elevation myocardial infarction (STEMI) and complex coronary artery disease (CAD) face a poor prognosis, including increased heart failure (HF) risk. Phase 2 clinical trials of anakinra have shown inhibition of the acute inflammatory response and prevention of HF after STEMI, but data on its effects based on CAD complexity are lacking. We performed a pooled secondary analysis of 139 patients with STEMI. The SYNTAX (Synergy Between Percutaneous Coronary Intervention with Taxus and Cardiac Surgery), SYNTAX II, and Gensini scores were calculated, and patients were divided into 2 groups below and above the median. We evaluated the effect of anakinra on the area-under-the-curve of high-sensitivity C-reactive protein (hsCRP-AUC) at 14 days, and the composite endpoint of new-onset HF, HF hospitalization, or all-cause death at 1-year follow-up using Kaplan-Meier survival curves, Cox regression analysis for hazard ratios (HRs), and tested interactions between subgroups. All 3 CAD complexity scores (SYNTAX, SYNTAX II, and Gensini) were associated with an increased risk of adverse events (HR 1.02-1.06, all P-values ≤0.025). We found no statistically significant interactions between CAD extent, measured as single-vessel or multivessel CAD, SYNTAX score ≤9 or >9, SYNTAX II score ≤24 or >24, Gensini score ≤32 or >32, and treatment effect of anakinra on hsCRP-AUC or the composite clinical endpoint (all P - values for interaction >0.05). In conclusion, among patients with STEMI, IL-1 blockade with anakinra significantly attenuated the acute inflammatory response and reduced the risk of HF-related events regardless of the spectrum of CAD complexity.
Collapse
Affiliation(s)
- Martin Denicolai
- Robert M. Berne Cardiovascular Research Center and Department of Medicine, University of Virginia, Charlottesville, VA
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Matteo Morello
- Robert M. Berne Cardiovascular Research Center and Department of Medicine, University of Virginia, Charlottesville, VA
| | - Michele Golino
- Robert M. Berne Cardiovascular Research Center and Department of Medicine, University of Virginia, Charlottesville, VA
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| | - Giuliana Corna
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Marco G Del Buono
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy ; and
| | - Carla R Agatiello
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Benjamin W Van Tassell
- Department of Pharmacotherapy & Outcomes Sciences, Virginia Commonwealth University, Richmond, VA
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Department of Medicine, University of Virginia, Charlottesville, VA
| |
Collapse
|
14
|
Casso-Chapa B, González NAV, Le NT, Palaskas NL, Nead KT, Eutsey LP, Samanthapudi VSK, Osborn AM, Lee J, Mejia G, Hoang O, Lin SH, Deswal A, Herrmann J, Wang G, Kirkland JL, Krishnan S, Wehrens XH, Chini EN, Yusuf SW, Iliescu CA, Jain A, Burks JK, Seeley E, Lorenzi PL, Chau KM, Mendoza KCO, Grumbach IM, Brookes PS, Hanssen NM, de Winther MP, Yvan-Charvet L, Kotla S, Schadler K, Abe JI. Reevaluating Anti-Inflammatory Therapy: Targeting Senescence to Balance Anti-Cancer Efficacy and Vascular Disease. Arterioscler Thromb Vasc Biol 2025; 45:372-385. [PMID: 39817327 PMCID: PMC11864897 DOI: 10.1161/atvbaha.124.319870] [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: 06/18/2024] [Revised: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 01/18/2025]
Abstract
Modulating immune function is a critical strategy in cancer and atherosclerosis treatments. For cancer, boosting or maintaining the immune system is crucial to prevent tumor growth. However, in vascular disease, mitigating immune responses can decrease inflammation and slow atherosclerosis progression. Anti-inflammatory therapy, therefore, presents a unique dilemma for cancer survivors: while it may decrease cardiovascular risk, it might also promote cancer growth and metastasis by suppressing the immune response. Senescence presents a potentially targetable solution to this challenge; senescence increases the risk of both cancer therapy resistance and vascular disease. Exercise, notably, shows promise in delaying this premature senescence, potentially improving cancer outcomes and lowering vascular disease risk post-treatment. This review focuses on the long-term impact of cancer therapies on vascular health. We underscore the importance of modulating senescence to balance cancer treatment's effectiveness and its vascular impact, and we emphasize investigating the role of exercise-mediated suppression of senescence in improving cancer survivorship.
Collapse
Affiliation(s)
- Bernardo Casso-Chapa
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | - Norma Alicia Vazquez González
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kevin T. Nead
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lydia P. Eutsey
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Cancer Center Support Grant & Extramural Research Development, UT MD Anderson Cancer Center, Houston, TX
| | | | - Abigail M Osborn
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonghae Lee
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gilbert Mejia
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Oanh Hoang
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - James L. Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xander H.T. Wehrens
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cezar A. Iliescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Jared K. Burks
- Department of Leukemia, Division of Center Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Erin Seeley
- Department of Chemistry, University of Texas at Austin, Austin, Texas, USA
| | - Philip L. Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Khanh M. Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Keila Carolina Ostos Mendoza
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | | | - Paul S. Brookes
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, USA
| | - Nordin M.J. Hanssen
- Department of (Experimental) Vascular and Internal Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- Diabeter Centrum Amsterdam, Amsterdam, the Netherlands
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam Institute for Immunology and Infectious Diseases (AII), Inflammatory Diseases Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Keri Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
15
|
Fan CH, Chen LF, Cheng J, Wang YQ, Xu LH, Li JM. Predicting plaque regression based on plaque characteristics identified by optical coherence tomography: A retrospective study. Photodiagnosis Photodyn Ther 2025; 51:104473. [PMID: 39761809 DOI: 10.1016/j.pdpdt.2025.104473] [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: 10/16/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Atherosclerosis is a lipid-driven, systemic immune-inflammatory disease characterized by the accumulation of plaque within the arterial walls. Plaque regression can occur following appropriate treatment interventions. Optical coherence tomography (OCT), a high-resolution imaging modality, is frequently employed to assess plaque morphology. This study aims to explore the correlation between plaque characteristics identified using OCT, particularly macrophage infiltration, and subsequent plaque regression. METHODS In this retrospective study, data from 112 individuals with coronary artery plaques, who underwent OCT imaging at our hospital, between June 2019 and June 2024, were evaluated. Plaques were classified as lipid-rich, fibrous, or calcified based on the initial OCT findings. Macrophage infiltration levels within each plaque type were quantified. After one year of follow-up, repeat OCT imaging was performed to evaluate plaque regression. Statistical analyses were conducted to assess the relationship between initial plaque characteristics and regression outcomes. RESULTS Plaques that underwent regression were more commonly lipid-rich and exhibited higher levels of macrophage infiltration compared to those without regression. Multivariate analysis identified the histological inflammation score (HIS) as an independent factor influencing plaque regression. CONCLUSION Macrophage-rich plaques, as detected by OCT, are significant predictors of plaque regression. The identification of vulnerable plaque features through OCT can enhance the early diagnosis and treatment strategies for atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Cheng-Hui Fan
- Department of Cardiology, Shanghai East Hospital, Nanjing Medical University, Nanjing 211166, China; Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Lyu-Fan Chen
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jing Cheng
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yi-Qiong Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Ling-Hao Xu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ji-Ming Li
- Department of Cardiology, Shanghai East Hospital, Nanjing Medical University, Nanjing 211166, China; Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
16
|
Chu J, Yuan D, Lai Y, Ye W, Liu L, Lin H, Ping F, Zhu G, Chen F, Yao Y, Yan W, Liu X. Prognostic Implications of Changes in Total Physiological Atherosclerotic Burden in Patients With Coronary Artery Disease-A Serial QFR Study. Angiology 2025; 76:174-182. [PMID: 37994827 DOI: 10.1177/00033197231218616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
The association between coronary physiological progression and clinical outcomes has not been investigated. A total of 421 patients who underwent serial coronary angiography at least 6 months apart were included. Total physiological atherosclerotic burden was characterized by sum of quantitative flow ratio in 3 epicardial vessels (3V-QFR). The relationships of the 3V-QFR and its longitudinal change (△3V-QFR) with major adverse cardiovascular events (MACE) were explored. 3V-QFR values derived from follow-up angiograms were slightly lower compared with baseline (2.85 [2.77, 2.90] vs 2.86 [2.80, 2.90], P < .001). The median △3V-QFR value was -0.01 (-0.05, 0.02). The multivariable models demonstrated that follow-up 3V-QFR and △3V-QFR were independently associated with MACE (both P < .05). Patients with both low follow-up 3V-QFR (≤2.78) and low △3V-QFR (≤-0.05) presented 3 times higher risk of MACE than those without (hazard ratio: 2.953, 95% confidence interval 1.428-6.104, P = .003). Furthermore, adding patient-level 3V-QFR and △3V-QFR to clinical model significantly improved the predictability for MACE. In conclusion, total physiological atherosclerotic burden and its progression can provide incremental prognostic value over clinical characteristics, supporting the use of coronary physiology in the evaluation of disease progression and for the identification of vulnerable patients.
Collapse
Affiliation(s)
- Jiapeng Chu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Deqiang Yuan
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yan Lai
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wen Ye
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lei Liu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Lin
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fan Ping
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guoqi Zhu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fei Chen
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yian Yao
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenwen Yan
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuebo Liu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
17
|
Rives J, Gil-Millan P, Viladés D, García-Osuna Á, Genua I, Miñambres I, Grau-Agramunt M, Gich I, Puig N, Benitez S, Julve J, Pérez A, Sánchez-Quesada JL. Low-Density Lipoprotein Subfraction Phenotype Is Associated with Epicardial Adipose Tissue Volume in Type 2 Diabetes. J Clin Med 2025; 14:862. [PMID: 39941533 PMCID: PMC11818426 DOI: 10.3390/jcm14030862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/14/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Increased epicardial adipose tissue (EAT) volume is a common feature in type 2 diabetes (T2DM) which is directly associated with heart failure and advanced atherosclerosis. We aimed to evaluate lipoprotein-related biomarkers of EAT volume in T2DM patients before and after glycemic control. Methods: This study included 36 T2DM patients before and after optimization of glycemic control and on 14 healthy controls (HCs). EAT volume was measured using computed tomography imaging indexed to the body surface area (iEAT). Biochemical and lipid profiles were determined using commercial methods. Lipoproteins were isolated by ultracentrifugation, and variables of lipoprotein function were assessed. Multivariable regression analysis was used to find variables independently associated with iEAT. Results: iEAT was higher in T2DM than in controls and decreased with glycemic optimization. HDLs from T2DM had less apoA-I and cholesterol and more apoC-III and triglycerides. LDLs from T2DM had more triglycerides and apoB and smaller sizes than those from HCs. Significant correlations were found between iEAT and age, BMI, HbA1c, GGT, VLDLc, triglycerides, LDL size, apoA-I in HDL, and apoC-III in HDL. In the multivariable regression analysis, age, LDL size, and GGT associations remained statistically significant, and predicted 50% of the variability in EAT volume. ROC analysis using these variables showed an AUC of 0.835. Conclusions: Qualitative characteristics of lipoproteins were altered in T2DM. Multivariable analysis showed that LDL size and GGT plasma levels were independently associated with iEAT volume, suggesting that these variables might be useful biomarkers for stratifying T2DM patients with increased EAT volume.
Collapse
Affiliation(s)
- José Rives
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Pedro Gil-Millan
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (P.G.-M.); (I.G.); (I.M.); (J.J.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - David Viladés
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- CIBER of Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Álvaro García-Osuna
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
| | - Idoia Genua
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (P.G.-M.); (I.G.); (I.M.); (J.J.)
| | - Inka Miñambres
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (P.G.-M.); (I.G.); (I.M.); (J.J.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Margarida Grau-Agramunt
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
| | - Ignasi Gich
- Epidemiology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Nuria Puig
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
| | - Sonia Benitez
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Josep Julve
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (P.G.-M.); (I.G.); (I.M.); (J.J.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Antonio Pérez
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (P.G.-M.); (I.G.); (I.M.); (J.J.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - José Luis Sánchez-Quesada
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), 08041 Barcelona, Spain; (J.R.); (Á.G.-O.); (M.G.-A.); (N.P.); (S.B.)
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| |
Collapse
|
18
|
Gordon‐Kundu B, Peyravi R, Garg A, Baker A, Salas S, Levien M, Faridi KF, de Havenon A, Krumholz HM, Sheth KN, Forman R, Sharma R. Lipid-Lowering Therapy Use When Indicated and Subsequent Ischemic Stroke Severity. J Am Heart Assoc 2025; 14:e033365. [PMID: 39699007 PMCID: PMC12054410 DOI: 10.1161/jaha.123.033365] [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: 12/09/2023] [Accepted: 09/09/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Elevated low-density lipoprotein cholesterol is a risk factor for atherosclerotic cardiovascular disease, including acute ischemic stroke (AIS), due to large- and small-vessel disease. Cholesterol management guidelines recommend lipid-lowering therapy (LLT) to prevent atherosclerotic cardiovascular disease events. This study assessed use of LLT at the time of AIS according to guideline recommendations and determined the association of prestroke LLT use with stroke severity. METHODS AND RESULTS We conducted a retrospective study of patients hospitalized with AIS from 2015 to 2020 at a large academic comprehensive stroke center. Patients with AIS secondary to either small-vessel disease or large-artery atherosclerosis recorded in the institutional Get With The Guidelines-Stroke registry and with a prestroke indication for LLT were included. Using propensity score subclassification, adjusted logistic regression models were built to explore the associations between LLT use before AIS when indicated and presenting National Institutes of Health Stroke Scale score >4. There were 384 patients with AIS who met guideline-recommended criteria for prestroke LLT (median age 70 years, 57% men), of whom only 207 patients (54%) were prescribed LLT before AIS. Not being prescribed LLT when indicated was associated with a significantly higher likelihood of a presenting with National Institutes of Health Stroke Scale score >4, even when adjusted for specific stroke cause (odds ratio, 1.13 [95% CI, 1.03-1.20]; P=0.006). CONCLUSIONS LLT is underused in patients who present with atherosclerosis-related AIS. Lack of prestroke LLT use was associated with more severe stroke symptoms upon presentation. These findings emphasize the need to prescribe LLT when indicated, because its use may mitigate poststroke disability.
Collapse
Affiliation(s)
- Barbara Gordon‐Kundu
- Department of Neurology, Division of Cerebrovascular DiseasesHackensack Meridian School of MedicineNutleyNJUSA
| | - Reza Peyravi
- Department of Neurology, Center for Brain and Mind Health, Division of Vascular NeurologyYale School of MedicineNew HavenCTUSA
| | - Anisha Garg
- Department of Neurology, Division of Neuromuscular DiseasesMount Sinai School of MedicineNew YorkNYUSA
| | - Anna Baker
- Department of Neurology, Center for Brain and Mind Health, Division of Vascular NeurologyYale School of MedicineNew HavenCTUSA
| | - Samantha Salas
- Department of Neurology, Center for Brain and Mind Health, Division of Vascular NeurologyYale School of MedicineNew HavenCTUSA
| | - Michael Levien
- Department of Neurology, Center for Brain and Mind Health, Division of Vascular NeurologyYale School of MedicineNew HavenCTUSA
| | - Kamil F. Faridi
- Department of Medicine, Section of Cardiovascular MedicineYale School of MedicineNew HavenCTUSA
| | - Adam de Havenon
- Department of Neurology, Center for Brain and Mind Health, Division of Vascular NeurologyYale School of MedicineNew HavenCTUSA
| | - Harlan M. Krumholz
- Department of Medicine, Section of Cardiovascular MedicineYale School of MedicineNew HavenCTUSA
| | - Kevin N. Sheth
- Department of Neurology, Center for Brain and Mind Health, Division of Vascular NeurologyYale School of MedicineNew HavenCTUSA
| | - Rachel Forman
- Department of Neurology, Center for Brain and Mind Health, Division of Vascular NeurologyYale School of MedicineNew HavenCTUSA
| | - Richa Sharma
- Department of Neurology, Center for Brain and Mind Health, Division of Vascular NeurologyYale School of MedicineNew HavenCTUSA
| |
Collapse
|
19
|
Li Y, Han Z, Zhao X, Liu Y, Wu Z, Wang J, Li X, Guo X, Tao L. Association between joint exposure to ambient air pollutants and carotid plaque: The mediating role of cardiometabolic risk factors. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117755. [PMID: 39854868 DOI: 10.1016/j.ecoenv.2025.117755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Research has shown that exposure to joint air pollution is related to atherosclerosis, but little evidence has been found for carotid plaques. Our objective is to assess the association between exposure to joint air pollutants and carotid plaque and explore the mediating role of cardiometabolic factors in this relationship. METHODS The Beijing Health Management Cohort (BMHC) study followed participants recruited from 2013 to 2014 until December 31, 2020. All participants underwent carotid ultrasound and were free of carotid plaque at baseline. A satellite-based land-use regression (LUR) model was applied to estimate air pollution exposure. The joint exposure to air pollutants was assessed by incorporating a weighted air pollution score. A modified Poisson regression model was conducted to investigate the relationship between exposure to air pollution and carotid plaque occurrence. Mediation analysis explored how cardiometabolic factors mediate the relationships between exposure to joint air pollution and carotid plaque risk. RESULTS During an average follow-up period 4 years, 1240 cases of carotid plaque were identified among 7358 participants. Each interquartile range (IQR) increase in air pollutants was associated with the following relative risk (RR) and 95 % confidence intervals (95 % CIs) for carotid plaque: 2.5-micrometer particulate matter (PM2.5), 1,04 (1.01, 1.07), 10-micrometer particulate matter (PM10), 1.10 (1.01, 1.20), sulfur dioxide (SO2), 1.28 (1.15, 1.42), ozone (O3), 1.18 (1.01, 1.37), and carbon monoxide (CO), 1.32 (1.15, 1.50). Joint exposure to air pollution was positively and linearly associated with the occurrence of carotid plaque, with low-density cholesterol (LDL-C) and mean arterial pressure (MAP) mediating 2.24 % and 4.28 % of the association, respectively. CONCLUSIONS Long-term joint exposure to ambient air pollutants elevates the risk of developing carotid plaque. LDL-C and MAP suggest partial mediating effects of joint air pollution on carotid plaques. Our results emphasize the need to thoroughly evaluate various air pollutants concerning carotid plaque.
Collapse
Affiliation(s)
- Yunfei Li
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.
| | - Ze Han
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.
| | - Xiaoyu Zhao
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.
| | - Yueruijing Liu
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.
| | - Zhiyuan Wu
- Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA.
| | - Jinqi Wang
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.
| | - Xia Li
- Department of Mathematics and Statistics, La Trobe University, Melbourne 3086, Australia.
| | - Xiuhua Guo
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia.
| | - Lixin Tao
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Ding Q, He Y, Zhao J, Liu W, Zhu Z, Pang Y, Zhao Y, Liu Y, Wang ZL, Zhu L, He Y, Li T. Impact of drug-coated balloon for vascular luminal dilatational remodeling after balloon angioplasty in intracranial atherosclerotic stenosis - a retrospective cohort study. J Neurointerv Surg 2025:jnis-2024-022281. [PMID: 39778933 DOI: 10.1136/jnis-2024-022281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/17/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVE This study aimed to examine the effect of drug-coated balloons (DCBs) on vascular luminal dilatational remodeling (VLDR) following simple balloon angioplasty. METHODS A retrospective cohort study was conducted using data from patients diagnosed with intracranial atherosclerotic stenosis (ICAS), who were treated exclusively with balloon angioplasty at Henan Provincial People's Hospital between June 2019 and April 2023. Inverse probability weighting (IPW) was used to create balanced cohorts of patients who underwent drug-coated balloon angioplasty (DCBA) and plain old balloon angioplasty (POBA). The primary endpoint was VLDR occurrence during follow-up, with the effect of DCBA on VLDR assessed by adjusted multivariate regression. RESULTS The study included 110 patients who underwent simple percutaneous transluminal angioplasty, with 60 in the DCBA group and 50 in the POBA group. At follow-up, the stenosis rate in the DCBA group was lower than in the POBA group (P<0.001). The decrease in stenosis rate (DSR) was greater in the DCBA group compared with the POBA group (P<0.001). Nineteen patients (31.7%) in the DCBA group experienced VLDR, whereas only four (8%) in the POBA group developed VLDR, a statistically significant difference (P=0.002). After IPW adjustment, differences in stenosis rate (34.17 (20.00, 46.72) vs 46.00 (37.88, 70.00), P<0.001), DSR (-1.66 (-16.71, 11.40) vs -18.00 (-28.00, -3.00), P<0.001) and VLDR incidence (32.2% vs 9.9%, P<0.001) between the DCBA and POBA groups remained significant. Multivariate regression analysis identified DCBA as an independent factor influencing VLDR occurrence. CONCLUSION This study demonstrated that, compared with POBA, DCBA increases VLDR occurrence in ICAS patients during follow-up.
Collapse
Affiliation(s)
- Qianhao Ding
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital; Henan Provincial Cerebrovascular Interventional Innovation Engineering Technology Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Zhengzhou, Henan, China
| | - Yingkun He
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital; Henan Provincial Cerebrovascular Interventional Innovation Engineering Technology Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Zhengzhou, Henan, China
| | - Jingge Zhao
- Department of Scientific Research and Foreign Affairs, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Wenbo Liu
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital; Henan Provincial Cerebrovascular Interventional Innovation Engineering Technology Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Zhengzhou, Henan, China
| | - Zhengpeng Zhu
- Department of Cerebrovascular Disease, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yukuan Pang
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital; Henan Provincial Cerebrovascular Interventional Innovation Engineering Technology Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Zhengzhou, Henan, China
| | - Yang Zhao
- School of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yang Liu
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital; Henan Provincial Cerebrovascular Interventional Innovation Engineering Technology Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Zhengzhou, Henan, China
| | - Zi-Liang Wang
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital; Henan Provincial Cerebrovascular Interventional Innovation Engineering Technology Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Zhengzhou, Henan, China
| | - Liangfu Zhu
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital; Henan Provincial Cerebrovascular Interventional Innovation Engineering Technology Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Zhengzhou, Henan, China
| | - Yanyan He
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital; Henan Provincial Cerebrovascular Interventional Innovation Engineering Technology Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Zhengzhou, Henan, China
| | - Tianxiao Li
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital; Henan Provincial Cerebrovascular Interventional Innovation Engineering Technology Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Zhengzhou, Henan, China
- Sanya Hospital of Traditional Chinese Medicine, Sanya, Hainan, China
| |
Collapse
|
21
|
Wang M, Zhou S, Hu Y, Tong W, Zhou H, Ma M, Cai X, Zhang Z, Zhang L, Chen Y. Macrophages overexpressing interleukin-10 target and prevent atherosclerosis: Regression of plaque formation and reduction in necrotic core. Bioeng Transl Med 2025; 10:e10717. [PMID: 39801756 PMCID: PMC11711221 DOI: 10.1002/btm2.10717] [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: 02/01/2024] [Revised: 07/27/2024] [Accepted: 08/08/2024] [Indexed: 01/16/2025] Open
Abstract
Atherosclerosis, a slowly progressing inflammatory disease, is characterized by the presence of monocyte-derived macrophages. Interventions targeting the inflammatory characteristics of atherosclerosis hold promising potential. Although interleukin (IL)-10 is widely acknowledged for its anti-inflammatory effects, systemic administration of IL-10 has limitations due to its short half-life and significant systemic side effects. In this study, we aimed to investigate the effectiveness of an approach designed to overexpress IL-10 in macrophages and subsequently introduce these genetically modified cells into ApoE-/- mice to promote atherosclerosis regression. We engineered RAW264.7 cells to overexpress IL-10 (referred to as IL-10M) using lentivirus vectors. The IL-10M exhibited robust IL-10 secretion, maintained phagocytic function, improved mitochondrial membrane potentials, reduced superoxide production and showed a tendency toward the M2 phenotype when exposed to inflammatory stimuli. IL-10M can selectively target plaques in ApoE-/- mice and has the potential to reduce plaque area and necrotic core at both early and late stages of plaque progression. Moreover, there was a significant reduction in MMP9, a biomarker associated with plaque rupture, in IL-10M-treated plaques from both the early and late intervention groups. Additionally, the administration of IL-10M showed no obvious side effects. This study serves as proof that cell therapy based on anti-inflammatory macrophages might be a promising strategy for the intervention of atherosclerosis.
Collapse
Affiliation(s)
- Mingyi Wang
- Medical School of Chinese PLABeijingChina
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
| | - Shanshan Zhou
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- Department of CardiologyThe First Medical Center of PLA General HospitalBeijingChina
| | - Yingyun Hu
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- The Medical School of Nankai UniversityTianjinChina
| | - Wei Tong
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- Department of CardiologyThe First Medical Center of PLA General HospitalBeijingChina
| | - Hao Zhou
- Department of CardiologyNo. 966 Hospital of Joint Logisties ForceDandongChina
| | - Mingrui Ma
- Medical School of Chinese PLABeijingChina
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
| | - Xingxuan Cai
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- The Second Medical School of Southern Medical UniversityGuangzhouChina
| | - Zhengbin Zhang
- Medical School of Chinese PLABeijingChina
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
| | - Luo Zhang
- Medical School of Chinese PLABeijingChina
- Research Center of BioengineeringThe Medical Innovation Research Division of PLA General HospitalBeijingChina
| | - Yundai Chen
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- Department of CardiologyThe First Medical Center of PLA General HospitalBeijingChina
| |
Collapse
|
22
|
Guajardo-Jauregui N, Cardenas-de la Garza JA, Galarza-Delgado DA, Azpiri-Lopez JR, Arvizu-Rivera RI, Polina-Lugo RL, Colunga-Pedraza IJ. Inadequate identification of high cardiovascular risk and carotid plaques in rheumatoid arthritis patients by the 2024 Predicting Risk of Cardiovascular EVENTs and the 2013 Atherosclerotic Cardiovascular Disease algorithms: findings from a Mexican cohort. Clin Rheumatol 2025; 44:161-169. [PMID: 39656397 DOI: 10.1007/s10067-024-07249-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 01/14/2025]
Abstract
The American College of Cardiology/American Heart Association introduced the Predicting Risk of Cardiovascular EVENTs (PREVENT™) algorithm to estimate the 10-year risk of developing cardiovascular disease. We aimed to assess the cardiovascular risk (CVR) reclassification among rheumatoid arthritis (RA) patients using traditional CVR algorithms-the 2024 PREVENT™ and the 2013 Atherosclerotic Cardiovascular Disease (ASCVD)-and the presence of carotid plaque (CP). This was a cross-sectional study nested of a RA patients' cohort. A certified radiologist performed a high-resolution B-mode carotid ultrasound to identify the presence of CP. The CVR evaluation was performed by a cardiologist, blinded to carotid ultrasound results, using the PREVENT™ and the ASCVD algorithms. Cohen's kappa (k) coefficient assessed concordance between high-risk classification by CVR algorithms and CP presence. ROC curve analysis evaluated the algorithms' capacity to identify RA patients with CP. The cutoff point was determined by the Youden-Index, with p < 0.05 as statistically significant. A total of 210 RA patients were included. The reclassification of CVR due to CP was 34.3% for the PREVENT™ algorithm and 30.0% for the ASCVD algorithm. Of these, 44.4% and 71.4%, respectively, were initially classified as low risk. Concordance between CVR algorithms and carotid ultrasound showed slight agreement (k = 0.032 and k = 0.130, respectively). The PREVENT™ algorithm did not identify more than one-third of high-CVR RA patients with indication of starting statin therapy based on carotid ultrasound findings. The PREVENT™ and ASCVD algorithms showed poor performance in identifying RA patients with CP. Key Points • The presence of CP was identified in more than a third of the evaluated RA patients (35.7%), classifying them as high CVR. • CVR reclassification by the presence of CP was observed in 34.3% RA patients with the PREVENTTM algorithm and in 30.0% RA patients with the ASCVD algorithm. • Most of the reclassified patients belonged to the low-risk category, 44.4% with the PREVENTTM algorithm and 71.4% with the ASCVD algorithm. • When evaluating the concordance between the ASCVD algorithm and the carotid ultrasound for high-risk classification, a slight agreement was found (k = 0.130).
Collapse
Affiliation(s)
- Natalia Guajardo-Jauregui
- Internal Medicine Department, Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Jesus Alberto Cardenas-de la Garza
- Rheumatology Service, Internal Medicine Department, Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Dionicio Angel Galarza-Delgado
- Rheumatology Service, Internal Medicine Department, Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Jose Ramon Azpiri-Lopez
- Cardiology Service, Internal Medicine Department, Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Rosa Icela Arvizu-Rivera
- Rheumatology Service, Internal Medicine Department, Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Rebeca Lizeth Polina-Lugo
- Rheumatology Service, Internal Medicine Department, Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Iris Jazmin Colunga-Pedraza
- Rheumatology Service, Internal Medicine Department, Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon, Monterrey, Mexico.
| |
Collapse
|
23
|
Bruun K, Mortensen MB. Rethinking atherosclerotic cardiovascular disease prevention in the era of expanding therapies: could plaque stabilization reduce the need for lifelong treatments and polypharmacy? Curr Opin Cardiol 2025; 40:50-55. [PMID: 39436382 DOI: 10.1097/hco.0000000000001188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
PURPOSE OF REVIEW This review examines current evidence on pharmacologically induced plaque stabilization in the context of a growing range of new therapies. It explores the potential for a paradigm shift in atherosclerotic cardiovascular disease (ASCVD) prevention, where treatments may not need to be lifelong to achieve lasting benefits. RECENT FINDINGS Since 2015, over 14 novel therapies have been introduced, each shown to reduce ASCVD risk when added to standard care with statins and aspirin. More than 80% of ischemic heart disease patients are now eligible for one or more of these treatments, increasing the risk of polypharmacy, treatment burden, and adverse side effects. As more therapies become available, this challenge is expected to grow. Many of these treatments have demonstrated plaque regression and stabilization, as evidenced by both intravascular ultrasound and computed tomography angiography, which likely explains much of their efficacy. SUMMARY The increasing number of novel therapies presents challenges in preventing ASCVD without leading to lifelong polypharmacy and increased patient burden. Since many of these drugs act through plaque stabilization, a new approach may be feasible - using these treatments for shorter durations to induce plaque regression, followed by less intensive maintenance therapies to preserve stability. This approach warrants further investigation in future studies.
Collapse
Affiliation(s)
- Kathrine Bruun
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Martin Bødtker Mortensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Ehrlich A, Pelli G, Foglia B, Molica F, Kwak BR. Protective role of Pannexin1 in lymphatic endothelial cells in the progression of atherosclerosis in female mice. PLoS One 2024; 19:e0315511. [PMID: 39775604 PMCID: PMC11684638 DOI: 10.1371/journal.pone.0315511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Atherosclerosis is a progressive arterial disease arising from imbalanced lipid metabolism and a maladaptive immune response. The lymphatic system ensures tissue fluid homeostasis, absorption of dietary fats and trafficking of immune cells to draining lymph nodes, thereby potentially affecting atherogenesis. Endothelial cell-specific deletion of Pannexin1 (Panx1) in apolipoprotein E-deficient (Apoe-/-) mice increased atherosclerosis, suggesting a protective role for Panx1 channels in arterial endothelial function. Here, we investigated the role of Panx1 in lymphatic endothelial cells (LECs) in the initiation and the progression of atherosclerosis. Male or female Prox1-CreERT2+Panx1fl/flApoe-/- and Panx1fl/flApoe-/- mice were fed a high cholesterol diet (HCD) for 6 or 10 weeks. Tamoxifen-induced deletion of Panx1 was performed before or after 4 weeks of HCD. Body weight and serum lipid profiles were determined. The atherosclerotic plaque burden was assessed by Sudan-IV staining on thoracic-abdominal aortas and in aortic roots. Plaque composition was determined by immunohistochemistry. No differences in serum cholesterol, LDL and HDL were observed between genotypes and between sexes after HCD. Bodyweight, serum triglycerides and free fatty acid levels were higher before and after 6 weeks of HCD in male Prox1-CreERT2+Panx1fl/flApoe-/- and control Panx1fl/flApoe-/- mice compared to females of the same genotypes, which was associated with more lipids and inflammatory cells in their atherosclerotic plaques. In contrast, the atherosclerotic plaque burden was higher in female mice. The progression of atherosclerosis in male mice was not different between genotypes. However, female Prox1-CreERT2+Panx1fl/flApoe-/- mice showed enhanced progression of atherosclerosis compared to Panx1fl/flApoe-/- controls of the same sex. In addition, atherosclerotic lesions in female, but not in male, Prox1-CreERT2+Panx1fl/flApoe-/- mice showed T cell enrichment. Altogether, our results reveal differential sex-dependent effects of Panx1 in lymphatic endothelium on the progression of atherosclerosis.
Collapse
Affiliation(s)
- Avigail Ehrlich
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Graziano Pelli
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Bernard Foglia
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Filippo Molica
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Brenda R. Kwak
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
25
|
Gibson CM, Duffy D, Bahit MC, Chi G, White H, Korjian S, Alexander JH, Lincoff AM, Heise M, Kingwell BA, Nicolau JC, Lopes RD, Cornel JH, Lewis BS, Vinereanu D, Goodman SG, Bode C, Steg PG, Libby P, Sacks FM, Bainey KR, Ridker PM, Mahaffey KW, Aylward P, Nicholls SJ, Pocock SJ, Mehran R, Harrington RA. Apolipoprotein A-I infusions and cardiovascular outcomes in acute myocardial infarction according to baseline LDL-cholesterol levels: the AEGIS-II trial. Eur Heart J 2024; 45:5023-5038. [PMID: 39221651 DOI: 10.1093/eurheartj/ehae614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/08/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS In the AEGIS-II trial (NCT03473223), CSL112, a human apolipoprotein A1 derived from plasma that increases cholesterol efflux capacity, did not significantly reduce the risk of the primary endpoint through 90 days vs. placebo after acute myocardial infarction (MI). Nevertheless, given the well-established relationship between higher low-density lipoprotein cholesterol (LDL-C) and plaque burden, as well as greater risk reductions seen with PCSK9 inhibitors in patients with baseline LDL-C ≥ 100 mg/dL on statin therapy, the efficacy of CSL112 may be influenced by baseline LDL-C. METHODS Overall, 18 219 patients with acute MI, multivessel coronary artery disease, and additional risk factors were randomized to either four weekly infusions of 6 g CSL112 or placebo. This exploratory post-hoc analysis evaluated cardiovascular outcomes by baseline LDL-C in patients prescribed guideline-directed statin therapy at the time of randomization (n = 15 731). RESULTS As baseline LDL-C increased, the risk of the primary endpoint at 90 days lowered in those treated with CSL112 compared with placebo. In patients with LDL-C ≥ 100 mg/dL at randomization, there was a significant risk reduction of cardiovascular death, MI, or stroke in the CSL112 vs. placebo group at 90, 180, and 365 days [hazard ratio .69 (.53-.90), .71 (.57-.88), and .78 (.65-.93)]. In contrast, there was no difference between treatment groups among those with LDL-C < 100 mg/dL at baseline. CONCLUSIONS In this population, treatment with CSL112 compared to placebo was associated with a significantly lower risk of recurrent cardiovascular events among patients with a baseline LDL-C ≥ 100 mg/dL. Further studies need to confirm that CSL112 efficacy is influenced by baseline LDL-C.
Collapse
Affiliation(s)
- C Michael Gibson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 930 Commonwealth Avenue, Boston, MA 02215, USA
| | - Danielle Duffy
- Clinical Development, CSL Behring, King of Prussia, PA, USA
| | | | - Gerald Chi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 930 Commonwealth Avenue, Boston, MA 02215, USA
| | - Harvey White
- Health New Zealand - Te Whatu Ora, Te Toka Tumai, Green Lane Cardiovascular Service, Auckland City Hospital, Auckland 1142, New Zealand
| | - Serge Korjian
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 930 Commonwealth Avenue, Boston, MA 02215, USA
| | - John H Alexander
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Duke Clinical Research Institute, Duke Health, Durham, NC, USA
| | - A Michael Lincoff
- The Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Mark Heise
- Clinical Development, CSL Behring, King of Prussia, PA, USA
- Deparment of Biostatistics, CSL Behring, King of Prussia, PA, USA
| | - Bronwyn A Kingwell
- Deparment of Research and Development, CSL Limited, Melbourne, Australia
| | - Jose C Nicolau
- Instituto do Coracao (InCor), Cardiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Renato D Lopes
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Duke Clinical Research Institute, Duke Health, Durham, NC, USA
- Brazilian Clinical Research Institute, Sao Paulo, SP, Brazil
| | - Jan H Cornel
- Radboud University Medical Center, Nijmegen and Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | | | - Dragos Vinereanu
- University of Medicine and Pharmacy Carol Davila, University and Emergency Hospital, Bucharest, Romania
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Canada
- Division of Cardiology, Department of Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
- Clinical Trials and Translation Unit, Peter Munk Cardiac Center, University Health Network, Toronto, Canada
| | - Christoph Bode
- Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ph Gabriel Steg
- Université Paris-Cité, INSERM U_1148, FACT and AP-HP, Hôpital Bichat, Paris, France
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Frank M Sacks
- Cardiovascular Disease Prevention, Department of Nutrition, Harvard T.H. Chan School of Public Health, Brigham and Women's Hospital, Boston, MA, USA
| | - Kevin R Bainey
- Walter Mackenzie Health Sciences Centre, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Boston, MA, USA
| | - Kenneth W Mahaffey
- Stanford Center for Clinical Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Philip Aylward
- Department of Cardiology, South Australian Health and Medical Research Institute/SAHMRI, Adelaide, Australia
| | - Stephen J Nicholls
- Department of Cardiology, Victorian Heart Institute, Monash University, Melbourne, VIC, Australia
| | - Stuart J Pocock
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Roxana Mehran
- Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, Zena and Michael A. Wiener Cardiovascular Institute, New York, NY, USA
| | | |
Collapse
|
26
|
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
|
27
|
Saenz-Pipaon G, Wacker BK, Bi L, Stamatikos A, Dichek DA. Exosome-Mediated Transfer of X-Motif-Tagged Anti-MiR-33a-5p Antagomirs to the Medial Cells of Transduced Rabbit Carotid Arteries. BIOLOGY 2024; 13:965. [PMID: 39765632 PMCID: PMC11673983 DOI: 10.3390/biology13120965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
Atherosclerosis is caused by the accumulation of cholesterol within intimal smooth muscle cells (SMCs) and macrophages. However, the transporter ATP-binding cassette subfamily A, member 1 (ABCA1), can remove cholesterol from these intimal, cells reducing atherosclerosis. Antagomir-mediated inhibition of miR-33a-5p, a microRNA that represses ABCA1 translation, promotes ABCA1-dependent cholesterol efflux and may impede atherosclerosis development. In our previous work, transducing cultured endothelial cells (ECs) with a helper-dependent adenoviral vector (HDAd) that expresses X-motif-tagged anti-miR-33a-5p enhanced antagomir packaging into EC-derived exosomes, which delivered the antagomir to cultured SMCs and macrophages. In this present study, we tested whether in vivo transduction of rabbit carotid artery endothelium can deliver an X-motif-tagged anti-miR-33a-5p to subendothelial cells. Rabbit carotid endothelial cells were transduced in vivo with an HDAd expressing anti-miR-33a-5p either with or without the X-motif (n = 11 arteries per vector). Contralateral carotids received HDAd that express scrambled oligonucleotides. Three days after transduction, the antagomir-without the X-motif-was detected in the intima but not in the media of transduced carotids (p = 0.062). The X-motif antagomir was detected in 82% of the intimal extracts (9 out of 11 carotids) and 27% of medial samples (3 out of 11 carotids, p = 0.031). However, the X-motif did not significantly enhance antagomir delivery to the media (p = 0.214 vs. non-X-motif antagomir). Expression of the antagomirs-with and without the X-motif-was sub-stoichiometric in ECs and SMCs. No antagomir-related changes in miR-33a-5p or ABCA1 expressions were detected. Despite its potential as a therapeutic strategy, our exosome-targeted gene transfer system requires further improvements to enhance antagomir expression and delivery to the subendothelial cells.
Collapse
Affiliation(s)
- Goren Saenz-Pipaon
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA 98195, USA; (B.K.W.); (L.B.); (D.A.D.)
| | - Bradley K. Wacker
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA 98195, USA; (B.K.W.); (L.B.); (D.A.D.)
| | - Lianxiang Bi
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA 98195, USA; (B.K.W.); (L.B.); (D.A.D.)
| | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA;
| | - David A. Dichek
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA 98195, USA; (B.K.W.); (L.B.); (D.A.D.)
| |
Collapse
|
28
|
Song J, Cao C, Wang Z, Li H, Yang L, Kang J, Meng H, Li L, Liu J. Mechanistic insights into the regression of atherosclerotic plaques. Front Physiol 2024; 15:1473709. [PMID: 39628943 PMCID: PMC11611857 DOI: 10.3389/fphys.2024.1473709] [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: 07/31/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
Atherosclerosis is a major contributor to cardiovascular diseases and mortality globally. The progression of atherosclerotic disease results in the expansion of plaques and the development of necrotic cores. Subsequent plaque rupture can lead to thrombosis, occluding blood vessels, and end-organ ischemia with consequential ischemic injury. Atherosclerotic plaques are formed by the accumulation of lipid particles overloaded in the subendothelial layer of blood vessels. Abnormally elevated blood lipid levels and impaired endothelial function are the initial factors leading to atherosclerosis. The atherosclerosis research has never been interrupted, and the previous view was that the pathogenesis of atherosclerosis is an irreversible and chronic process. However, recent studies have found that the progression of atherosclerosis can be halted when patients' blood lipid levels are reversed to normal or lower. A large number of studies indicates that it can inhibit the progression of atherosclerosis lesions and promote the regression of atherosclerotic plaques and necrotic cores by lowering blood lipid levels, improving the repair ability of vascular endothelial cells, promoting the reverse cholesterol transport in plaque foam cells and enhancing the ability of macrophages to phagocytize and clear the necrotic core of plaque. This article reviews the progress of research on the mechanism of atherosclerotic plaque regression. Our goal is to provide guidance for developing better therapeutic approaches to atherosclerosis by reviewing and analyzing the latest scientific findings.
Collapse
Affiliation(s)
- Jianshu Song
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- Research Institute of Traditional Chinese Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Ce Cao
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Ziyan Wang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Haoran Li
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- Research Institute of Traditional Chinese Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Lili Yang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Jing Kang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Hongxu Meng
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Lei Li
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Jianxun Liu
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
29
|
Hong D, Lee SH, Heo J, Shin D, Cho J, Guallar E, Joh HS, Kim HK, Ha J, Choi KH, Park TK, Yang JH, Song YB, Hahn JY, Choi SH, Gwon HC, Kang D, Lee JM. Safety and efficacy of antiplatelet therapy in patients with intermediate coronary artery stenosis and deferred revascularization. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024:S1885-5857(24)00331-1. [PMID: 39542207 DOI: 10.1016/j.rec.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION AND OBJECTIVES This study investigated the safety and efficacy of antiplatelet therapy in patients with intermediate coronary artery stenosis who underwent deferred revascularization due to their fractional flow reserve (FFR). METHODS A nationwide cohort study was conducted using the Korean National Health Insurance Service database. A total of 4657 patients with intermediate coronary artery stenosis who underwent deferred revascularization due to their FFR were identified from 2013 to 2020. FFR was indicated in patients with no prior evidence of myocardial ischemia and intermediate coronary artery stenosis (50%-70%) as determined by quantitative coronary angiography. Patients were classified according to whether antiplatelet therapy was initiated after the index procedure. The primary efficacy outcome was major adverse cardiac and cerebrovascular events (MACCE), a composite of all-cause death, myocardial infarction, unplanned revascularization, and stroke, during a 5-year follow-up period. The primary safety outcome was any gastrointestinal bleeding. RESULTS After propensity score matching, there were 1634 patients in the antiplatelet therapy group and 1634 in the nonantiplatelet therapy group. The risk of MACCE was similar between the 2 groups (24.8% vs 24.7%; adjusted HR, 0.97; 95%CI, 0.84-1.13; P=0.745). The risk of gastrointestinal bleeding was higher in the antiplatelet therapy group than in the nonantiplatelet therapy group (2.2% vs 1.2%; aHR, 2.07; 95%CI, 1.08-4.00). These results were similar in subgroup analyses. CONCLUSIONS In patients with intermediate coronary artery stenosis who underwent deferred revascularization due to their FFR, antiplatelet therapy may increase the risk of gastrointestinal bleeding without reducing the risk of future ischemic events.
Collapse
Affiliation(s)
- David Hong
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Hun Lee
- Division of Cardiology, Department of Internal Medicine, Heart Center, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, South Korea
| | - Jihye Heo
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea; Department of Clinical Research Design and Evaluation, Samsung Advances Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
| | - Doosup Shin
- Department of Cardiology, St Francis Hospital and Heart Center, Roslyn, New York, United States
| | - Juhee Cho
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea; Department of Clinical Research Design and Evaluation, Samsung Advances Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
| | - Eliseo Guallar
- Department of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States
| | - Hyun Sung Joh
- Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Kuk Kim
- Department of Internal Medicine and Cardiovascular Center, Chosun University Hospital, University of Chosun College of Medicine, Gwangju, Republic of Korea
| | - Junho Ha
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ki Hong Choi
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Taek Kyu Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeong Hoon Yang
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Bin Song
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Joo-Yong Hahn
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung-Hyuk Choi
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyeon-Cheol Gwon
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Danbee Kang
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea; Department of Clinical Research Design and Evaluation, Samsung Advances Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea.
| | - Joo Myung Lee
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Chen W, Liu Y, Deng X, Li B, Wang H, Wei G, Chen K, Wang S. CYP2C19 Loss-of-Function is an Associated Risk Factor for Premature Coronary Artery Disease: A Case-Control Study. Int J Gen Med 2024; 17:5049-5058. [PMID: 39512259 PMCID: PMC11542493 DOI: 10.2147/ijgm.s486187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/26/2024] [Indexed: 11/15/2024] Open
Abstract
Objective Cytochrome P450 2C19 (CYP2C19) is a major enzyme involved in the biotransformation and metabolism of various substances. Loss-of-function of the CYP2C19 gene represents downregulation of CYP2C19 enzyme indication limited or no enzymatic function, which may be, in turn, associated with some disease susceptibility. The relationship between CYP2C19 polymorphisms and susceptibility to premature coronary artery disease (PCAD) is not fully understood. This study aimed to assess this relationship. Methods This study included 635 PCAD patients, and 548 age-matched non-CAD individuals as controls, from November 2019 to August 2023. The CYP2C19 rs4244285 (681G > A, *2) and rs4986893 (636G > A, *3) were genotyped, and the distribution of CYP2C19 polymorphisms between patients and controls and the relationship between CYP2C19 polymorphisms and PCAD risk were analyzed. Results A total of 442 (37.4%), 543 (45.9%), and 198 (16.7%) individuals had CYP2C19 extensive metabolizer (EM) (*1/*1), intermediate metabolizer (IM) (*1/*2 and *1/*3), and poor metabolizer (PM) (*2/*2, *2/*3, and *3/*3) phenotypes, respectively. CYP2C19 *2/*2 genotype frequency was higher, *1/*1 genotype was lower in PCAD patients than controls. Individuals with CYP2C19 PM phenotype had higher triglyceride (TG) levels than those with CYP2C19 EM or IM phenotypes. Logistic regression analysis showed that body mass index (BMI) ≥24.0 kg/m2 (≥24.0 kg/m2 vs 18.5-23.9 kg/m2, odds ratio (OR): 1.326, 95% confidence interval (CI): 1.041-1.688, p = 0.022), smoking (OR: 1.974, 95% CI: 1.283-3.306, p = 0.002), hypertension (OR: 1.327, 95% CI: 1.044-1.687, p = 0.021), diabetes mellitus (OR: 1.390, 95% CI: 1.054-1.834, p = 0.020), CYP2C19 PM phenotype (PM phenotype vs EM phenotype, OR: 1.701, 95% CI: 1.200-2.411, p = 0.003), and CYP2C19 IM+PM phenotypes (IM+PM vs EM phenotype, OR: 1.369, 95% CI: 1.077-1.740, p = 0.010) were associated with PCAD. Conclusion CYP2C19 PM or IM+PM phenotypes, overweight, smoking, hypertension, and diabetes mellitus were associated with PCAD.
Collapse
Affiliation(s)
- Wenhao Chen
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Yuanliang Liu
- Department of Computer Tomography, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Xunwei Deng
- Research Experimental Center, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Bin Li
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Hao Wang
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Guoliang Wei
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Kehui Chen
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Shen Wang
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| |
Collapse
|
31
|
Ueki Y, Itagaki T, Kuwahara K. Lipid-lowering Therapy and Coronary Plaque Regression. J Atheroscler Thromb 2024; 31:1479-1495. [PMID: 39111840 PMCID: PMC11537793 DOI: 10.5551/jat.rv22024] [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: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 11/06/2024] Open
Abstract
Lipid-lowering therapy plays a central role in reducing cardiovascular events. Over the past few decades, clinical trials utilizing several imaging techniques have consistently shown that lipid-lowering therapy can reduce the coronary plaque burden and improve plaque composition. Although intravascular ultrasound has been the most extensively used modality to assess plaque burden, other invasive modalities, such as optical coherence tomography and near-infrared spectroscopy, provide relevant data on plaque vulnerability, and computed tomography angiography detects both plaque volume and characteristics non-invasively. A large body of evidence supports the notion that reducing low-density lipoprotein cholesterol using statins combined with ezetimibe and proprotein convertase subtillisin/kexin type 9 inhibitors consistently shows improvements in plaque burden and favorable morphological changes. This review summarizes previously obtained data on the impact of lipid-lowering treatment strategies on atherosclerotic plaque regression, as assessed using several imaging modalities.
Collapse
Affiliation(s)
- Yasushi Ueki
- Department of Cardiovascular Medicine, Shinshu University Hospital, Nagano, Japan
| | - Tadashi Itagaki
- Department of Cardiovascular Medicine, Shinshu University Hospital, Nagano, Japan
| | - Koichiro Kuwahara
- Department of Cardiovascular Medicine, Shinshu University Hospital, Nagano, Japan
| |
Collapse
|
32
|
Gać P, Jakubowska-Martyniuk A, Żórawik A, Hajdusianek W, Żytkowski D, Matys T, Poręba R. Diagnostic Methods of Atherosclerotic Plaque and the Assessment of Its Prognostic Significance-A Narrative Review. J Cardiovasc Dev Dis 2024; 11:343. [PMID: 39590186 PMCID: PMC11594366 DOI: 10.3390/jcdd11110343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Cardiovascular diseases (CVD) are a leading cause of death. The most notable cause of CVD is an atherosclerotic plaque. The aim of this review is to provide an overview of different diagnostic methods for atherosclerotic plaque relevant to the assessment of cardiovascular risk. The methods can be divided into invasive and non-invasive. This review focuses on non-invasive with attention paid to ultrasonography, contrast-enhanced ultrasonography, intravascular ultrasonography, and assessment of intima-media complex, coronary computed tomography angiography, and magnetic resonance. In the review, we discuss a number of Artificial Intelligence technologies that support plaque imaging.
Collapse
Affiliation(s)
- Paweł Gać
- Department of Environmental Health, Occupational Medicine and Epidemiology, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368 Wroclaw, Poland
- Centre of Diagnostic Imaging, 4th Military Hospital, Rudolfa Weigla 5, 50-981 Wrocław, Poland
| | - Anna Jakubowska-Martyniuk
- Department of Environmental Health, Occupational Medicine and Epidemiology, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368 Wroclaw, Poland
| | - Aleksandra Żórawik
- Department of Environmental Health, Occupational Medicine and Epidemiology, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368 Wroclaw, Poland
| | - Wojciech Hajdusianek
- Department of Environmental Health, Occupational Medicine and Epidemiology, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368 Wroclaw, Poland
| | - Dawid Żytkowski
- Department of Environmental Health, Occupational Medicine and Epidemiology, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368 Wroclaw, Poland
| | - Tomasz Matys
- Department of Angiology and Internal Diseases, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Rafał Poręba
- Centre of Diagnostic Imaging, 4th Military Hospital, Rudolfa Weigla 5, 50-981 Wrocław, Poland
| |
Collapse
|
33
|
Han W, Xiong N, Zhong R, Pan Z. CYP2C19 Poor Metabolizer Status and High System Inflammation Response Index are Independent Risk Factors for Premature Myocardial Infarction: A Hospital-Based Retrospective Study. Int J Gen Med 2024; 17:4959-4969. [PMID: 39494358 PMCID: PMC11529344 DOI: 10.2147/ijgm.s489235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Objective Atherosclerosis (AS) is a sustained chronic vascular inflammatory response caused by lipid metabolism disorders and immune response disorders and is the main cause of premature (men ≤ 55 years old, women ≤ 65 years old) myocardial infarction (PMI). Cytochrome P450 2C19 (CYP2C19) (related to vascular function and lipid metabolism) and peripheral immune cell levels and plays an important role in the course of AS. The association CYP2C19 polymorphisms, comprehensive immunoinflammatory indices with PMI susceptibility is unclear. Methods This study included 485 PMI patients, and 639 age-matched non-PMI individuals as controls, from January 2019 to March 2024. The relationship between CYP2C19 polymorphisms, peripheral immunoinflammatory indices (pan-immune inflammation value (PIV), systemic immune inflammation index (SII), and system inflammation response index (SIRI)) and PMI risk were analyzed. Results The inflammatory indices levels in PMI patients were higher than those in controls (all p<0.05). The frequencies of the CYP2C19 *1/*2 and *2/*2 genotypes were higher, while the frequency of the *1/*1 genotype was lower in the PMI patients than those in controls. The cut-off values of TC, TG, LDL-C, PIV, SII, and SIRI were 5.065, 1.305, 2.805, 410.485, 869.645, and 1.495 for distinguishing PMI, respectively. Logistic regression analysis showed that male (odds ratio (OR): 1.607, 95% confidence interval (CI): 1.134-2.277, p=0.008), history of smoking (OR: 7.108, 95% CI: 4.351-11.614, p<0.001), diabetes mellitus (OR: 4.906, 95% CI: 3.333-7.223, p<0.001), CYP2C19 poor metabolizer (PM) (*2/*2, *2/*3, and *3/*3) (OR: 2.147, 95% CI: 1.279-3.603, p=0.004), and high TG (≥1.305 vs <1.305, OR: 2.598, 95% CI: 1.864-3.623, p<0.001) and SIRI level (≥1.495 vs <1.495, OR: 2.495, 95% CI: 1.432-4.349, p=0.001) were independent risk factors for PMI. Conclusion CYP2C19 PM phenotype, high SIRI level (≥1.495) and TG level (≥1.305), male, history of smoking, and diabetes mellitus were independently associated with PMI susceptibility.
Collapse
Affiliation(s)
- Wendao Han
- Department of Blood Transfusion, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Nating Xiong
- Department of Blood Transfusion, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Renkai Zhong
- Department of Blood Transfusion, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Zhongyi Pan
- Department of Blood Transfusion, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| |
Collapse
|
34
|
Papafaklis MI, Koros R, Tsigkas G, Karanasos A, Moulias A, Davlouros P. Reversal of Atherosclerotic Plaque Growth and Vulnerability: Effects of Lipid-Modifying and Anti-Inflammatory Therapeutic Agents. Biomedicines 2024; 12:2435. [PMID: 39595002 PMCID: PMC11591594 DOI: 10.3390/biomedicines12112435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Atherosclerotic plaque development constitutes the primary substrate of coronary artery disease (CAD) and is the outcome of an intricate process involving endothelial damage, inflammation, and lipid retention. The clinical efficacy of many lipid-lowering therapies in patients with CAD has been well established. Over the past few decades, a substantial and significant advance regarding the use of invasive and non-invasive imaging modalities has been observed. Numerous studies have been conducted using these imaging techniques and have investigated the changes in morphology (e.g., atheroma volume) and composition (e.g., lipid burden, fibrous cap thickness, macrophage accumulation) at the plaque level that explain the improved clinical outcomes by various pharmacological interventions. Lipid-lowering agents, such as statins and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, demonstrate direct effects on plaque volume and composition that enhance plaque stabilization and/or regression beyond the reduction of low-density lipoproteins. An increasing amount of clinical research is also focused on the role of inflammation in plaque vulnerability and future adverse cardiac events. Consequently, there is a pressing need to explore therapeutic strategies that are capable of disrupting the inflammatory response as well as reducing atheroma burden and modifying high-risk plaque characteristics. This review provides a comprehensive analysis of the current evidence regarding the effects of traditional and novel therapeutic strategies targeting modification of the lipid profile and inflammatory processes on reversing plaque growth and attenuating vulnerable features, thereby promoting plaque stabilization and passivation.
Collapse
Affiliation(s)
- Michail I. Papafaklis
- Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Cardiology Division, University Hospital of Patras, 26504 Rio, Greece
| | - Rafail Koros
- Cardiology Division, University Hospital of Patras, 26504 Rio, Greece
| | - Grigorios Tsigkas
- Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Cardiology Division, University Hospital of Patras, 26504 Rio, Greece
| | - Antonios Karanasos
- Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Cardiology Division, University Hospital of Patras, 26504 Rio, Greece
| | | | - Periklis Davlouros
- Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Cardiology Division, University Hospital of Patras, 26504 Rio, Greece
| |
Collapse
|
35
|
Barbieri L, Tumminello G, Fichtner I, Corsini A, Santos RD, Carugo S, Ruscica M. PCSK9 and Coronary Artery Plaque-New Opportunity or Red Herring? Curr Atheroscler Rep 2024; 26:589-602. [PMID: 39150672 PMCID: PMC11393034 DOI: 10.1007/s11883-024-01230-6] [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: 07/30/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE OF REVIEW Although the clinical benefit of reducing low-density lipoprotein cholesterol (LDLc) in patients with coronary artery disease (CAD) is well-established, the impact on plaque composition and stability is less clear. Our narrative review aimed to assess the clinical effects of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors on coronary plaque characteristics specifically focusing from atheroma progression to regression and stabilization. RECENT FINDINGS The combination of statin therapy and PCSK9 inhibitors (evolocumab and alirocumab) promotes plaque stability in patients following an acute coronary syndrome. The GLAGOV study highlighted the relationship between achieved LDLc levels and changes in percentage atheroma volume. Similarly, the PACMAN-AMI study concluded that the qualitative and quantitative changes in coronary plaque were associated with the levels of LDLc. Assessing the severity of coronary artery stenosis and the extent of atherosclerotic burden by means of imaging techniques (e.g., IVUS, OCT and near-infrared spectroscopic) have significantly advanced our understanding of the benefits from promoting plaque regression and achieving to features of plaque stabilization through increasingly intensive lipid-lowering strategies.
Collapse
Affiliation(s)
- Lucia Barbieri
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriele Tumminello
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Raul D Santos
- Heart Institute (InCor), Lipid Clinic, University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
36
|
Yu M, Yang Y, Dong SL, Zhao C, Yang F, Yuan YF, Liao YH, He SL, Liu K, Wei F, Jia HB, Yu B, Cheng X. Effect of Colchicine on Coronary Plaque Stability in Acute Coronary Syndrome as Assessed by Optical Coherence Tomography: The COLOCT Randomized Clinical Trial. Circulation 2024; 150:981-993. [PMID: 39166327 DOI: 10.1161/circulationaha.124.069808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND Colchicine has been approved to reduce cardiovascular risk in patients with coronary heart disease on the basis of its potential benefits demonstrated in the COLCOT (Colchicine Cardiovascular Outcomes Trial) and LoDoCo2 (Low-Dose Colchicine 2) studies. Nevertheless, there are limited data available about the specific impact of colchicine on coronary plaques. METHODS This was a prospective, single-center, randomized, double-blind clinical trial. From May 3, 2021, until August 31, 2022, a total of 128 patients with acute coronary syndrome aged 18 to 80 years with lipid-rich plaque (lipid pool arc >90°) detected by optical coherence tomography were included. The subjects were randomly assigned in a 1:1 ratio to receive either colchicine (0.5 mg once daily) or placebo for 12 months. The primary end point was the change in the minimal fibrous cap thickness from baseline to the 12-month follow-up. RESULTS Among 128 patients, 52 in the colchicine group and 52 in the placebo group completed the study. The mean age of the 128 patients was 58.0±9.8 years, and 25.0% were female. Compared with placebo, colchicine therapy significantly increased the minimal fibrous cap thickness (51.9 [95% CI, 32.8 to 71.0] μm versus 87.2 [95% CI, 69.9 to 104.5] μm; difference, 34.2 [95% CI, 9.7 to 58.6] μm; P=0.006), and reduced average lipid arc (-25.2° [95% CI, -30.6° to -19.9°] versus -35.7° [95% CI, -40.5° to -30.8°]; difference, -10.5° [95% CI, -17.7° to -3.4°]; P=0.004), mean angular extension of macrophages (-8.9° [95% CI, -13.3° to -4.6°] versus -14.0° [95% CI, -18.0° to -10.0°]; difference, -6.0° [95% CI, -11.8° to -0.2°]; P=0.044), high-sensitivity C-reactive protein level (geometric mean ratio, 0.6 [95% CI, 0.4 to 1.0] versus 0.3 [95% CI, 0.2 to 0.5]; difference, 0.5 [95% CI, 0.3 to 1.0]; P=0.046), interleukin-6 level (geometric mean ratio, 0.8 [95% CI, 0.6 to 1.1] versus 0.5 [95% CI, 0.4 to 0.7]; difference, 0.6 [95% CI, 0.4 to 0.9]; P=0.025), and myeloperoxidase level (geometric mean ratio, 1.0 [95% CI, 0.8 to 1.2] versus 0.8 [95% CI, 0.7 to 0.9]; difference, 0.8 [95% CI, 0.6 to 1.0]; P=0.047). CONCLUSIONS Our findings suggested that colchicine resulted in favorable effects on coronary plaque stabilization at optical coherence tomography in patients with acute coronary syndrome. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT04848857.
Collapse
Affiliation(s)
- Miao Yu
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si-Lai Dong
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Zhao
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China (C.Z., H.-B.J., B.Y.)
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, China (C.Z., H.-B.J., B.Y.)
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin (C.Z., H.-B.J., B.Y.)
| | - Fen Yang
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Fan Yuan
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Hua Liao
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shao-Lin He
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Liu
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Wei
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hai-Bo Jia
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China (C.Z., H.-B.J., B.Y.)
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, China (C.Z., H.-B.J., B.Y.)
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin (C.Z., H.-B.J., B.Y.)
| | - Bo Yu
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China (C.Z., H.-B.J., B.Y.)
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, China (C.Z., H.-B.J., B.Y.)
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin (C.Z., H.-B.J., B.Y.)
| | - Xiang Cheng
- Department of Cardiology (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases (M.Y., Y.Y., S.-L.D., F.Y., Y.-F.Y., Y.-H.L., S.-L.H., K.L., F.W., X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Li H, Liu J, Liang Q, Yu Y, Sun G. Effect of Vascular Senescence on the Efficacy and Safety of Warfarin: Insights from Rat Models and a Prospective Cohort Study. J Pharmacol Exp Ther 2024; 391:39-50. [PMID: 39095206 DOI: 10.1124/jpet.124.002265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/03/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024] Open
Abstract
Warfarin, with its narrow therapeutic range, requires the understanding of various influencing factors for personalized medication. Vascular senescence, marked by vascular stiffening and endothelial dysfunction, has an unclear effect on the efficacy and safety of warfarin. Based on previous studies, we hypothesized that vascular senescence increases the risk of bleeding during warfarin therapy. This study aimed to explore these effects using animal models and clinical cohorts. We established rat models of vascular senescence and calcification using d-galactose, vitamin D, and nicotine. After validating the models, we examined changes in the international normalized ratio (INR) at fixed warfarin doses (0.20 and 0.35 mg/kg). We found that vascular senescence caused significantly elevated INR values and increased bleeding risk. In the prospective clinical cohort study (NCT06428110), hospitalized warfarin patients with standard dose adjustments were divided into vascular senescence and control groups based on ultrasound and computed tomography diagnosis. Using propensity score matching to exclude the influence of confounding factors, we found that the vascular senescence group had lower steady-state warfarin doses and larger dose adjustments, with a higher probability of INR exceeding the therapeutic range. The vascular senescence group tended to experience more bleeding or thromboembolic/ischemic events during 1 year of follow-up, while there was no statistical difference. In conclusion, vascular senescence leads to unstable INR values and increases higher bleeding risk during warfarin therapy, highlighting the importance of considering vascular senescence in future precision warfarin therapies. SIGNIFICANCE STATEMENT: Many factors influence warfarin efficacy; however, the effect of vascular senescence remains unclear. This study aimed to investigate the effects of vascular senescence on the efficacy and safety of warfarin. Through both rat models and clinical cohort studies, our findings indicated that vascular senescence may compromise the stability of warfarin, presenting challenges in maintaining its efficacy and safety.
Collapse
Affiliation(s)
- Haobin Li
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jing Liu
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Qing Liang
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Yan Yu
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Guangchun Sun
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
38
|
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
|
39
|
Barkas F, Sener YZ, Golforoush PA, Kheirkhah A, Rodriguez-Sanchez E, Novak J, Apellaniz-Ruiz M, Akyea RK, Bianconi V, Ceasovschih A, Chee YJ, Cherska M, Chora JR, D'Oria M, Demikhova N, Kocyigit Burunkaya D, Rimbert A, Macchi C, Rathod K, Roth L, Sukhorukov V, Stoica S, Scicali R, Storozhenko T, Uzokov J, Lupo MG, van der Vorst EPC, Porsch F. Advancements in risk stratification and management strategies in primary cardiovascular prevention. Atherosclerosis 2024; 395:117579. [PMID: 38824844 DOI: 10.1016/j.atherosclerosis.2024.117579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/29/2024] [Accepted: 05/14/2024] [Indexed: 06/04/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of morbidity and mortality worldwide, highlighting the urgent need for advancements in risk assessment and management strategies. Although significant progress has been made recently, identifying and managing apparently healthy individuals at a higher risk of developing atherosclerosis and those with subclinical atherosclerosis still poses significant challenges. Traditional risk assessment tools have limitations in accurately predicting future events and fail to encompass the complexity of the atherosclerosis trajectory. In this review, we describe novel approaches in biomarkers, genetics, advanced imaging techniques, and artificial intelligence that have emerged to address this gap. Moreover, polygenic risk scores and imaging modalities such as coronary artery calcium scoring, and coronary computed tomography angiography offer promising avenues for enhancing primary cardiovascular risk stratification and personalised intervention strategies. On the other hand, interventions aiming against atherosclerosis development or promoting plaque regression have gained attention in primary ASCVD prevention. Therefore, the potential role of drugs like statins, ezetimibe, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, omega-3 fatty acids, antihypertensive agents, as well as glucose-lowering and anti-inflammatory drugs are also discussed. Since findings regarding the efficacy of these interventions vary, further research is still required to elucidate their mechanisms of action, optimize treatment regimens, and determine their long-term effects on ASCVD outcomes. In conclusion, advancements in strategies addressing atherosclerosis prevention and plaque regression present promising avenues for enhancing primary ASCVD prevention through personalised approaches tailored to individual risk profiles. Nevertheless, ongoing research efforts are imperative to refine these strategies further and maximise their effectiveness in safeguarding cardiovascular health.
Collapse
Affiliation(s)
- Fotios Barkas
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece.
| | - Yusuf Ziya Sener
- Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | | | - Azin Kheirkhah
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elena Rodriguez-Sanchez
- Division of Cardiology, Department of Medicine, Department of Physiology, and Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Jan Novak
- 2(nd) Department of Internal Medicine, St. Anne's University Hospital in Brno and Faculty of Medicine of Masaryk University, Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Maria Apellaniz-Ruiz
- Genomics Medicine Unit, Navarra Institute for Health Research - IdiSNA, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Ralph Kwame Akyea
- Centre for Academic Primary Care, School of Medicine, University of Nottingham, United Kingdom
| | - Vanessa Bianconi
- Department of Medicine and Surgery, University of Perugia, Italy
| | - Alexandr Ceasovschih
- Internal Medicine Department, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Ying Jie Chee
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore
| | - Mariia Cherska
- Cardiology Department, Institute of Endocrinology and Metabolism, Kyiv, Ukraine
| | - Joana Rita Chora
- Unidade I&D, Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal; Universidade de Lisboa, Faculdade de Ciências, BioISI - Biosystems & Integrative Sciences Institute, Lisboa, Portugal
| | - Mario D'Oria
- Division of Vascular and Endovascular Surgery, Department of Medical Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Nadiia Demikhova
- Sumy State University, Sumy, Ukraine; Tallinn University of Technology, Tallinn, Estonia
| | | | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l'institut du Thorax, Nantes, France
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy
| | - Krishnaraj Rathod
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom; Barts Interventional Group, Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Vasily Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Svetlana Stoica
- "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania; Institute of Cardiovascular Diseases Timisoara, Timisoara, Romania
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Tatyana Storozhenko
- Cardiovascular Center Aalst, OLV Clinic, Aalst, Belgium; Department of Prevention and Treatment of Emergency Conditions, L.T. Malaya Therapy National Institute NAMSU, Kharkiv, Ukraine
| | - Jamol Uzokov
- Republican Specialized Scientific Practical Medical Center of Therapy and Medical Rehabilitation, Tashkent, Uzbekistan
| | | | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074, Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074, Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336, Munich, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074, Aachen, Germany
| | - Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
40
|
Sarraju A, Nissen SE. Atherosclerotic plaque stabilization and regression: a review of clinical evidence. Nat Rev Cardiol 2024; 21:487-497. [PMID: 38177454 DOI: 10.1038/s41569-023-00979-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 01/06/2024]
Abstract
Atherosclerotic plaque results from a complex interplay between lipid deposition, inflammatory changes, cell migration and arterial wall injury. Over the past two decades, clinical trials utilizing invasive arterial imaging modalities, such as intravascular ultrasonography, have shown that reducing levels of atherogenic lipoproteins, mainly serum LDL-cholesterol (LDL-C), to very low levels can safely reduce overall atherosclerotic plaque burden and favourably modify plaque composition. Classically, this outcome has been achieved with intensive statin therapy. Since 2016, newer and potent lipid-lowering strategies, such as proprotein convertase subtilisin-kexin type 9 inhibition, have shown incremental effects on plaque regression and risk of clinical events. Despite maximal reduction in plasma LDL-C levels, considerable residual cardiovascular risk remains in some patients. Therefore, there is a need to study therapeutic approaches that address residual risk beyond LDL-C reduction to promote plaque stabilization or regression. Contemporary imaging modalities, such as coronary computed tomography angiography, enable non-invasive assessment of the overall atherosclerotic plaque burden as well as of certain local plaque characteristics. This technology could allow further study of plaque stabilization and regression using novel therapeutic approaches. Non-invasive plaque assessment might also offer the potential to guide personalized management strategies if validated for this purpose.
Collapse
Affiliation(s)
- Ashish Sarraju
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Steven E Nissen
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
41
|
Pietruszyńska-Reszetarska A, Pietruszyński R, Irzmański R. The Significance of Genetically Determined Methylation and Folate Metabolism Disorders in the Pathogenesis of Coronary Artery Disease: A Target for New Therapies? Int J Mol Sci 2024; 25:6924. [PMID: 39000032 PMCID: PMC11241586 DOI: 10.3390/ijms25136924] [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: 05/18/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Methylation is a biochemical process involving the addition of a methyl group (-CH3) to various chemical compounds. It plays a crucial role in maintaining the homeostasis of the endothelium, which lines the interior surface of blood vessels, and has been linked, among other conditions, to coronary artery disease (CAD). Despite significant progress in CAD diagnosis and treatment, intensive research continues into genotypic and phenotypic CAD biomarkers. This review explores the significance of the methylation pathway and folate metabolism in CAD pathogenesis, with a focus on endothelial dysfunction resulting from deficiency in the active form of folate (5-MTHF). We discuss emerging areas of research into CAD biomarkers and factors influencing the methylation process. By highlighting genetically determined methylation disorders, particularly the MTHFR polymorphism, we propose the potential use of the active form of folate (5-MTHF) as a novel CAD biomarker and personalized pharmaceutical for selected patient groups. Our aim is to improve the identification of individuals at high risk of CAD and enhance their prognosis.
Collapse
Affiliation(s)
| | - Robert Pietruszyński
- Cardiology Outpatient Clinic, Military Medical Academy Memorial Teaching Hospital of the Medical University of Lodz—Central Veterans’ Hospital, 90-549 Lodz, Poland;
| | - Robert Irzmański
- Department of Internal Medicine, Rehabilitation and Physical Medicine, Medical University of Lodz, 90-645 Lodz, Poland;
| |
Collapse
|
42
|
Poniku A, Batalli A, Shita D, Rexhaj Z, Ferati A, Leka R, Bajraktari A, Abdyli G, Haliti E, Ibrahimi P, Karahoda R, Elezi S, Shatri F, Bytyçi I, Henein M, Bajraktari G. Smoking and Hypertriglyceridemia Predict ST-Segment Elevation Myocardial Infarction in Kosovo Patients with Acute Myocardial Infarction. Clin Pract 2024; 14:1149-1158. [PMID: 38921269 PMCID: PMC11202547 DOI: 10.3390/clinpract14030091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/28/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Myocardial infarction (MI), presented as ST-segment elevation MI (STEMI) and non-ST-segment elevation MI (NSTEMI), is influenced by atherosclerosis risk factors. AIM The aim of this study was to assess the patterns of presentation of patients with acute MI in Kosovo. METHODS This was a cross-sectional study conducted at the University Clinical Center of Kosovo, which included all patients hospitalized with acute MI over a period of 7 years. RESULTS Among the 7353 patients admitted with acute MI (age 63 ± 12 years, 29% female), 59.4% had STEMI and 40.6% had NSTEMI. The patients with NSTEMI patients less (48.3% vs. 54%, p < 0.001), but more of them had diabetes (37.8% vs. 33.6%, p < 0.001), hypertension (69.6% vs. 63%, p < 0.001), frequently had a family history of coronary artery disease (CAD) (40% vs. 38%, p = 0.009), and had more females compared to the patients with STEMI (32% vs. 27%, p < 0.001). The patients with NSTEMI underwent less primary percutaneous interventions compared with the patients with STEMI (43.6% vs. 55.2%, p < 0.001). Smoking [1.277 (1.117-1.459), p ˂ 0.001] and high triglycerides [0.791 (0.714-0.878), p = 0.02] were independent predictors of STEMI. CONCLUSIONS In Kosovo, patients with STEMI are more common than those with NSTEMI, and they were mostly males and more likely to have diabetes, hypertension, and a family history of CAD compared to those with NSTEMI. Smoking and high triglycerides proved to be the strongest predictors of acute STEMI in Kosovo, thus highlighting the urgent need for optimum atherosclerosis risk control and education strategies.
Collapse
Affiliation(s)
- Afrim Poniku
- Medical Faculty, University of Prishtina, 10000 Prishtina, Kosovo; (A.P.); (D.S.); (G.A.); (E.H.); (G.B.)
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
| | - Arlind Batalli
- Medical Faculty, University of Prishtina, 10000 Prishtina, Kosovo; (A.P.); (D.S.); (G.A.); (E.H.); (G.B.)
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
| | - Dua Shita
- Medical Faculty, University of Prishtina, 10000 Prishtina, Kosovo; (A.P.); (D.S.); (G.A.); (E.H.); (G.B.)
| | - Zarife Rexhaj
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
| | - Arlind Ferati
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
| | - Rita Leka
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
| | - Artan Bajraktari
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
- Institute of Public Health and Clinical Medicine, Umeå University, 90187 Umeå, Sweden;
| | - Genc Abdyli
- Medical Faculty, University of Prishtina, 10000 Prishtina, Kosovo; (A.P.); (D.S.); (G.A.); (E.H.); (G.B.)
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
| | - Edmond Haliti
- Medical Faculty, University of Prishtina, 10000 Prishtina, Kosovo; (A.P.); (D.S.); (G.A.); (E.H.); (G.B.)
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
| | - Pranvera Ibrahimi
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
- Institute of Public Health and Clinical Medicine, Umeå University, 90187 Umeå, Sweden;
| | - Rona Karahoda
- Research Unit, Heimerer College, 10000 Prishtina, Kosovo;
| | - Shpend Elezi
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
| | - Faik Shatri
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
| | - Ibadete Bytyçi
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
- Institute of Public Health and Clinical Medicine, Umeå University, 90187 Umeå, Sweden;
| | - Michael Henein
- Institute of Public Health and Clinical Medicine, Umeå University, 90187 Umeå, Sweden;
| | - Gani Bajraktari
- Medical Faculty, University of Prishtina, 10000 Prishtina, Kosovo; (A.P.); (D.S.); (G.A.); (E.H.); (G.B.)
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo; (Z.R.); (A.F.); (R.L.); (A.B.); (P.I.); (S.E.); (F.S.); (I.B.)
- Institute of Public Health and Clinical Medicine, Umeå University, 90187 Umeå, Sweden;
| |
Collapse
|
43
|
Kim H, Ahn JM, Kang DY, Lee J, Choi Y, Park SJ, Park DW. Management of Coronary Vulnerable Plaque With Medical Therapy or Local Preventive Percutaneous Coronary Intervention. JACC. ASIA 2024; 4:425-443. [PMID: 39100699 PMCID: PMC11291350 DOI: 10.1016/j.jacasi.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/20/2024] [Accepted: 04/04/2024] [Indexed: 08/06/2024]
Abstract
Acute coronary syndromes (ACS) often result from the rupture or erosion of high-risk coronary atherosclerotic plaques (ie, vulnerable plaques). Advances in intracoronary imaging such as intravascular ultrasound, optical coherence tomography, or near-infrared spectroscopy have improved the identification of vulnerable plaques, characterized by large plaque burden, small minimal luminal area, thin fibrous cap, and large lipid content. Although pharmacology, including lipid-lowering agents, and intensive risk-factor control are pivotal for management of vulnerable plaques and secondary prevention, recurrent events tend to accrue despite intensive pharmacotherapy. Therefore, it has been hypothesized that local preventive percutaneous coronary intervention may passivate these vulnerable plaques, preventing the occurrence of plaque-related ACS. However, solid evidence is lacking on its use for treatment of non-flow-limiting vulnerable plaques. As such, the optimal management of vulnerable plaques has not been established. Herein, we have reviewed the diagnosis and management of vulnerable plaques, focusing on systematic pharmacology and focal treatments.
Collapse
Affiliation(s)
- Hoyun Kim
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jung-Min Ahn
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Do-Yoon Kang
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jinho Lee
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yeonwoo Choi
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung-Jung Park
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Duk-Woo Park
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
44
|
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
|
45
|
Poznyak AV, Yakovlev AA, Popov MА, Zhuravlev AD, Sukhorukov VN, Orekhov AN. WITHDRAWN: Coronary atherosclerotic plaque regression strategies. J Biomed Res 2024; 39:1-21. [PMID: 38808553 DOI: 10.7555/jbr.37.20230223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Abstract
Ahead of Print article withdrawn by publisher.
Collapse
Affiliation(s)
| | - Alexey A Yakovlev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 109240, Russia
| | - Mikhail А Popov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - Alexander D Zhuravlev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - Vasily N Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| |
Collapse
|
46
|
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
|
47
|
Jiang Y, Yang ZG, Wang J, Jiang L, Han PL, Shi R, Li Y. Type 2 diabetes mellitus aggravates coronary atherosclerosis in hypertensive individuals based on coronary CT angiography: a retrospective propensity score-based study. Front Cardiovasc Med 2024; 11:1372519. [PMID: 38836061 PMCID: PMC11149417 DOI: 10.3389/fcvm.2024.1372519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Background The effect of type 2 diabetes mellitus (T2DM) on coronary atherosclerosis detected on coronary computed tomography angiography (CCTA) in hypertensive patients has attracted increasing attention. This study investigated the relationships of T2DM with coronary artery plaque characteristics and semiquantitative CCTA scores in hypertensive patients. Materials and methods In this single-center study, 1,700 hypertensive patients, including 850 T2DM [HT(T2DM+)] and 850 non-T2DM [HT(T2DM-)] individuals, were retrospectively analyzed after propensity matching. Plaque type, extent, coronary stenosis, segment involvement score (SIS), segment stenosis score (SSS), and CT-based Leaman score (CT-LeSc) based on CCTA were assessed and compared between the two groups. Results HT(T2DM+) patients had more coronary segments with calcified plaque (2.08 ± 2.20 vs. 1.40 ± 1.91), mixed plaque (2.90 ± 2.87 vs. 2.50 ± 2.66), nonobstructive stenosis (4.23 ± 2.44 vs. 3.62 ± 2.42), and obstructive stenosis (1.22 ± 2.18 vs. 0.78 ± 1.51), a lower proportion of 1-vessel disease (15.3% vs. 25.5%), a higher proportion of 3-vessel disease (59.6% vs. 46.7%), and higher SIS (5.5 ± 3.1 vs. 4.4 ± 3.0), SSS (10.3 ± 8.5 vs. 7.7 ± 7.1), and CT-LeSc (9.4 ± 5.6 vs. 7.9 ± 5.2) than HT(T2DM-) patients (all P-values <0.05). Multivariable analysis revealed that T2DM was an independent risk factor for calcified plaque [odds ratio (OR) = 2.213], obstructive coronary artery disease (CAD) (OR = 1.271), multivessel disease (OR = 1.838), SIS > 4 (OR = 1.910), SSS > 6 (OR = 1.718), and CT-LeSc > 5 (OR = 1.584) in hypertension population (all P-values <0.05). Conclusion T2DM was independently associated with the presence of calcified coronary artery plaque and increased the risk of obstructive CAD, multivessel disease, and CT-LeSc > 5 in hypertensive patients. More attention should be given to the assessment and management for coronary atherosclerosis in hypertensive patients with T2DM, as this population may have a higher risk of cardiovascular events.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhi-Gang Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jin Wang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Jiang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pei-Lun Han
- West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Shi
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
48
|
Cheng Q, Sun J, Zhong H, Wang Z, Liu C, Zhou S, Deng J. Research trends in lipid-lowering therapies for coronary heart disease combined with hyperlipidemia: a bibliometric study and visual analysis. Front Pharmacol 2024; 15:1393333. [PMID: 38828451 PMCID: PMC11140088 DOI: 10.3389/fphar.2024.1393333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/03/2024] [Indexed: 06/05/2024] Open
Abstract
Background Cardiovascular disease (CVD) poses a significant global health and economic challenge, with atherosclerosis being a primary cause. Over the past 40 years, substantial research has been conducted into the prevention and reversal of atherosclerosis, resulting in the development of lipid-lowering agents such as statins and fibrates. Despite the extensive literature and formulation of numerous therapeutic guidelines in this domain, a comprehensive bibliometric analysis of the current research landscape and trends has not been performed. This study aimed to elucidate the evolution and milestones of research into lipid-lowering treatments for coronary heart disease (CHD) in conjunction with hyperlipidemia through bibliometric analysis, offering insights into future directions for treatment strategies. Methods This study examined publications from 1986 to 2023 retrieved from the Web of Science database (Core Collection). Utilizing tools such as VOSviewer, Pajek, and CiteSpace, we analyzed publication and citation numbers, H-indexes, contributions by countries and institutions, authorship, journal sources, and keyword usage to uncover research trajectories and areas of focus. Results Our analysis of 587 publications revealed a recent surge in research output, particularly post-2003. The American Journal of Cardiology published the highest number of studies, with 40 articles, whereas Circulation received the highest number of citations (6,266). Key contributors included the United States, Japan, and China, with the United States leading in citation numbers and the H-index. Harvard University and Leiden University emerged as pivotal institutions, and Professors J. Wouter Jukema and Robert P. Giugliano were identified as leading experts. Keyword analysis disclosed five thematic clusters, indicating a shift in research towards new drug combinations and strategies, signaling future research directions. Conclusion The last 4 decades have seen a notable rise in publications on lipid-lowering therapies for CHD and hyperlipidemia, with the United States retaining world-leading status. The increase in international collaboration aids the shift towards research into innovative lipid-lowering agents and therapeutic approaches. PCSK9 inhibitors and innovative combination therapies, including antisense oligonucleotides and angiopoietin-like protein 3 inhibitors, provide avenues for future research, intending to maximize the safety and efficacy of treatment approaches.
Collapse
Affiliation(s)
- Quankai Cheng
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jingjing Sun
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Haicheng Zhong
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ziming Wang
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chang Liu
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Sheng Zhou
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jie Deng
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
49
|
Fezzi S, Malakouti S, Sivalingam J, Khater J, Ribichini F, Cortese B. Drug-Coated Balloon in Acute Coronary Syndromes: Ready for the Prime Time? Curr Cardiol Rep 2024; 26:359-372. [PMID: 38619711 DOI: 10.1007/s11886-024-02037-2] [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] [Accepted: 02/28/2024] [Indexed: 04/16/2024]
Abstract
PURPOSE OF REVIEW Acute coronary syndromes (ACS) are a major global health concern. Percutaneous coronary intervention (PCI) with new-generation drug-eluting stents (DES) has been endorsed as safe and effective in the management of culprit and non-culprit lesions of ACS. However, permanent metallic implants may have drawbacks, including the need for prolonged dual antiplatelet therapy (DAPT) and the risk of long-term stent-related complications. An alternative approach using drug-coated balloons (DCBs) is gaining growing interest, having the potential of delivering therapy directly to vulnerable plaques, avoiding the need for permanent metallic implants, and potentially allowing for better long-term medical treatment. Despite limited evidence, DCB is being explored in several patients' subgroups. This review aims to discuss the existing evidence regarding DCB in ACS management. RECENT FINDINGS DCB appears to be a promising strategy in the management of ACS, showing comparable angiographic and clinical results as compared to new-generation DES in relatively small clinical trials or large prospective registries. The advantage of avoiding permanent implants is particularly appealing in this setting, where DCB has the potential of delivering anti-atherogenic local therapy directly to vulnerable plaques still amenable to atherogenic regression. This review seeks to underline the theoretical background of DCB use and reports the available evidence in its support in the specific setting of ACS. In the context of ACS, the use of DCB is highly attractive, offering a dedicated anti-atherogenic local therapy, capable of addressing a broad range of vulnerable plaques and patients.
Collapse
Affiliation(s)
- Simone Fezzi
- Fondazione Ricerca e Innovazione Cardiovascolare, Milan, Italy
- University of Verona, Verona, Italy
| | | | | | - Jacinthe Khater
- DCB Academy, Milan, Italy
- Faculty of Medical Sciences, Lebanese University Rafic Hariri University Campus, Hadath, Lebanon
| | | | - Bernardo Cortese
- Fondazione Ricerca e Innovazione Cardiovascolare, Milan, Italy.
- DCB Academy, Milan, Italy.
| |
Collapse
|
50
|
Song Y, Wang Q, Jia L. Omega-3 fatty acids and their influence on hypertension and coronary atherosclerosis: Insights from a Mendelian randomization approach. J Clin Hypertens (Greenwich) 2024; 26:382-390. [PMID: 38450969 PMCID: PMC11007784 DOI: 10.1111/jch.14782] [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: 11/10/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 03/08/2024]
Abstract
It has been suggested that Omega-3 fatty acids may improve endothelial thickness and thereby reduce the onset of cardiovascular diseases such as coronary atherosclerosis and hypertension. However, published observational epidemiological studies on the relationship between cardiovascular disease (CVD) and Omega-3 fatty acids remain inconclusive. Here, we performed a two-sample Mendelian randomisation analysis using publicly available GWAS pooled statistics to study a GWAS dataset of 16 380 466 SNPs in 23 363 cases and 195 429 controls (also of European ancestry) to determine genetic susceptibility to hypertension. We performed random-effects Inverse Variance Weighted (IVW) Mendelian Randomization (MR) analyses supplemented by a series of sensitivity assessments to measure the robustness of the findings and to detect any violations of the MR assumptions. During the course of the study, we used IVW, MR-Egger, and weighted median regression to infer that Omega-3 intake has a potentially adverse effect against atherosclerosis, although the trend was not significant (OR = 1.1198; 95%; CI: 0.9641-1.3006, p = .130). Meanwhile, our analyses showed a statistically significant negative association between Omega-3 fatty acid levels and risk of hypertension (OR = 0.9006; 95% CI: 0.8179-0.9917, p = .033). In addition, we explored the causal relationship between atherosclerosis and hypertension and found a significant correlation (OR = 1.3036; 95% CI: 1.0672-1.5923, p = .009). In conclusion, our extensive data investigated by MR suggest that elevated levels of Omega-3 fatty acids may be associated with an decreased risk of hypertension. Although there is no direct link between hypertension and atherosclerosis, the possibility of a subtle association cannot be categorically excluded.
Collapse
Affiliation(s)
- Yuchen Song
- College of Integrated Chinese and Western MedicineLiaoning University of Traditional Chinese MedicineShenyangLiaoningChina
| | - Qun Wang
- Key Laboratory of Ministry of Education for TCM Viscera‐State Theory and ApplicationsMinistry of Education of ChinaLiaoning University of Traditional Chinese MedicineShenyangLiaoningChina
| | - Lianqun Jia
- Key Laboratory of Ministry of Education for TCM Viscera‐State Theory and ApplicationsMinistry of Education of ChinaLiaoning University of Traditional Chinese MedicineShenyangLiaoningChina
| |
Collapse
|