1
|
Cho S, Cho H, Min H, Lee JG, Kim TO, Lee PH, Lee SW, Kang SJ. Clinical impact of deep learning-derived intravascular ultrasound characteristics in patients with deferred coronary artery. Int J Cardiol 2024; 417:132543. [PMID: 39265789 DOI: 10.1016/j.ijcard.2024.132543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/23/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Prognostic markers for long-term outcomes are lacking in patients with deferred (nonculprit) coronary artery lesions. This study aimed to identify the morphological criteria for predicting adverse outcomes and validate their clinical impact. Using deep learning models, we extracted geometrical parameters and maximal attenuation (or calcium) burden index (ABI-max or CBI-max) from the intravascular ultrasound (IVUS) images of nonculprit vessels in 1115 patients. The endpoints included cardiac death, myocardial infarction, and target vessel revascularization of nonculprit vessel. Cardiac death occurred in 27 (2.4 %) patients at 3 years and 39 (3.5 %) patients at 5 years. At 5 years, the cardiac death-free survival rate was significantly lower with ABP-max ≥11.37 % vs. < 11.37 % (90.0 % vs. 98.7 %), CBI-max ≥13.40 % vs. < 13.40 % (92.8 % vs. 98.4 %), and percent atheroma volume ≥ 51.35 % vs. < 51.35 % (94.0 % vs. 97.7) (all log-rank p < 0.001). The independent predictors of 5-year cardiovascular mortality were age (hazard ratio [HR] 1.21), female sex (HR 0.33), history of heart failure (HR 6.06), chronic kidney disease (HR 18.28), ABI-max (HR 1.04), and CBI-max (HR 1.05). The independent determinants of 5-year target vessel revascularization of nonculprit vessel were fractional flow reserve (HR 0.95 per 0.01 increase), minimal lumen area (HR 0.63), and plaque burden (HR 1.15). In patients with nonculprit coronary artery lesions, a large burden of attenuated or calcified plaques predicted cardiac mortality, while IVUS geometry was associated with repeat revascularization. Thus, deep learning-based IVUS analysis of the whole target vessel may help clinicians identify high-risk lesions.
Collapse
Affiliation(s)
- Sungsoo Cho
- Division of Cardiology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyungjoo Cho
- Department of Cardiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hyunseok Min
- Department of Cardiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - June-Goo Lee
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Tae Oh Kim
- Department of Cardiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Pil Hyung Lee
- Department of Cardiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Seung-Whan Lee
- Department of Cardiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Soo-Jin Kang
- Department of Cardiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Di Vito L, Di Giusto F, Mazzotta S, Scalone G, Bruscoli F, Silenzi S, Selimi A, Angelini M, Galieni P, Grossi P. Management of vulnerable patient phenotypes and acute coronary syndrome mechanisms. Int J Cardiol 2024; 415:132365. [PMID: 39029561 DOI: 10.1016/j.ijcard.2024.132365] [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: 05/22/2024] [Revised: 07/07/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Atherosclerosis is a chronic vascular disease. Its prevalence increases with aging. However, atherosclerosis may also affect young subjects without significant exposure to the classical risk factors. Recent evidence indicates clonal hematopoiesis of indeterminate potential (CHIP) as a novel cardiovascular risk factor that should be suspected in young patients. CHIP represents a link between impaired bone marrow and atherosclerosis. Atherosclerosis may present with an acute symptomatic manifestation or subclinical events that favor plaque growth. The outcome of a plaque relies on a balance of innate and environmental factors. These factors can influence the processes that initiate and propagate acute plaque destabilization leading to intraluminal thrombus formation or subclinical vessel healing. Thirty years ago, the first autopsy study revealed that coronary plaques can undergo rupture even in subjects without a known cardiovascular history. Nowadays, cardiac magnetic resonance studies demonstrate that this phenomenon is not rare. Myocardial infarction is mainly due to plaque rupture and plaque erosion that have different pathophysiological mechanisms. Plaque erosion carries a better prognosis as compared to plaque rupture. Thus, a tailored conservative treatment has been proposed and some studies demonstrated it to be safe. On the contrary, plaque rupture is typically associated with inflammation and anti-inflammatory treatments have been proposed in response to persistently elevate biomarkers of systemic inflammation. In conclusion, atherosclerosis may present in different forms or phenotypes. Vulnerable patient phenotypes, identified by using intravascular imaging techniques, biomarkers, or even genetic analyses, are characterized by distinctive pathophysiological mechanisms. These different phenotypes merit tailored management.
Collapse
Affiliation(s)
- Luca Di Vito
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy.
| | | | - Serena Mazzotta
- Department of Haematology and Stem Cell Transplantation Unit C. e G, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Giancarla Scalone
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy
| | - Filippo Bruscoli
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy
| | - Simona Silenzi
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy
| | - Adelina Selimi
- University Hospital "Umberto I-Lancisi-Salesi", Ancona, Italy
| | - Mario Angelini
- Department of Haematology and Stem Cell Transplantation Unit C. e G, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Piero Galieni
- Department of Haematology and Stem Cell Transplantation Unit C. e G, Mazzoni Hospital, Ascoli Piceno, Italy
| | | |
Collapse
|
3
|
Gastanadui MG, Margaroli C, Litovsky S, Richter RP, Wang D, Xing D, Wells JM, Gaggar A, Nanda V, Patel RP, Payne GA. Spatial Transcriptomic Approach to Understanding Coronary Atherosclerotic Plaque Stability. Arterioscler Thromb Vasc Biol 2024; 44:e264-e276. [PMID: 39234691 PMCID: PMC11499036 DOI: 10.1161/atvbaha.123.320330] [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: 10/26/2023] [Accepted: 08/08/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Coronary atherosclerotic plaques susceptible to acute coronary syndrome have traditionally been characterized by their surrounding cellular architecture. However, with the advent of intravascular imaging, novel mechanisms of coronary thrombosis have emerged, challenging our contemporary understanding of acute coronary syndrome. These intriguing findings underscore the necessity for a precise molecular definition of plaque stability. Considering this, our study aimed to investigate the vascular microenvironment in patients with stable and unstable plaques using spatial transcriptomics. METHODS Autopsy-derived coronary arteries were preserved and categorized by plaque stability (n=5 patients per group). We utilized the GeoMx spatial profiling platform and Whole Transcriptome Atlas to link crucial histological morphology markers in coronary lesions with differential gene expression in specific regions of interest, thereby mapping the vascular transcriptome. This innovative approach allowed us to conduct cell morphological and spatially resolved transcriptional profiling of atherosclerotic plaques while preserving crucial intercellular signaling. RESULTS We observed intriguing spatial and cell-specific transcriptional patterns in stable and unstable atherosclerotic plaques, showcasing regional variations within the intima and media. These regions exhibited differential expression of proinflammatory molecules (eg, IFN-γ [interferon-γ], MHC [major histocompatibility complex] class II, proinflammatory cytokines) and prothrombotic signaling pathways. By using lineage tracing through spatial deconvolution of intimal CD68+ (cluster of differentiation 68) cells, we characterized unique, intraplaque subpopulations originating from endothelial, smooth muscle, and myeloid lineages with distinct regional locations associated with plaque instability. In addition, unique transcriptional signatures were observed in vascular smooth muscle and CD68+ cells among plaques exhibiting coronary calcification. CONCLUSIONS Our study illuminates distinct cell-specific and regional transcriptional alterations present in unstable plaques. Furthermore, we characterize spatially resolved, in situ evidence supporting cellular transdifferentiation and intraplaque plasticity as significant contributors to plaque instability in human coronary atherosclerosis. Our results provide a powerful resource for the identification of novel mediators of acute coronary syndrome, opening new avenues for preventative and therapeutic treatments.
Collapse
Affiliation(s)
- Maria G Gastanadui
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Camilla Margaroli
- Department of Pathology, Division of Molecular & Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Silvio Litovsky
- Department of Pathology, Division of Anatomic Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert P. Richter
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pediatrics, Division of Pediatric Critical Care, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dezhi Wang
- Department of Pathology, Pathology Core Research Laboratory, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dongqi Xing
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J. Michael Wells
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL
| | - Amit Gaggar
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vivek Nanda
- Department of Pathology, Division of Molecular & Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rakesh P. Patel
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pathology, Division of Molecular & Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory A. Payne
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL
| |
Collapse
|
4
|
Tian Y, Shao S, Feng H, Zeng R, Li S, Zhang Q. Targeting senescent cells in atherosclerosis: Pathways to novel therapies. Ageing Res Rev 2024; 101:102502. [PMID: 39278272 DOI: 10.1016/j.arr.2024.102502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/16/2024] [Accepted: 09/08/2024] [Indexed: 09/18/2024]
Abstract
Targeting senescent cells has recently emerged as a promising strategy for treating age-related diseases, such as atherosclerosis, which significantly contributes to global cardiovascular morbidity and mortality. This review elucidates the role of senescent cells in the development of atherosclerosis, including persistently damaging DNA, inducing oxidative stress and secreting pro-inflammatory factors known as the senescence-associated secretory phenotype. Therapeutic approaches targeting senescent cells to mitigate atherosclerosis are summarized in this review, which include the development of senotherapeutics and immunotherapies. These therapies are designed to either remove these cells or suppress their deleterious effects. These emerging therapies hold potential to decelerate or even alleviate the progression of AS, paving the way for new avenues in cardiovascular research and treatment.
Collapse
Affiliation(s)
- Yuhan Tian
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai-Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Sihang Shao
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore
| | - Haibo Feng
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Rui Zeng
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai-Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Shanshan Li
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai-Tibet Plateau, Southwest Minzu University, Chengdu 610041, China.
| | - Qixiong Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Department of Pharmacy, Sichuan Provincial People's Hospital East Sichuan Hospital & Dazhou First People's Hospital, Dazhou 635000, China.
| |
Collapse
|
5
|
Cai GF, Chen SW, Huang JK, Lin SR, Huang GH, Lin CH. Decoding marker genes and immune landscape of unstable carotid plaques from cellular senescence. Sci Rep 2024; 14:26196. [PMID: 39478143 PMCID: PMC11525637 DOI: 10.1038/s41598-024-78251-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/29/2024] [Indexed: 11/02/2024] Open
Abstract
Recently, cellular senescence-induced unstable carotid plaques have gained increasing attention. In this study, we utilized bioinformatics and machine learning methods to investigate the correlation between cellular senescence and the pathological mechanisms of unstable carotid plaques. Our aim was to elucidate the causes of unstable carotid plaque progression and identify new therapeutic strategies. First, differential expression analysis was performed on the test set GSE43292 to identify differentially expressed genes (DEGs) between the unstable plaque group and the control group. These DEGs were intersected with cellular senescence-associated genes to obtain 40 cellular senescence-associated DEGs. Subsequently, key genes were then identified through weighted gene co-expression network analysis, random forest, Recursive Feature Elimination for Support Vector Machines algorithm and cytoHubba plugin. The intersection yielded 3 CSA-signature genes, which were validated in the external validation set GSE163154. Additionally, we assessed the relationship between these CSA-signature genes and the immune landscape of the unstable plaque group. This study suggests that cellular senescence may play an important role in the progression mechanism of unstable plaques and is closely related to the influence of the immune microenvironment. Our research lays the foundation for studying the progression mechanism of unstable carotid plaques and provides some reference for targeted therapy.
Collapse
Affiliation(s)
- Gang-Feng Cai
- Department of Neurosurgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Shao-Wei Chen
- Department of Neurosurgery, Quanzhou Orthopedic-Traumatological Hospital, Quanzhou, Fujian, China
| | - Jin-Kai Huang
- Department of Neurosurgery, Quanzhou Orthopedic-Traumatological Hospital, Quanzhou, Fujian, China
| | - Shi-Rong Lin
- Department of Neurosurgery, Quanzhou Orthopedic-Traumatological Hospital, Quanzhou, Fujian, China
| | - Guo-He Huang
- Department of Neurosurgery, Quanzhou Orthopedic-Traumatological Hospital, Quanzhou, Fujian, China
| | - Cai-Hou Lin
- Department of Neurosurgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
6
|
Tong J, Wang Z, Zhang J, Gao R, Liu X, Liao Y, Guo X, Wei Y. Advanced Applications of Nanomaterials in Atherosclerosis Diagnosis and Treatment: Challenges and Future Prospects. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58072-58099. [PMID: 39432384 DOI: 10.1021/acsami.4c13607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Atherosclerosis-induced coronary artery disease is a major cause of cardiovascular mortality. Clinically, conservative treatment strategies for atherosclerosis still focus on lifestyle interventions and the use of lipid-lowering and anticoagulant medications. Despite achieving some therapeutic effects, these approaches are limited by low bioavailability, long intervention periods, and significant side effects. With the advancement of nanotechnology, nanomaterials have demonstrated extraordinary potential in the biomedical field. Their excellent biocompatibility, surface modifiability, and high targeting capability not only enable efficient diagnosis of plaque progression but also allow precise drug delivery within atherosclerotic plaques, significantly enhancing drug bioavailability and reducing systemic side effects. Here, we systematically review the current research progress of nanomaterials in the field of atherosclerosis to summarize not only the types of nanomaterials but also their applications in both the diagnosis and treatment of atherosclerosis. Notably, in the context of plaque therapy, we provide a comprehensive overview of current nanomaterial applications based on their targeted therapeutic systems for different cell types within plaques. Additionally, we address the persistent challenge of clinical translation of nanomaterials by summarizing current issues and providing directions for innovation and improvement in nanomaterial design. Overall, we believe that this review systematically summarizes the applications and challenges of biomedical nanomaterials in atherosclerosis diagnosis and therapy, thereby offering insights and references for the development of therapeutic materials for atherosclerosis.
Collapse
Affiliation(s)
- Junran Tong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiwen Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiahui Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ran Gao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangfei Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yuhan Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaopeng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
7
|
Yamaji K, Kanenawa K, Morofuji T, Nishikawa R, Imada K, Kohjitani H, Watanabe H, Tazaki J, Taniwaki M, Koga S, Akashi R, Kubo S, Ohya M, Kikuchi T, Ohira H, Numasawa Y, Arikawa M, Iwama M, Kitai T, Kobayashi Y, Shiomi H, Tada T, Yamaji Y, Daidoji H, Ohtani H, Furukawa Y, Kadota K, Toyofuku M, Ando K, Ono K, Kimura T. Serial Optical Coherence Tomography Assessment of Coronary Atherosclerosis and Long-Term Clinical Outcomes. J Am Heart Assoc 2024:e034458. [PMID: 39435729 DOI: 10.1161/jaha.123.034458] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 06/24/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND The impact of high-risk coronary artery plaques identified using optical coherence tomography on late luminal narrowing and clinical events remains poorly understood. METHODS AND RESULTS This multicenter prospective study included 176 patients who underwent percutaneous coronary intervention and serial optical coherence tomography at baseline and 1-year follow-up to investigate nontarget regions with angiographically intermediate stenosis. At 1 year after percutaneous coronary intervention, the coronary artery lumen area decreased significantly from 6.06 (95% CI, 5.60-6.53) mm2 to 5.88 (95% CI, 5.41-6.35) mm2 (difference, -0.18; 95% CI, -0.22 to -0.14 mm2; P<0.001), particularly in thin-cap fibroatheromas, thick-cap fibroatheromas, mixed plaques, and fibrous plaques. The prevalence of fibroatheroma decreased from 38% to 36% (P<0.001), whereas calcified plaque increased from 31% to 34% (P<0.001), accompanied by a significant increase in calcium thickness and angle. Diabetes and current smoking habits were independently associated with increasing calcium prevalence. Patients with thin-cap fibroatheroma had a significantly higher 3-year risk of ischemia-driven nontarget vessel revascularization (hazard ratio, 2.42 [95% CI, 1.03-5.71]; P=0.04), primarily due to revascularization in the imaged region. No significant association was observed between coronary artery calcium prevalence and clinical outcomes within 3 years. CONCLUSIONS The coronary artery lumen area significantly decreased over a 1-year interval, particularly in thin-cap fibroatheromas, thick-cap fibroatheromas, mixed plaques, and fibrous plaques. Although thin-cap fibroatheroma prevalence was associated with higher risk of ischemia-driven nontarget vessel revascularization, no significant association was noted between coronary artery calcium prevalence and clinical outcomes within 3 years. The interaction between calcium progression and long-term clinical events necessitates further investigation. REGISTRATION URL: https://www.umin.ac.jp/ctr/; Unique Identifier: UMIN000031937.
Collapse
Affiliation(s)
- Kyohei Yamaji
- Department of Cardiovascular Medicine Kyoto University Graduate School of Medicine Kyoto Japan
- Department of Cardiology Kokura Memorial Hospital Kitakyushu Japan
| | - Kenji Kanenawa
- Department of Cardiology Kokura Memorial Hospital Kitakyushu Japan
| | - Toru Morofuji
- Department of Cardiology Kokura Memorial Hospital Kitakyushu Japan
| | - Ryusuke Nishikawa
- Department of Cardiovascular Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Kazuaki Imada
- Department of Cardiovascular Medicine Kyoto University Graduate School of Medicine Kyoto Japan
- Department of Cardiology Kokura Memorial Hospital Kitakyushu Japan
| | - Hirohiko Kohjitani
- Department of Cardiovascular Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Hiroki Watanabe
- Department of Cardiology Japanese Red Cross Wakayama Medical Center Wakayama Japan
| | - Junichi Tazaki
- Department of Cardiology Japanese Red Cross Wakayama Medical Center Wakayama Japan
| | | | - Seiji Koga
- Department of Cardiovascular Medicine Nagasaki University Graduate School of Biomedical Sciences Nagasaki Japan
| | - Ryohei Akashi
- Department of Cardiovascular Medicine Nagasaki University Graduate School of Biomedical Sciences Nagasaki Japan
| | - Shunsuke Kubo
- Department of Cardiology Kurashiki Central Hospital Kurashiki Japan
| | - Masanobu Ohya
- Department of Cardiology Kurashiki Central Hospital Kurashiki Japan
| | | | | | - Yohei Numasawa
- Department of Cardiology Japanese Red Cross Ashikaga Hospital Ashikaga Japan
| | - Masaya Arikawa
- Department of Cardiology National Hospital Organization Oita Medical Center Yokota Oita Japan
| | - Makoto Iwama
- Department of Cardiology Gifu Prefectural General Medical Center Gifu Japan
| | - Takeshi Kitai
- Department of Cardiovascular Medicine Kobe City Medical Center General Hospital Kobe Japan
- Department of Cardiovascular Medicine National Cerebral and Cardiovascular Center Suita Japan
| | - Yohei Kobayashi
- Department of Cardiology Osaka Red Cross Hospital Osaka Japan
| | - Hiroki Shiomi
- Department of Cardiovascular Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Tomohisa Tada
- Department of Cardiology Shizuoka General Hospital Shizuoka Japan
| | - Yuhei Yamaji
- Department of Cardiology The Tazuke Kofukai Medical Research Institute, Kitano Hospital Osaka Japan
| | - Hyuma Daidoji
- Department of Cardiology Yamagata Prefectural Central Hospital Yamagata Japan
| | - Hayato Ohtani
- Department of Cardiology, Internal Medicine 3 Hamamatsu University School of Medicine Hamamatsu Japan
| | - Yutaka Furukawa
- Department of Cardiovascular Medicine Kobe City Medical Center General Hospital Kobe Japan
| | - Kazushige Kadota
- Department of Cardiology Kurashiki Central Hospital Kurashiki Japan
| | - Mamoru Toyofuku
- Department of Cardiology Japanese Red Cross Wakayama Medical Center Wakayama Japan
| | - Kenji Ando
- Department of Cardiology Kokura Memorial Hospital Kitakyushu Japan
| | - Koh Ono
- Department of Cardiovascular Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine Kyoto University Graduate School of Medicine Kyoto Japan
- Division of Cardiology Hirakata Kohsai Hospital Osaka Japan
| |
Collapse
|
8
|
Deng C, Liu Z, Li C, Xu G, Zhang R, Bai Z, Hu X, Xia Q, Pan L, Wang S, Xia J, Zhao R, Shi B. Predictive models for cholesterol crystals and plaque vulnerability in acute myocardial infarction: Insights from an optical coherence tomography study. Int J Cardiol 2024; 418:132610. [PMID: 39366560 DOI: 10.1016/j.ijcard.2024.132610] [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: 07/12/2024] [Revised: 09/08/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Cholesterol crystals (CCs) are recognized as a risk factor for vulnerable atherosclerotic plaque rupture (PR) and major adverse cardiovascular events. However, their predictive factors and association with plaque vulnerability in patients with acute myocardial infarction (AMI) remain insufficiently explored. Therefore, This study aims to investigate the association between CCs and plaque vulnerability in culprit lesions of AMI patients, identify the factors influencing CCs formation, and develop a predictive model for CCs. METHODS A total of 431 culprit lesions from AMI patients who underwent pre-intervention optical coherence tomography (OCT) imaging were analyzed. Patients were divided into groups based on the presence or absence of CCs and PR. The relationship between CCs and plaque vulnerability was evaluated. A risk nomogram for predicting CCs was developed using the least absolute shrinkage and selection operator and logistic regression analysis. RESULTS CCs were identified in 64.5 % of patients with AMI. The presence of CCs was associated with a higher prevalence of vulnerable plaque features, such as thin-cap fibroatheroma (TCFA), PR, macrophage infiltration, neovascularization, calcification, and thrombus, compared to patients without CCs. The CCs model demonstrated an area under the curve (AUC) of 0.676 for predicting PR. Incorporating CCs into the TCFA model (AUC = 0.656) significantly enhanced predictive accuracy, with a net reclassification improvement index of 0.462 (95 % confidence interval [CI]: 0.263-0.661, p < 0.001) and an integrated discrimination improvement index of 0.031 (95 % CI: 0.013-0.048, p = 0.001). Multivariate regression analysis identified the atherogenic index of plasma (odds ratio [OR] = 2.417), TCFA (OR = 1.759), macrophage infiltration (OR = 3.863), neovascularization (OR = 2.697), calcification (OR = 1.860), and thrombus (OR = 2.430) as independent risk factors for CCs formation. The comprehensive model incorporating these factors exhibited reasonable discriminatory ability, with an AUC of 0.766 (95 % CI: 0.717-0.815) in the training set and 0.753 (95 % CI: 0.704-0.802) in the internal validation set, reflecting good calibration. Decision curve analysis suggested that the model has potential clinical utility within a threshold probability range of approximately 18 % to 85 %. CONCLUSIONS CCs were associated with plaque vulnerability in the culprit lesions of AMI patients. Additionally, this study identified key factors influencing CCs formation and developed a predictive model with potential clinical applicability.
Collapse
Affiliation(s)
- Chancui Deng
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhijiang Liu
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chaozhong Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Guanxue Xu
- Department of Cardiology, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| | - Renyi Zhang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhixun Bai
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xingwei Hu
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qianhang Xia
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Li Pan
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Sha Wang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jie Xia
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
9
|
Ference BA, Braunwald E, Catapano AL. The LDL cumulative exposure hypothesis: evidence and practical applications. Nat Rev Cardiol 2024; 21:701-716. [PMID: 38969749 DOI: 10.1038/s41569-024-01039-5] [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] [Accepted: 05/02/2024] [Indexed: 07/07/2024]
Abstract
The trapping of LDL and other apolipoprotein B-containing lipoproteins within the artery wall causes atherosclerosis. As more LDL becomes trapped within the artery wall over time, the atherosclerotic plaque burden gradually increases, raising the risk of an acute cardiovascular event. Therefore, the biological effect of LDL on the risk of atherosclerotic cardiovascular disease (ASCVD) depends on both the magnitude and duration of exposure. Maintaining low levels of LDL-cholesterol (LDL-C) over time decreases the number of LDL particles trapped within the artery wall, slows the progression of atherosclerosis and, by delaying the age at which mature atherosclerotic plaques develop, substantially reduces the lifetime risk of ASCVD events. Summing LDL-C measurements over time to calculate cumulative exposure to LDL generates a unique biomarker that captures both the magnitude and duration of exposure, which facilitates the estimation of the absolute risk of having an acute cardiovascular event at any point in time. Titrating LDL-C lowering to keep cumulative exposure to LDL below the threshold at which acute cardiovascular events occur can effectively prevent ASCVD. In this Review, we provide the first comprehensive overview of how the LDL cumulative exposure hypothesis can guide the prevention of ASCVD. We also discuss the benefits of maintaining lower LDL-C levels over time and how this knowledge can be used to inform clinical practice guidelines as well as to design novel primary prevention trials and ASCVD prevention programmes.
Collapse
Affiliation(s)
- Brian A Ference
- DeepCausalAI Institute for Clinical Translation, Cambridge, UK.
| | - Eugene Braunwald
- TIMI Study Group, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Milan, Italy.
- Multimedica IRCCS, Milan, Italy.
| |
Collapse
|
10
|
Konishi T, Kamiyama K, Osato T, Yoshimoto T, Aoki T, Anzai T, Tanaka S. Increased Piezo1 expression in myofibroblasts in patients with symptomatic carotid atherosclerotic plaques undergoing carotid endarterectomy: A pilot study. Vascular 2024; 32:1063-1069. [PMID: 37499697 DOI: 10.1177/17085381231192380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
OBJECTIVES We aimed to investigate Piezo1 expression in myofibroblasts in symptomatic and asymptomatic patients undergoing carotid endarterectomy and its relationship with atherosclerotic plaque formation. METHODS This cross-sectional study analyzed carotid plaques of 17 randomly selected patients who underwent carotid endarterectomy from May 2015 to August 2017. In total, 51 sections (the most stenotic lesion, and the sections 5-mm proximal and distal) stained with hematoxylin-eosin and elastica-Masson were examined. Immunohistochemistry was performed using antibodies to Piezo1. The Piezo1 score of a section was calculated semiquantitatively, averaged across 30 randomly selected myofibroblasts in the fibrous cap of the plaque. RESULTS Of 17 patients (mean age: 74.2 ± 7.1 years), 15 were men, 9 had diabetes mellitus, and 13 had hypertension. Symptomatic patients had higher mean Piezo1 score than asymptomatic patients (1.78 ± 0.23 vs 1.34 ± 0.17, p < .001). Univariate linear regression analyses suggested an association between plaque rupture, thin-cap fibroatheroma and microcalcifications and the Piezo1 score (p = .001, .008, and 0.003, respectively). CONCLUSIONS Increased Piezo1 expression of myofibroblasts may be associated with atherosclerotic carotid plaque instability. Further study is warranted to support this finding.
Collapse
Affiliation(s)
- Takao Konishi
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kenji Kamiyama
- Department of Neurosurgery, Nakamura Memorial Hospital, Sapporo, Japan
| | - Toshiaki Osato
- Department of Neurosurgery, Nakamura Memorial Hospital, Sapporo, Japan
| | - Tetsuyuki Yoshimoto
- Department of Neurosurgery, Hokkaido Neurosurgical Memorial Hospital, Sapporo, Japan
| | - Takeshi Aoki
- Department of Neurosurgery, Hokkaido Neurosurgical Memorial Hospital, Sapporo, Japan
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| |
Collapse
|
11
|
Buonpane A, Trimarchi G, Ciardetti M, Coceani MA, Alagna G, Benedetti G, Berti S, Andò G, Burzotta F, De Caterina AR. Optical Coherence Tomography in Myocardial Infarction Management: Enhancing Precision in Percutaneous Coronary Intervention. J Clin Med 2024; 13:5791. [PMID: 39407851 PMCID: PMC11477163 DOI: 10.3390/jcm13195791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
In acute myocardial infarction (AMI), the urgency of coronary revascularization through percutaneous coronary intervention (PCI) is paramount, offering notable advantages over pharmacologic treatment. However, the persistent risk of adverse events, including recurrent AMI and heart failure post-revascularization, underscores the necessity for enhanced strategies in managing coronary artery disease. Traditional angiography, while widely employed, presents significant limitations by providing only two-dimensional representations of complex three-dimensional vascular structures, hampering the accurate assessment of plaque characteristics and stenosis severity. Intravascular imaging, specifically optical coherence tomography (OCT), significantly addresses these limitations with superior spatial resolution compared to intravascular ultrasound (IVUS). Within the context of AMI, OCT serves dual purposes: as a diagnostic tool to accurately identify culprit lesions in ambiguous cases and as a guide for optimizing PCI procedures. Its capacity to differentiate between various mechanisms of acute coronary syndrome, such as plaque rupture and spontaneous coronary dissection, enhances its diagnostic potential. Furthermore, OCT facilitates precise lesion preparation, optimal stent sizing, and confirms stent deployment efficacy. Recent meta-analyses indicate that OCT-guided PCI markedly improves safety and efficacy in revascularization, subsequently decreasing the risks of mortality and complications. This review emphasizes the critical role of OCT in refining patient-specific therapeutic approaches, aligning with the principles of precision medicine to enhance clinical outcomes for individuals experiencing AMI.
Collapse
Affiliation(s)
- Angela Buonpane
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Largo Agostino Gemelli, 1, 00168 Roma, Italy; (A.B.); (F.B.)
| | - Giancarlo Trimarchi
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (G.T.); (G.A.)
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Marco Ciardetti
- Cardiology and Pneumology Division, Fondazione Toscana G. Monasterio, 56124 Pisa, Italy; (M.C.); (M.A.C.)
| | - Michele Alessandro Coceani
- Cardiology and Pneumology Division, Fondazione Toscana G. Monasterio, 56124 Pisa, Italy; (M.C.); (M.A.C.)
| | - Giulia Alagna
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (G.T.); (G.A.)
| | - Giovanni Benedetti
- Fondazione Toscana G. Monasterio, Ospedale del Cuore G., Pasquinucci, 54100 Massa, Italy; (G.B.); (S.B.); (A.R.D.C.)
| | - Sergio Berti
- Fondazione Toscana G. Monasterio, Ospedale del Cuore G., Pasquinucci, 54100 Massa, Italy; (G.B.); (S.B.); (A.R.D.C.)
| | - Giuseppe Andò
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (G.T.); (G.A.)
| | - Francesco Burzotta
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Largo Agostino Gemelli, 1, 00168 Roma, Italy; (A.B.); (F.B.)
| | - Alberto Ranieri De Caterina
- Fondazione Toscana G. Monasterio, Ospedale del Cuore G., Pasquinucci, 54100 Massa, Italy; (G.B.); (S.B.); (A.R.D.C.)
| |
Collapse
|
12
|
Li M, Wu M, Pack J, Wu P, Yan P, De Man B, Wang A, Nieman K, Wang G. Coronary atherosclerotic plaque characterization with silicon-based photon-counting computed tomography (CT): A simulation-based feasibility study. Med Phys 2024. [PMID: 39321385 DOI: 10.1002/mp.17422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 08/23/2024] [Accepted: 08/30/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Recent photon-counting computed tomography (PCCT) development brings great opportunities for plaque characterization with much-improved spatial resolution and spectral imaging capability. While existing coronary plaque PCCT imaging results are based on CZT- or CdTe-materials detectors, deep-silicon photon-counting detectors offer unique performance characteristics and promise distinct imaging capabilities. PURPOSE This study aims to numerically investigate the feasibility of characterizing plaques with a deep-silicon PCCT scanner and to demonstrate its potential performance advantages over traditional CT scanners using energy-integrating detectors (EID). METHODS We conducted a systematic simulation study of a deep-silicon PCCT scanner using a newly developed digital plaque phantom with clinically relevant geometrical and chemical properties. Through qualitative and quantitative evaluations, this study investigates the effects of spatial resolution, noise, and motion artifacts on plaque imaging. RESULTS Noise-free simulations indicated that PCCT imaging could delineate the boundary of necrotic cores with a much finer resolution than EID-CT imaging, achieving a structural similarity index metric (SSIM) score of 0.970 and reducing the root mean squared error (RMSE) by two-thirds. Measuring necrotic core area errors were reduced from 91.5% to 24%, and fibrous cap thickness errors were reduced from 349.8% to 33.3%. In the presence of noise, the optimal reconstruction was achieved using 0.25 mm voxels and a soft reconstruction kernel, yielding the highest contrast-to-noise ratio (CNR) of 3.48 for necrotic core detection and the best image quality metrics among all choices. However, the ultrahigh resolution of PCCT increased sensitivity to motion artifacts, which could be mitigated by keeping residual motion amplitude below 0.4 mm. CONCLUSIONS The findings suggest that deep-silicon PCCT scanner can offer sufficient spatial resolution and tissue contrast for effective plaque characterization, potentially improving diagnostic accuracy in cardiovascular imaging, provided image noise and motion blur can be mitigated using advanced algorithms. This simulation study involves several simplifications, which may result in some idealized outcomes that do not directly translate to clinical practice. Further validation studies with physical scans are necessary and will be considered for future work.
Collapse
Affiliation(s)
- Mengzhou Li
- Biomedical Imaging Center, Center for Biotechnology and Interdisciplinary Research, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Mingye Wu
- GE HealthCare Technology & Innovation Center, Niskayuna, New York, USA
| | - Jed Pack
- GE HealthCare Technology & Innovation Center, Niskayuna, New York, USA
| | - Pengwei Wu
- GE HealthCare Technology & Innovation Center, Niskayuna, New York, USA
| | - Pingkun Yan
- Biomedical Imaging Center, Center for Biotechnology and Interdisciplinary Research, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Bruno De Man
- GE HealthCare Technology & Innovation Center, Niskayuna, New York, USA
| | - Adam Wang
- Department of Radiology, Stanford University, Stanford, California, USA
| | - Koen Nieman
- Department of Radiology, Stanford University, Stanford, California, USA
- Department of Medicine (Cardiovascular Medicine), Stanford University, Stanford, California, USA
| | - Ge Wang
- Biomedical Imaging Center, Center for Biotechnology and Interdisciplinary Research, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| |
Collapse
|
13
|
Reda Abdelaziz Morsy MM, Mensink FB, Los J, Damman P, van Royen N, Abdelhafez MAH, Mohamed HSE, Demitry SR, Ten Cate TJF, van Geuns RJ. Comparison of high-risk characteristics of non-culprit plaques in relation to plaque severity in acute coronary syndrome. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2024:S1553-8389(24)00666-3. [PMID: 39322479 DOI: 10.1016/j.carrev.2024.09.006] [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: 07/12/2024] [Revised: 08/12/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION Patients with acute coronary syndrome (ACS) have high event rates related to non-culprit (NC) lesions, therefore plaque composition of these lesions is of great interest. Although marginal atherosclerotic lesions were studied extensively, more significant lesions might have more high-risk characteristics. AIM To compare differences in high-risk lesion characteristics between significant versus non-stenotic NC plaques in ACS and the discrepancies with chronic coronary syndrome (CCS) patients. METHODS Non-culprit vessels of 26 ACS patients with 26 angiographically significant lesions and 37 patients (17 ACS and 20 CCS) with 48 non-stenotic lesions were investigated with intravascular ultrasound (IVUS) and near-infrared spectroscopy (NIRS). Overall, 74 segments of 30 mm length were analyzed in 1 mm intervals. External elastic lamina (EEM), plaque burden (PB), minimal luminal area (MLA), percent atheroma volume (PAV) and lipid core burden index maximum 4 mm (maxLCBI4mm) were determined for each segment. RESULTS Cardiovascular risk factors were similar in all groups. PB was higher and MLA smaller in significant non-culprit ACS lesions vs non-stenotic lesions: PB 73.5% (IQR 68.7-78.5) vs 59.2 (IQR 49.6-71.5), p = 0.003, MLA 3.0 mm2 (IQR 2.3-3.9) vs 4.0 mm2 (IQR 2.8-4.7). MaxLCBI4mm was similar 308.1 (±155.4) vs 287.8 (±165.7), p = 0.67. Among non-stenotic plaques, MaxLCBI4mm was comparable between ACS and CCS patients, 275.7 (±151.5) in CCS patients vs 287.8 (±165.7) in ACS patients, p = 0.79. CONCLUSION Although visually significant non-culprit lesions had a higher plaque burden compared to non-stenotic lesions, a significant relation between MaxLCBI4mm and hemodynamic significance of the plaques couldn't be established.
Collapse
Affiliation(s)
| | | | - Jonathan Los
- Radboud University medical center, the Netherlands
| | - Peter Damman
- Radboud University medical center, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
14
|
Girish MP, Gupta MD, Maehara A, Matsumura M, Bansal A, Kunal S, Batra V, Mohanty A, Qamar A, Mintz GS, Ali ZA, Yusuf J. OCT-based comparative evaluation of culprit lesion morphology in very young versus older adult patients with STEMI. ASIAINTERVENTION 2024; 10:177-185. [PMID: 39347107 PMCID: PMC11413641 DOI: 10.4244/aij-d-24-00013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/24/2024] [Indexed: 10/01/2024]
Abstract
Background The clinical and pathophysiological characteristics of coronary artery disease in very young adults are poorly described. Aims Using optical coherence tomography (OCT), we compared culprit lesion morphology in very young adult patients (≤35 years) versus older adult patients (>60 years) with ST-segment elevation myocardial infarction (STEMI). Methods Culprit lesion morphology was classified as plaque rupture, plaque erosion, or calcified nodule. Thrombus age was subclassified into acute (intraluminal thrombus with surface irregularity) or subacute (mostly mural thrombus with a smooth surface). Results A total of 61 patients who underwent thrombolysis within 24 hours from symptom onset were included, with 38 (59.7%) subjects ≤35 years and 23 (40.3%) subjects >60 years of age. As an underlying mechanism of STEMI thrombosis, plaque erosion was more common in very young patients (52.6% vs 21.7%; p=0.02) while plaque rupture was more common in elderly patients (65.2% vs 36.8%; p=0.03). Acute or subacute thrombus was identified in 68.9% (42/61) of patients, with red thrombus being more frequent in very young patients. In the entire patient cohort, acute thrombus was more frequent in plaque rupture compared with plaque erosion (62.0% vs 28.0%; p=0.01), whereas subacute thrombus was more common in plaque erosion versus plaque rupture (52.0% vs 10.3%; p=0.0008). Conclusions OCT showed that plaque erosion and plaque rupture were the most common underlying STEMI mechanisms in very young patients and older patients, respectively, and that subjects with plaque erosion had greater evidence of subacute thrombus.
Collapse
Affiliation(s)
- M P Girish
- Department of Cardiology, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, Delhi, India
| | - Mohit D Gupta
- Department of Cardiology, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, Delhi, India
| | - Akiko Maehara
- Division of Cardiology, Department of Medicine, Columbia University Medical Center/NewYork-Presbyterian Hospital, New York, NY, USA
- Cardiovascular Research Foundation, New York, NY, USA
| | | | - Ankit Bansal
- Department of Cardiology, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, Delhi, India
| | - Shekhar Kunal
- Department of Cardiology, ESIC Medical College and Hospital, Faridabad, India
| | - Vishal Batra
- Department of Cardiology, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, Delhi, India
| | - Arun Mohanty
- Department of Cardiology, Sir Ganga Ram Hospital, New Delhi, India
| | - Arman Qamar
- Section of Interventional Cardiology, NorthShore University Health System, Evanston, IL, USA
| | - Gary S Mintz
- Cardiovascular Research Foundation, New York, NY, USA
| | - Ziad A Ali
- Cardiovascular Research Foundation, New York, NY, USA
- Department of Cardiology, St. Francis Hospital, Roslyn, NY, USA
| | - Jamal Yusuf
- Department of Cardiology, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, Delhi, India
| |
Collapse
|
15
|
Koga JI, Umezu R, Kondo Y, Shirouzu T, Orkhonselenge N, Ueno H, Katsuki S, Matoba T, Nishimura Y, Kataoka M. Cyclophilin D induces necrotic core formation by mediating mitochondria-associated macrophage death in advanced atherosclerotic lesions. Atherosclerosis 2024; 396:118524. [PMID: 38972156 DOI: 10.1016/j.atherosclerosis.2024.118524] [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: 10/17/2023] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND AND AIMS In advanced atherosclerotic lesions, macrophage deaths result in necrotic core formation and plaque vulnerability. Cyclophilin D (CypD) is a mitochondria-specific cyclophilin involved in the process of cell death after organ ischemia-reperfusion. However, the role of CypD in atherosclerosis, especially in necrotic core formation, is unknown. Therefore, this experiment aims to clarify the role of CypD in necrotic core formation. METHODS To clarify the specific role of CypD, encoded by Ppif in mice, apolipoprotein-E/CypD-double knockout (Apoe-/-Ppif-/-) mice were generated. These mice were fed a high-fat diet containing 0.15 % cholesterol for 24 weeks to accelerate atherosclerotic lesion development. RESULTS Deletion of CypD decreased the necrotic core size, accompanied by a reduction of macrophage apoptosis compared to control Apoe-/- mice. In RAW264.7 cells, siRNA-mediated knockdown of CypD attenuated the release of cytochrome c from the mitochondria to the cytosol induced by endoplasmic reticulum stress inducer thapsigargin. In addition, necroptosis, induced by TNF-α and caspase inhibitor, was attenuated by knockdown of CypD. Ly-6Chigh inflammatory monocytes in peripheral blood leukocytes and mRNA expression of Il1b in the aorta were decreased by deletion of CypD. In contrast, siRNA-mediated knockdown of CypD did not significantly decrease Il1b nor Ccl2 mRNA expression in RAW264.7 cells treated with LPS and IFN-γ, suggesting that inhibition of inflammation in vivo is likely due to decreased cell death in the atherosclerotic lesions rather than a direct action of CypD deletion on the macrophage. CONCLUSIONS These results indicate that CypD induces macrophage death and mediates necrotic core formation in advanced atherosclerotic lesions. CypD could be a novel therapeutic target for treating atherosclerotic vascular diseases.
Collapse
Affiliation(s)
- Jun-Ichiro Koga
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Ryuta Umezu
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuki Kondo
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; The Department of Cardiovascular Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tomohiro Shirouzu
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Nasanbadrakh Orkhonselenge
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hiromichi Ueno
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shunsuke Katsuki
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Matoba
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yosuke Nishimura
- The Department of Cardiovascular Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masaharu Kataoka
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
16
|
Dai N, Tang X, Ling R, Zhou F, Chen S, Zhang L, Duan S, Pan W, Zhang J, Zhou D, Ge J. Prognostic implications of pre-transcatheter aortic valve replacement computed tomography-derived coronary plaque characteristics and stenosis severity. Eur Radiol 2024; 34:5923-5933. [PMID: 38308681 DOI: 10.1007/s00330-024-10633-7] [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: 09/10/2023] [Revised: 11/27/2023] [Accepted: 12/08/2023] [Indexed: 02/05/2024]
Abstract
OBJECTIVES The study aimed to investigate the prognostic value of pre-transcatheter aortic valve replacement (TAVR) computed tomography angiography (CTA) in assessing physiological stenosis severity (CTA-derived fractional flow reserve (CT-FFR)) and high-risk plaque characteristics (HRPC). MATERIALS AND METHODS Among TAVR patients who underwent pre-procedure CTA, the presence and number of HRPCs (minimum lumen area of < 4 mm2, plaque burden ≥ 70%, low-attenuating plaques, positive remodeling, napkin-ring sign, or spotty calcification) as well as CT-FFR were assessed. The risk of vessel-oriented composite outcome (VOCO, a composite of vessel-related ischemia-driven revascularization, vessel-related myocardial infarction, or cardiac death) was compared according to the number of HRPC and CT-FFR categories. RESULTS Four hundred and twenty-seven patients (68.4% were male) with 1072 vessels were included. Their mean age was 70.6 ± 10.6 years. Vessels with low CT-FFR (≤ 0.80) (41.7% vs. 15.8%, adjusted hazard ratio (HRadj) 1.96; 95% confidence interval (CI): 1.28-2.96; p = 0.001) or lesions with ≥ 3 HRPC (38.7% vs. 16.0%, HRadj 1.81; 95%CI 1.20-2.71; p = 0.005) demonstrated higher VOCO risk. In the CT-FFR (> 0.80) group, lesions with ≥ 3 HRPC showed a significantly higher risk of VOCO than those with < 3 HRPC (34.7% vs. 13.0%; HRadj 2.04; 95%CI 1.18-3.52; p = 0.011). However, this relative increase in risk was not observed in vessels with positive CT-FFR (≤ 0.80). CONCLUSIONS In TAVR candidates, both CT-FFR and the presence of ≥ 3 HRPC were associated with an increased risk of adverse clinical events. However, the value of HRPC differed with the CT-FFR category, with more incremental predictability among vessels with negative CT-FFR but not among vessels with positive CT-FFR. CLINICAL RELEVANCE STATEMENT In transcatheter aortic valve replacement (TAVR) candidates, pre-TAVR CTA provided the opportunity to assess coronary physiological stenosis severity and high-risk plaque characteristics, both of which are associated with worse clinical outcomes. KEY POINTS • The current study investigated the prognostic value of coronary physiology significance and plaque characteristics in transcatheter aortic valve replacement patients. • The combination of coronary plaque vulnerability and physiological significance showed improved accuracy in predicting clinical outcomes in transcatheter aortic valve replacement patients. • Pre-transcatheter aortic valve replacement CT can be a one-stop-shop tool for coronary assessments in clinical practice.
Collapse
Affiliation(s)
- Neng Dai
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Xianglin Tang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Runjianya Ling
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Fan Zhou
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Shasha Chen
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Lei Zhang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | | | - Wenzhi Pan
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Jiayin Zhang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, #85 Wujin Rd, Shanghai, 200080, China.
| | - Daxin Zhou
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Xuhui District, Shanghai, 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
17
|
Cesaro A, Acerbo V, Indolfi C, Filardi PP, Calabrò P. The clinical relevance of the reversal of coronary atherosclerotic plaque. Eur J Intern Med 2024:S0953-6205(24)00348-0. [PMID: 39164156 DOI: 10.1016/j.ejim.2024.08.010] [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: 03/25/2024] [Revised: 07/12/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of death globally despite advances in preventive therapies. Understanding of the initiation and progression of atherosclerosis, the interplay between lipoproteins, endothelial dysfunction, inflammation, and immune responses is critical to treating this disease. The development of vulnerable coronary plaques prone to thrombosis, can lead to acute coronary syndromes, for these reasons, the potential plaque stabilization and regression through pharmacological interventions, particularly lipid-lowering agents like statins and PCSK9 inhibitors is crucial. The imaging techniques such as intravascular ultrasound (IVUS), near-infrared spectroscopy (NIRS), and optical coherence tomography (OCT) play a key role in assessing plaque composition and guiding interventional therapeutic strategies. Clinical evidence supports the efficacy of intensive lipid-lowering therapy in inducing plaque regression, with studies demonstrating reductions in plaque volume and improvements in plaque morphology assessed by IVUS, OCT and NIRS. While pharmacological interventions show promise in promoting plaque regression and stabilization, their impact on long-term cardiovascular events requires further investigation. Multimodality imaging and comprehensive outcome trials are proposed as essential tools for elucidating the relationship between plaque modification and clinical benefit in coronary atherosclerosis. The stabilization or regression of atherosclerotic plaque might serve as the phenomenon linking the reduction in LDL-C levels to the decrease in cardiovascular events. Overall, this review emphasizes the ongoing efforts to advance our understanding of ASCVD pathophysiology and optimize therapeutic approaches for improving patient outcomes.
Collapse
Affiliation(s)
- Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy
| | - Vincenzo Acerbo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | | | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; Division of Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy.
| |
Collapse
|
18
|
HashemizadehKolowri S, Akcicek EY, Akcicek H, Ma X, Ferguson MS, Balu N, Hatsukami TS, Yuan C. Efficient and Accurate 3D Thickness Measurement in Vessel Wall Imaging: Overcoming Limitations of 2D Approaches Using the Laplacian Method. J Cardiovasc Dev Dis 2024; 11:249. [PMID: 39195157 DOI: 10.3390/jcdd11080249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 08/29/2024] Open
Abstract
The clinical significance of measuring vessel wall thickness is widely acknowledged. Recent advancements have enabled high-resolution 3D scans of arteries and precise segmentation of their lumens and outer walls; however, most existing methods for assessing vessel wall thickness are 2D. Despite being valuable, reproducibility and accuracy of 2D techniques depend on the extracted 2D slices. Additionally, these methods fail to fully account for variations in wall thickness in all dimensions. Furthermore, most existing approaches are difficult to be extended into 3D and their measurements lack spatial localization and are primarily confined to lumen boundaries. We advocate for a shift in perspective towards recognizing vessel wall thickness measurement as inherently a 3D challenge and propose adapting the Laplacian method as an outstanding alternative. The Laplacian method is implemented using convolutions, ensuring its efficient and rapid execution on deep learning platforms. Experiments using digital phantoms and vessel wall imaging data are conducted to showcase the accuracy, reproducibility, and localization capabilities of the proposed approach. The proposed method produce consistent outcomes that remain independent of centerlines and 2D slices. Notably, this approach is applicable in both 2D and 3D scenarios. It allows for voxel-wise quantification of wall thickness, enabling precise identification of wall volumes exhibiting abnormal wall thickness. Our research highlights the urgency of transitioning to 3D methodologies for vessel wall thickness measurement. Such a transition not only acknowledges the intricate spatial variations of vessel walls, but also opens doors to more accurate, localized, and insightful diagnostic insights.
Collapse
Affiliation(s)
| | - Ebru Yaman Akcicek
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84108, USA
| | - Halit Akcicek
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84108, USA
| | - Xiaodong Ma
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84108, USA
| | - Marina S Ferguson
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Niranjan Balu
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Thomas S Hatsukami
- Department of Surgery, Division of Vascular Surgery, University of Washington, Seattle, WA 98195, USA
| | - Chun Yuan
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84108, USA
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
19
|
Sirignano P, Margheritini C, Mansour W, Aloisi F, Setacci C, Speziale F, Stabile E, Taurino M. Sex as a Predictor of Outcomes for Symptomatic Carotid Stenosis: A Comparative Analysis between CAS and CEA. J Pers Med 2024; 14:830. [PMID: 39202021 PMCID: PMC11355406 DOI: 10.3390/jpm14080830] [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: 05/16/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
PURPOSE Reporting gender-related outcomes for symptomatic carotid lesion revascularization after both endarterectomy (CEA) and carotid artery stenting (CAS) procedures in an unselected group of patients treated by Italian Vascular Specialists. MATERIAL AND METHODS A retrospective study was conducted on patients presenting with recently symptomatic carotid stenosis treated by CAS and by CEA. The primary endpoint was the 30 days any stroke occurrence rate; secondary endpoints were technical success, occurrence of transient ischemic attack (TIA), acute myocardial infarction (AMI) and death. Demographic, clinical and procedural data were all noted in order to identify the outcome's determining factor. RESULTS A total of 265 patients (193 males and 72 females) were enrolled, and of these 134 (50.5%) underwent CEA and 131 CAS (49.5%). At 30 days, the overall new stroke rate was 3.4% (one fatal), and no TIA, AMI or deaths were observed. Among strokes, seven major and two minor strokes were reported, with six after CEA and three after CAS (p = 0.32; OR: 2; CI95%: 0.48-8.17). The timing of revascularization has been found to be slightly associated with new stroke occurrence: seven out nine strokes were observed in patients treated within 14 days from symptom onset (5.5% vs. 1.4%; p = 0.08, OR: 3.8, CI95%: 0.77-18.56). Lastly, female patients presented a significantly higher risk of post-operative stroke compared to male patients: 6.9% vs. 2.1% (p: 0.05; OR: 3.52; CI95%: 0.91-13.52). CONCLUSIONS Our experience seems to suggest that both CEA and CAS provide safe and effective results in treating patients presenting with symptomatic carotid stenosis. Regardless of the type of revascularization, female sex is an independent risk factor for stroke recurrence after treatment.
Collapse
Affiliation(s)
- Pasqualino Sirignano
- Vascular and Endovascular Surgery Unit, Sant’Andrea Hospital of Rome, Department of General and Specialistic Surgery, “Sapienza” University of Rome, 00189 Rome, Italy
| | - Costanza Margheritini
- Vascular and Endovascular Surgery Unit, Sant’Andrea Hospital of Rome, Department of Molecular and Clinical Medicine, “Sapienza” University of Rome, 00189 Rome, Italy
| | - Wassim Mansour
- Vascular and Endovascular Surgery Unit, Policlinico Umberto I Hospital of Rome, Department of General and Specialistic Surgery, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Francesco Aloisi
- Vascular and Endovascular Surgery Unit, Sant’Andrea Hospital of Rome, Department of Molecular and Clinical Medicine, “Sapienza” University of Rome, 00189 Rome, Italy
| | - Carlo Setacci
- Vascular and Endovascuar Surgery Unit, “Le Scotte” Hospital of Siena, Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Francesco Speziale
- Vascular and Endovascular Surgery Unit, Policlinico Umberto I Hospital of Rome, Department of General and Specialistic Surgery, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Eugenio Stabile
- Division of Cardiology, Cardiovascular Department, San Carlo Regional Hospital, “Federico II” University of Naples, 85100 Potenza, Italy
| | - Maurizio Taurino
- Vascular and Endovascular Surgery Unit, Sant’Andrea Hospital of Rome, Department of Molecular and Clinical Medicine, “Sapienza” University of Rome, 00189 Rome, Italy
| | | |
Collapse
|
20
|
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
|
21
|
Buckler AJ, Abbara S, Budoff MJ, Carr JJ, De Cecco CN, DeMarco JK, Ferencik M, Figtree GA, Ikuta I, Kolossváry M, Konrad M, Lal BK, Marques H, Moss AJ, Obuchowski NA, van Beek EJR, Virmani R, Williams MC, Saba L, Joseph Schoepf U. Special Report on the Consensus QIBA Profile for Objective Analytical Validation of Non-calcified and High-risk Plaque and Other Biomarkers using Computed Tomography Angiography. Acad Radiol 2024:S1076-6332(24)00448-3. [PMID: 39060206 DOI: 10.1016/j.acra.2024.07.014] [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: 05/07/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024]
Abstract
RATIONALE AND OBJECTIVES Evidence is building in support of the clinical utility of atherosclerotic plaque imaging by computed tomography angiography (CTA). There is increasing organized activity to embrace non-calcified plaque (NCP) as a formally defined biomarker for clinical trials, and high-risk plaque (HRP) for clinical care, as the most relevant measures for the field to advance and worthy of community efforts to validate. Yet the ability to assess the quantitative performance of any given specific solution to make these measurements or classifications is not available. Vendors use differing definitions, assessment metrics, and validation data sets to describe their offerings without clinician users having the capability to make objective assessments of accuracy and precision and how this affects diagnostic confidence. MATERIALS AND METHODS The QIBA Profile for Atherosclerosis Biomarkers by CTA was created by the Quantitative Imaging Biomarkers Alliance (QIBA) to improve objectivity and decrease the variability of noninvasive plaque phenotyping. The Profile provides claims on the accuracy and precision of plaque measures individually and when combined. RESULTS Individual plaque morphology measurements are evaluated in terms of bias (accuracy), slope (consistency of the bias across the measurement range, needed for measurements of change), and variability. The multiparametric plaque stability phenotype is evaluated in terms of agreement with expert pathologists. The Profile is intended for a broad audience, including those engaged in discovery science, clinical trials, and patient care. CONCLUSION This report provides a rationale and overview of the Profile claims and how to comply with the Profile in research and clinical practice. SUMMARY STATEMENT This article summarizes objective means to validate the analytical performance of non-calcified plaque (NCP), other emerging plaque morphology measurements, and multiparametric histology-defined high-risk plaque (HRP), as outlined in the QIBA Profile for Atherosclerosis Biomarkers by CTA.
Collapse
Affiliation(s)
| | | | - Matthew J Budoff
- Department of Medicine, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA (M.J.B.)
| | - John Jeffrey Carr
- Vanderbilt University Medical Center, Nashville, Tennessee, USA (J.J.C.)
| | | | - J Kevin DeMarco
- Walter Reed National Military Medical Center and Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA (J.K.D.)
| | - Maros Ferencik
- Oregon Health & Science University, Portland, Oregon, USA (M.F.)
| | - Gemma A Figtree
- Department of Cardiology, Royal North Shore Hospital, St Leonards, NSW, Australia (G.A.F.); Cardiovascular Discovery Group, Kolling Institute of Medical Research, St Leonards, Australia (G.A.F.); Faculty of Medicine & Health, University of Sydney, Camperdown, Royal North Shore Hospital, St Leonards, NSW, Australia (G.A.F.)
| | - Ichiro Ikuta
- Mayo Clinic Arizona, Phoenix, Arizona, USA (I.I.)
| | - Márton Kolossváry
- Gottsegen National Cardiovascular Center, Budapest, Hungary (M.K.); Physiological Controls Research Center, University Research and Innovation Center, Óbuda University, Budapest, Hungary (M.K.)
| | - Mathis Konrad
- Department of Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany (M.K.)
| | - Brajesh K Lal
- Department of Vascular Surgery, University of Maryland, Baltimore, Maryland, USA (B.K.L.); Vascular Service, VA Medical Center, Baltimore, Maryland, USA (B.K.L.)
| | - Hugo Marques
- Hospital da Luz, Imaging Department - Católica Medical School, Lisboa, Portugal (H.M.)
| | - Alastair J Moss
- Department of Cardiovascular Sciences and National Institute for Health Research Leicester Biomedical Research Centre, University of Leicester, Leicester, UK (A.J.M.)
| | - Nancy A Obuchowski
- Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio, USA (N.A.O.)
| | | | - Renu Virmani
- CV Path Institute, Gaithersburg, Maryland, USA (R.V.)
| | - Michelle C Williams
- Emory University, Atlanta, Georgia, USA (C.N.D.C., M.C.W.); Centre for Cardiovascular Science, University of Edinburgh, Scotland (M.C.W.)
| | - Luca Saba
- University of Cagliari, Sardinia, Italy (L.S.)
| | - U Joseph Schoepf
- Medical University of South Carolina, Charleston, South Carolina, USA (U.J.S.)
| |
Collapse
|
22
|
Onea HL, Olinic M, Lazar FL, Homorodean C, Ober MC, Spinu M, Achim A, Tataru DA, Olinic DM. A Review Paper on Optical Coherence Tomography Evaluation of Coronary Calcification Pattern: Is It Relevant Today? J Cardiovasc Dev Dis 2024; 11:231. [PMID: 39195139 DOI: 10.3390/jcdd11080231] [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: 06/17/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
The process of coronary calcification represents one of the numerous pathophysiological mechanisms involved in the atherosclerosis continuum. Optical coherence tomography (OCT) represents an ideal imaging modality to assess plaque components, especially calcium. Different calcification patterns have been contemporarily described in both early stages and advanced atherosclerosis. Microcalcifications and spotty calcifications correlate positively with macrophage burden and inflammatory markers and are more frequently found in the superficial layers of ruptured plaques in acute coronary syndrome patients. More compact, extensive calcification may reflect a later stage of the disease and was traditionally associated with plaque stability. Nevertheless, a small number of culprit coronary lesions demonstrates the presence of dense calcified plaques. The purpose of the current paper is to review the most recent OCT data on coronary calcification and the interrelation between calcification pattern and plaque vulnerability. How different calcified plaques influence treatment strategies and associated prognostic implications is of great interest.
Collapse
Affiliation(s)
- Horea-Laurentiu Onea
- Department of Internal Medicine, Medical Clinic Number 1, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- County Clinical Emergency Hospital Sibiu, 550024 Sibiu, Romania
| | - Maria Olinic
- Department of Internal Medicine, Medical Clinic Number 1, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- Second Cardiology Department, County Clinical Emergency Hospital Cluj-Napoca, 400347 Cluj-Napoca, Romania
| | - Florin-Leontin Lazar
- Department of Internal Medicine, Medical Clinic Number 1, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- County Clinical Emergency Hospital Sibiu, 550024 Sibiu, Romania
| | - Calin Homorodean
- Department of Internal Medicine, Medical Clinic Number 1, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- Second Cardiology Department, County Clinical Emergency Hospital Cluj-Napoca, 400347 Cluj-Napoca, Romania
| | - Mihai Claudiu Ober
- Second Cardiology Department, County Clinical Emergency Hospital Cluj-Napoca, 400347 Cluj-Napoca, Romania
| | - Mihail Spinu
- Department of Internal Medicine, Medical Clinic Number 1, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- Second Cardiology Department, County Clinical Emergency Hospital Cluj-Napoca, 400347 Cluj-Napoca, Romania
| | - Alexandru Achim
- Niculae Stancioiu Heart Institute Cluj-Napoca, 400001 Cluj-Napoca, Romania
| | - Dan Alexandru Tataru
- Department of Internal Medicine, Medical Clinic Number 1, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- Second Cardiology Department, County Clinical Emergency Hospital Cluj-Napoca, 400347 Cluj-Napoca, Romania
| | - Dan Mircea Olinic
- Department of Internal Medicine, Medical Clinic Number 1, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- Second Cardiology Department, County Clinical Emergency Hospital Cluj-Napoca, 400347 Cluj-Napoca, Romania
| |
Collapse
|
23
|
Lu F, Lin Y, Zhou J, Chen Z, Liu Y, Zhong M, Wang L. Obesity and the obesity paradox in abdominal aortic aneurysm. Front Endocrinol (Lausanne) 2024; 15:1410369. [PMID: 39055063 PMCID: PMC11269098 DOI: 10.3389/fendo.2024.1410369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Obesity, characterized by its complexity and heterogeneity, has emerged as a significant public health concern. Its association with increased incidence and mortality of cardiovascular diseases stems not only from its complications and comorbidities but also from the endocrine effects of adipose tissue. Abdominal aortic aneurysm (AAA), a chronic inflammatory condition, has been closely linked to obesity. Intriguingly, mild obesity appears to confer a protective effect against AAA mortality, whereas severe obesity and being underweight do not, giving rise to the concept of the "obesity paradox". This review aims to provide an overview of obesity and its paradoxical relationship with AAA, elucidate its underlying mechanisms, and discuss the importance of preoperative weight loss in severely obese patients with AAA.
Collapse
Affiliation(s)
- Feng Lu
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Ganzhou Key Laboratory of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yong Lin
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Ganzhou Key Laboratory of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jianshun Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Ganzhou Key Laboratory of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhen Chen
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Ganzhou Key Laboratory of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yingying Liu
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Ganzhou Key Laboratory of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Maolin Zhong
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Ganzhou Key Laboratory of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Lifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Ganzhou Key Laboratory of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
24
|
Jawaid MM, Narejo S, Riaz F, Reyes-Aldasoro CC, Slabaugh G, Brown J. Non-calcified plaque-based coronary stenosis grading in contrast enhanced CT. Med Eng Phys 2024; 129:104182. [PMID: 38906576 DOI: 10.1016/j.medengphy.2024.104182] [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: 02/13/2024] [Revised: 04/08/2024] [Accepted: 05/17/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND The high mortality rate associated with coronary heart disease has led to state-of-the-art non-invasive methods for cardiac diagnosis including computed tomography and magnetic resonance imaging. However, stenosis computation and clinical assessment of non-calcified plaques has been very challenging due to their ambiguous intensity response in CT i.e. a significant overlap with surrounding muscle tissues and blood. Accordingly, this research presents an approach for computation of coronary stenosis by investigating cross-sectional lumen behaviour along the length of 3D coronary segments. METHODS Non-calcified plaques are characterized by comparatively lower-intensity values with respect to the surrounding. Accordingly, segment-wise orthogonal volume was reconstructed in 3D space using the segmented coronary tree. Subsequently, the cross sectional volumetric data was investigated using proposed CNN-based plaque quantification model and subsequent stenosis grading in clinical context was performed. In the last step, plaque-affected orthogonal volume was further investigated by comparing vessel-wall thickness and lumen area obstruction w.r.t. expert-based annotations to validate the stenosis grading performance of model. RESULTS The experimental data consists of clinical CT images obtained from the Rotterdam CT repository leading to 600 coronary segments and subsequent 15786 cross-sectional images. According to the results, the proposed method quantified coronary vessel stenosis i.e. severity of the non-calcified plaque with an overall accuracy of 83%. Moreover, for individual grading, the proposed model show promising results with accuracy equal to 86%, 90% and 79% respectively for severe, moderate and mild stenosis. The stenosis grading performance of the proposed model was further validated by performing lumen-area versus wall-thickness analysis as per annotations of manual experts. The statistical results for lumen area analysis precisely correlates with the quantification performance of the model with a mean deviation of 5% only. CONCLUSION The overall results demonstrates capability of the proposed model to grade the vessel stenosis with reasonable accuracy and precision equivalent to human experts.
Collapse
Affiliation(s)
| | - Sanam Narejo
- Mehran University of Engineering and Technology, Jamshoro, Pakistan.
| | | | | | | | | |
Collapse
|
25
|
Wu J, Zou Y, Meng X, Fan Z, van der Geest R, Cui F, Li J, Zhang T, Zhang F. Increased incidence of napkin-ring sign plaques on cervicocerebral computed tomography angiography associated with the risk of acute ischemic stroke occurrence. Eur Radiol 2024; 34:4438-4447. [PMID: 38001250 DOI: 10.1007/s00330-023-10404-w] [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: 04/05/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 11/26/2023]
Abstract
OBJECTIVES Carotid atherosclerosis plays an essential role in the occurrence of ischemic stroke. This study aimed to investigate whether a larger burden of napkin-ring sign (NRS) plaques on cervicocerebral computed tomography angiography (CTA) increased the risk of acute ischemic stroke (AIS). METHODS This retrospective, single-center, cross-sectional study enrolled patients with NRS plaques identified in the subclavian arteries, brachiocephalic trunk, carotid arterial system, and vertebrobasilar circulation on contrast-enhanced cervicocerebral CTA. Patients were divided into AIS and non-AIS groups based on imaging within 12 h of symptom onset. Univariate and multivariate logistic regression analyses were performed to determine the risk factor of AIS occurrence. RESULTS A total of 202 patients (66.72 years ± 8.97, 157 men) were evaluated. Plaques with NRS in each subject of the AIS group (N = 98) were significantly more prevalent than that in the control group (N = 104) (1.96 ± 1.17 vs 1.41 ± 0.62). In the AIS group, there were substantially more NRS plaques on the ipsilateral side than contralateral side (1.55 ± 0.90 vs. 0.41 ± 0.66). NRS located on the ipsilateral side of the AIS showed an area under the receiver curve (AUC) of 0.86 to identify ischemic stroke. NRS plaque amounts were an independent risk factor for AIS occurrence (odds ratio, 1.86) after adjusting for other factors. CONCLUSIONS Increased incidence of napkin-ring sign plaques on cervicocerebral CTA was positively associated with AIS occurrence, which could aid in detecting asymptomatic atherosclerotic patients at high risk of AIS in routine screening or emergency settings. CLINICAL RELEVANCE STATEMENT Napkin-ring sign plaque provides an important imaging target for estimating acute ischemic stroke risk and identifying high-risk patients in routine screening or emergency settings, so that timely anti-atherosclerotic therapy can be used for prevention. KEY POINTS • This cross-sectional study investigated the association between high-risk carotid artery plaques and acute ischemic stroke. • Increased incidence of napkin-ring sign plaques on cervicocerebral computed tomography angiography is positively associated with acute ischemic stroke occurrence. • Napkin-ring signs help identify risky patients prone to acute ischemic stroke to facilitate prevention.
Collapse
Affiliation(s)
- Jingping Wu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Radiology, Hainan Hospital of PLA General Hospital, Sanya, China
| | - Ying Zou
- Department of Radiology, Hainan Hospital of PLA General Hospital, Sanya, China
| | - Xiao Meng
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Zhaoyang Fan
- Department of Radiology, University of Southern California, Los Angeles, CA, USA
| | - Rob van der Geest
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Fang Cui
- Department of Neurology, Hainan Hospital of PLA General Hospital, Sanya, China
| | - Jianyong Li
- Department of Neurology, Hainan Hospital of PLA General Hospital, Sanya, China
| | - Tengyuan Zhang
- Department of Neurology, Hainan Hospital of PLA General Hospital, Sanya, China
| | - Fan Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
- Department of Radiology, Hainan Hospital of PLA General Hospital, Sanya, China.
| |
Collapse
|
26
|
Jiao J, Hu B, Mou T, Li Q, Tian Y, Zhang N, Zhang Y, Yun M, Nan N, Tian J, Yu W, Mi H, Dong W, Song X. Translocator Protein 18 kDa Tracer 18F-FDPA PET/CTA Imaging for the Evaluation of Inflammation in Vulnerable Plaques. Mol Pharm 2024; 21:3623-3633. [PMID: 38819959 DOI: 10.1021/acs.molpharmaceut.4c00344] [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: 06/02/2024]
Abstract
Inflammation induced by activated macrophages within vulnerable atherosclerotic plaques (VAPs) constitutes a significant risk factor for plaque rupture. Translocator protein (TSPO) is highly expressed in activated macrophages. This study investigated the effectiveness of TSPO radiotracers, 18F-FDPA, in detecting VAPs and quantifying plaque inflammation in rabbits. 18 New Zealand rabbits were divided into 3 groups: sham group A, VAP model group B, and evolocumab treatment group C. 18F-FDPA PET/CTA imaging was performed at 12, 16, and 24 weeks in all groups. Optical coherence tomography (OCT) was performed on the abdominal aorta at 24 weeks. The VAP was defined through OCT images, and ex vivo aorta PET imaging was also performed at 24 weeks. The SUVmax and SUVmean of 18F-FDPA were measured on the target organ, and the target-to-background ratio (TBRmax) was calculated as SUVmax/SUVblood pool. The arterial sections of the isolated abdominal aorta were analyzed by HE staining, CD68 and TSPO immunofluorescence staining, and TSPO Western blot. The results showed that at 24 weeks, the plaque TBRmax of 18F-FDPA in group B was significantly higher than in groups A and C. Immunofluorescence staining of CD68 and TSPO, as well as Western blot, confirmed the increased expression of macrophages and TSPO in the corresponding regions of group B. HE staining revealed an increased presence of the lipid core, multiple foam cells, and inflammatory cell infiltration in the area with high 18F-FDPA uptake. This indicates a correlation between 18F-FDPA uptake, inflammation severity, and VAPs. The TSPO-targeted tracer 18F-FDPA shows specific uptake in macrophage-rich regions of atherosclerotic plaques, making it a valuable tool for assessing inflammation in VAPs.
Collapse
Affiliation(s)
- Jian Jiao
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Biao Hu
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Tiantian Mou
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Quan Li
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yi Tian
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Nan Zhang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ying Zhang
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mingkai Yun
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Nan Nan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Jing Tian
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Wei Yu
- Department of Pathology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Hongzhi Mi
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Wei Dong
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
27
|
Wolny R, Geers J, Grodecki K, Kwiecinski J, Williams MC, Slomka PJ, Hasific S, Lin AK, Dey D. Noninvasive Atherosclerotic Phenotyping: The Next Frontier into Understanding the Pathobiology of Coronary Artery Disease. Curr Atheroscler Rep 2024; 26:305-315. [PMID: 38727963 DOI: 10.1007/s11883-024-01205-7] [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: 04/25/2024] [Indexed: 06/22/2024]
Abstract
PURPOSE OF REVIEW Despite recent advances, coronary artery disease remains one of the leading causes of mortality worldwide. Noninvasive imaging allows atherosclerotic phenotyping by measurement of plaque burden, morphology, activity and inflammation, which has the potential to refine patient risk stratification and guide personalized therapy. This review describes the current and emerging roles of advanced noninvasive cardiovascular imaging methods for the assessment of coronary artery disease. RECENT FINDINGS Cardiac computed tomography enables comprehensive, noninvasive imaging of the coronary vasculature, and is used to assess luminal stenoses, coronary calcifications, and distinct adverse plaque characteristics, helping to identify patients prone to future events. Novel software tools, implementing artificial intelligence solutions, can automatically quantify and characterize atherosclerotic plaque from standard computed tomography datasets. These quantitative imaging biomarkers have been shown to improve patient risk stratification beyond clinical risk scores and current clinical interpretation of cardiac computed tomography. In addition, noninvasive molecular imaging in higher risk patients can be used to assess plaque activity and plaque thrombosis. Noninvasive imaging allows unique insight into the burden, morphology and activity of atherosclerotic coronary plaques. Such phenotyping of atherosclerosis can potentially improve individual patient risk prediction, and in the near future has the potential for clinical implementation.
Collapse
Affiliation(s)
- Rafal Wolny
- Department of Interventional Cardiology and Angiology, National Institute of Cardiology, Warsaw, Poland
| | - Jolien Geers
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA
- Department of Cardiology, Centrum Voor Hart- en Vaatziekten (CHVZ), Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Kajetan Grodecki
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Jacek Kwiecinski
- Department of Interventional Cardiology and Angiology, National Institute of Cardiology, Warsaw, Poland
| | - Michelle C Williams
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Piotr J Slomka
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA
| | - Selma Hasific
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Andrew K Lin
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA
- Monash Cardiovascular Research Centre, Victorian Heart Institute, Monash University and MonashHeart, Monash Health, Melbourne, VIC, Australia
| | - Damini Dey
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA.
| |
Collapse
|
28
|
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
|
29
|
Li C, Deng C, Shi B, Zhao R. Thin-cap fibroatheroma in acute coronary syndrome: Implication for intravascular imaging assessment. Int J Cardiol 2024; 405:131965. [PMID: 38492863 DOI: 10.1016/j.ijcard.2024.131965] [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: 12/17/2023] [Revised: 02/15/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
Acute coronary syndrome (ACS), a significant cardiovascular disease threat, has garnered increased focus concerning its etiological mechanisms. Thin-cap fibroatheroma (TCFA) are central to ACS pathogenesis, characterized by lipid-rich plaques, profuse foam cells, cholesterol crystals, and fragile fibrous caps predisposed to rupture. While TCFAs may be latent and asymptomatic, their pivotal role in ACS risk is undeniable. High-resolution imaging techniques like Optical coherence tomography (OCT) and Intravascular ultrasound (IVUS) are instrumental for effective TCFA detection. Therapeutic strategies encompass pharmacological and interventional measures, including antiplatelet agents, statins, and Percutaneous Coronary Intervention (PCI), aiding in plaque stabilization, inflammation reduction, and rupture risk mitigation. Despite the strong correlation between TCFAs and adverse prognoses in ACS patients, early detection and rigorous treatment significantly enhance patient prognosis and diminish cardiovascular events. This review aims to encapsulate recent advancements in TCFA research within ACS, covering formation mechanisms, clinical manifestations, and prognostic implications.
Collapse
Affiliation(s)
- Chaozhong Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chancui Deng
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
30
|
Hou S, Zhan Z, Fan J, Li M, Chen S, Zhang Y, Long Y, Pan W, Zhang X, Zhou D, Ge J. Association of in situ thrombus within the patent foramen ovale and patients with migraine: A prospective cohort study. Heliyon 2024; 10:e32105. [PMID: 38882380 PMCID: PMC11176847 DOI: 10.1016/j.heliyon.2024.e32105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Background Patent foramen ovale (PFO) is associated with migraine; however, the mechanism of PFO-associated migraine is not well known; additionally, percutaneous closure is controversial. This study aimed to investigate in situ thrombi within the PFO and explore the possible predictors of the effectiveness of PFO closure in migraineurs. Methods This prospective cohort study included 48 asymptomatic patients and 92 migraineurs with PFO. Optical coherence tomography (OCT) was used to evaluate the PFO microstructure. Only migraineurs underwent percutaneous closure. Migraineurs were divided into two cohorts based on the presence of a thrombus within the PFO. The symptoms were assessed at the 12-month follow-up visit. Predictors were evaluated employing multivariate logistic regression and receiver operating characteristic curve analyses. Results In situ thrombi within PFO were identified in 69 migraineurs and in two asymptomatic patients (76.7 % vs. 4.3 %; P < 0.001). Additionally, endocardial irregularity, discontinuity, low signal, and spasm were found in 59 (65.6 %), 15 (16.7 %), 13 (14.4 %), and six (6.7 %) patients, respectively, in the migraine group. In situ thrombus was associated with migraine risk (OR 49.03; 95%CI 8.52-282.18; P < 0.001). At the 12-month follow-up of the migraineur cohort, the primary endpoint, a 50 % reduction in migraine frequency after closure (with or without thrombus in PFO) was met (85.3 % vs. 25.0 %; P < 0.001). In situ thrombus was associated with migraine relief (OR 6.75; 95%CI 1.28-35.56; P = 0.024). Conclusions In situ thrombus and abnormal endocardium within PFOs were common in migraineurs, and in situ thrombus was a risk factor for migraine. Percutaneous closure was more effective in migraineurs with thrombi within the PFO. OCT imaging improved the understanding of pathogenic PFOs and may be helpful in selecting suitable migraineurs for PFO closure.
Collapse
Affiliation(s)
- Shiqiang Hou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Zhi Zhan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Jianing Fan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Mingfei Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Shasha Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yuan Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yuliang Long
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Wenzhi Pan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xiaochun Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Daxin Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| |
Collapse
|
31
|
Del Val D, Berta B, Roleder T, Malinowski K, Bastante T, Hermanides RS, Wojakowski W, Fabris E, Cuesta J, De Luca G, Rivero F, Alfonso F, Kedhi E. Vulnerable plaque features and adverse events in patients with diabetes mellitus: a post hoc analysis of the COMBINE OCT-FFR trial. EUROINTERVENTION 2024; 20:e707-e717. [PMID: 38840580 PMCID: PMC11148652 DOI: 10.4244/eij-d-23-00628] [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: 08/02/2023] [Accepted: 04/02/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Thin-cap fibroatheroma (TCFA) lesions are associated with a high risk of future major adverse cardiovascular events. However, the impact of other optical coherence tomography-detected vulnerability features (OCT-VFs) and their interplay with TCFA in predicting adverse events remains unknown. AIMS We aimed to evaluate the individual as well as the combined prognostic impact of OCT-VFs in predicting the incidence of the lesion-oriented composite endpoint (LOCE) in non-ischaemic lesions in patients with diabetes mellitus (DM). METHODS COMBINE OCT-FFR (ClinicalTrials.gov: NCT02989740) was a prospective, double-blind, international, natural history study that included DM patients with ≥1 non-culprit lesions with a fractional flow reserve>0.80 undergoing systematic OCT assessment. OCT-VFs included the following: TCFA, reduced minimal lumen area (r-MLA), healed plaque (HP), and complicated plaque (CP). The primary endpoint, LOCE - a composite of cardiac mortality, target vessel myocardial infarction, or clinically driven target lesion revascularisation up to 5 years - was analysed according to the presence of these OCT-VFs, both individually and in combination. RESULTS TCFA, r-MLA, HP and CP were identified in 98 (25.3%), 190 (49.0%), 87 (22.4%), and 116 (29.9%) patients, respectively. The primary endpoint rate increased progressively from 6.3% to 55.6% (hazard ratio 15.2, 95% confidence interval: 4.53-51.0; p<0.001) in patients without OCT-VFs as compared to patients with concomitant HP, r-MLA, CP, and TCFA. The coexistence of TCFA with other OCT-VFs resulted in an increased risk of the LOCE at 5 years. CONCLUSIONS In DM patients with non-ischaemic lesions, TCFA was the strongest predictor of future LOCE events. However, lesions that present additional OCT-VFs are associated with a higher risk of adverse events than OCT-detected TCFA alone. Further randomised studies are warranted to confirm these findings and their potential clinical implications.
Collapse
Affiliation(s)
- David Del Val
- Cardiology Department, Hospital Universitario de La Princesa, Madrid, Spain
- CIBERCV; Instituto de Investigación Sanitaria, IIS-IP, Hospital Universitario de La Princesa, Madrid, Spain
| | - Balazs Berta
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Isala Hartcentrum, Zwolle, the Netherlands
| | - Tomasz Roleder
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Katowice, Poland
- Department of Cardiology, Wroclaw Medical University, Wroclaw, Poland
| | - Krzysztof Malinowski
- Department of Bioinformatics and Telemedicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Teresa Bastante
- Cardiology Department, Hospital Universitario de La Princesa, Madrid, Spain
- CIBERCV; Instituto de Investigación Sanitaria, IIS-IP, Hospital Universitario de La Princesa, Madrid, Spain
| | | | - Wojciech Wojakowski
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Katowice, Poland
| | - Enrico Fabris
- Cardiovascular Department, University of Trieste, Trieste, Italy
| | - Javier Cuesta
- Cardiology Department, Hospital Universitario de La Princesa, Madrid, Spain
- CIBERCV; Instituto de Investigación Sanitaria, IIS-IP, Hospital Universitario de La Princesa, Madrid, Spain
| | - Giuseppe De Luca
- Division of Cardiology, AOU "Policlinico G. Martino", Messina, Italy and Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
- Division of Cardiology, IRCCS Hospital Galeazzi-Sant'Ambrogio, Milan, Italy
| | - Fernando Rivero
- Cardiology Department, Hospital Universitario de La Princesa, Madrid, Spain
- CIBERCV; Instituto de Investigación Sanitaria, IIS-IP, Hospital Universitario de La Princesa, Madrid, Spain
| | - Fernando Alfonso
- Cardiology Department, Hospital Universitario de La Princesa, Madrid, Spain
- CIBERCV; Instituto de Investigación Sanitaria, IIS-IP, Hospital Universitario de La Princesa, Madrid, Spain
| | - Elvin Kedhi
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Katowice, Poland
- Department of Interventional Cardiology, Royal VIctoria Hospital, McGill University Health Center, McGill University, Montreal, Canada
| |
Collapse
|
32
|
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
|
33
|
Mansouri P, Nematipour E, Rajablou N, Ghorashi SM, Azari S, Omidi N. Left anterior descending coronary artery-left circumflex coronary artery bifurcation angle and severity of coronary artery disease; is there any correlation? A cross-sectional study. Health Sci Rep 2024; 7:e2182. [PMID: 38868537 PMCID: PMC11168269 DOI: 10.1002/hsr2.2182] [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: 12/10/2023] [Revised: 04/05/2024] [Accepted: 05/16/2024] [Indexed: 06/14/2024] Open
Abstract
Background and Aims The aim of this study is to evaluate the association of coronary computed tomography angiography derived (CCTA) plaque characteristics and the left anterior descending coronary artery (LAD) and left circumflex coronary artery (LCX) bifurcation angle with severity of coronary artery disease (CAD). Methods All the stable patients with suspected CAD who underwent CCTA between January to December 2021 were included. Correlation between CCTA-derived aggregated plaque volume (APV), LAD-LCX angle, remodeling index (RI), coronary calcium score with Gensini score in conventional angiography were assessed. One hundred and twenty-two patients who underwent both CCTA and coronary angiography were analyzed. Results Our analysis showed that the median (percentile 25% to percentile 75%) of the APV, LAD-LCx angle, and calcium score were 31% (17%-47%), 58° (39°-89°), and 31 (0-186), respectively. Also, the mean ± SD of the RI was 1.05 ± 0.20. Significant correlation between LAD-LCx bifurcation angle (0.0001-0.684), APV (0.002-0.281), RI (0.0001-0.438), and calcium score (0.016-0.217) with Gensini score were detected. There was a linear correlation between the mean LAD-LCx bifurcation angle and the Gensini score. The sensitivity and specificity for the cut-off value of 47.5° for the LAD-LCX angle were 86.7% and 82.1%, respectively. Conclusion There is a direct correlation between the LAD-LCx angle and the Gensini score. In addition to plaque characteristics, anatomic-based CCTA-derived indices can be used to identify patients at higher risk for CAD.
Collapse
Affiliation(s)
- Pejman Mansouri
- Tehran Heart Center, Cardiovascular Disease Research InstituteTehran University of Medical SciencesTehranIran
| | - Ebrahim Nematipour
- Tehran Heart Center, Cardiovascular Disease Research InstituteTehran University of Medical SciencesTehranIran
| | - Nadia Rajablou
- School of MedicineTehran University of Medical SciencesTehranIran
| | - Seyyed Mojtaba Ghorashi
- Tehran Heart Center, Cardiovascular Disease Research InstituteTehran University of Medical SciencesTehranIran
| | - Samad Azari
- Hospital Management Research Center, Health Management Research InstituteIran University of Medical SciencesTehranIran
- Research Center for Emergency and Disaster ResilienceRed Crescent Society of the Islamic Republic of IranTehranIran
| | - Negar Omidi
- Cardiovascular Imaging Departement, Tehran Heart Center, School of Medicin, Tehran University of Medical SciencesTehran heart centerTehranIran
- Cardiac Primary Prevention Research Center, Cardiovascular Institute, Tehran University of Medical ScienceTehran heart centerTehranIran
| |
Collapse
|
34
|
Oliveri F, Van Oort MJH, Al Amri I, Bingen BO, Van der Kley F, Jukema JW, Jurado-Roman A, Montero Cabezas J. Coronary calcified nodules versus nonnodular coronary calcifications: a systematic review and meta-analysis. J Cardiovasc Med (Hagerstown) 2024; 25:438-449. [PMID: 38818813 DOI: 10.2459/jcm.0000000000001625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
BACKGROUND Percutaneous coronary intervention (PCI) on severely calcified coronary lesions is challenging. Coronary calcified nodule (CN) refers to an eccentric and protruding coronary calcification associated with plaque vulnerability and adverse clinical events. This study aims to conduct an extensive review of CNs, focusing on its prognostic impact in comparison with nonnodular coronary calcification (N-CN). METHOD A systematic literature review on PubMed, MEDLINE, and EMBASE databases was conducted for relevant articles. Observational studies or randomized controlled trials comparing CNs and N-CNs were included. RESULTS Five studies comparing CNs and N-CNs were pertinent for inclusion. The total number of individuals across these studies was 1456. There were no significant differences in the baseline demographic, clinical, and angiographic data between the CN and N-CN groups. Intracoronary imaging was always utilized. At follow-up, CNs were associated with significantly increased, target vessel revascularization [odds ratio (OR) 2.16; 95% confidence interval (CI): 1.39-3.36, P-value < 0.01, I2 = 0%] and stent thrombosis (OR 9.29; 95% CI: 1.67-51.79, P-value = 0.01, I2 = 0%) compared with N-CN. A trend for greater cardiac death was also assessed in the CN group (OR 1.75; 95% CI: 0.98-3.13, P-value = 0.06, I2 = 0%). CONCLUSION CN has a significantly negative impact on outcomes when compared with N-CN.
Collapse
Affiliation(s)
- F Oliveri
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, University of Pavia, Pavia, Italy
| | - M J H Van Oort
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - I Al Amri
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - B O Bingen
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - F Van der Kley
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J W Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - A Jurado-Roman
- Department of Cardiology, Hospital Universitario La Paz, Madrid, Spain
| | - J Montero Cabezas
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
35
|
Lancaster GI, Morgan PK, Murphy AJ. Iron Macrophages: Dance of Death and MMP Release in Intraplaque Hemorrhage. Thromb Haemost 2024; 124:581-583. [PMID: 38196076 DOI: 10.1055/s-0043-1778071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Affiliation(s)
- Graeme I Lancaster
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Pooranee K Morgan
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| |
Collapse
|
36
|
Chen M, Neverova N, Xu S, Suwannaphoom K, Lluri G, Tamboline M, Duarte S, Fishbein MC, Luo Y, Packard RRS. Invasive electrochemical impedance spectroscopy with phase delay for experimental atherosclerosis phenotyping. FASEB J 2024; 38:e23700. [PMID: 38787606 DOI: 10.1096/fj.202302544rr] [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: 12/11/2023] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Distinguishing quiescent from rupture-prone atherosclerotic lesions has significant translational and clinical implications. Electrochemical impedance spectroscopy (EIS) characterizes biological tissues by assessing impedance and phase delay responses to alternating current at multiple frequencies. We evaluated invasive 6-point stretchable EIS sensors over a spectrum of experimental atherosclerosis and compared results with intravascular ultrasound (IVUS), molecular positron emission tomography (PET) imaging, and histology. Male New Zealand White rabbits (n = 16) were placed on a high-fat diet, with or without endothelial denudation via balloon injury of the infrarenal abdominal aorta. Rabbits underwent in vivo micro-PET imaging of the abdominal aorta with 68Ga-DOTATATE, 18F-NaF, and 18F-FDG, followed by invasive interrogation via IVUS and EIS. Background signal-corrected values of impedance and phase delay were determined. Abdominal aortic samples were collected for histology. Analyses were performed blindly. EIS impedance was associated with markers of plaque activity including macrophage infiltration (r = .813, p = .008) and macrophage/smooth muscle cell (SMC) ratio (r = .813, p = .026). Moreover, EIS phase delay correlated with anatomic markers of plaque burden, namely intima/media ratio (r = .883, p = .004) and %stenosis (r = .901, p = .002), similar to IVUS. 68Ga-DOTATATE correlated with intimal macrophage infiltration (r = .861, p = .003) and macrophage/SMC ratio (r = .831, p = .021), 18F-NaF with SMC infiltration (r = -.842, p = .018), and 18F-FDG correlated with macrophage/SMC ratio (r = .787, p = .036). EIS with phase delay integrates key atherosclerosis features that otherwise require multiple complementary invasive and non-invasive imaging approaches to capture. These findings indicate the potential of invasive EIS to comprehensively evaluate human coronary artery disease.
Collapse
Affiliation(s)
- Michael Chen
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Natalia Neverova
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Ronald Reagan UCLA Medical Center, Los Angeles, California, USA
- West Los Angeles Veterans Affairs Medical Center, Los Angeles, California, USA
| | - Shili Xu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| | - Krit Suwannaphoom
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Gentian Lluri
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Ronald Reagan UCLA Medical Center, Los Angeles, California, USA
| | - Mikayla Tamboline
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Sandra Duarte
- Division of Laboratory and Animal Medicine, University of California, Los Angeles, California, USA
| | - Michael C Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Yuan Luo
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - René R Sevag Packard
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Ronald Reagan UCLA Medical Center, Los Angeles, California, USA
- West Los Angeles Veterans Affairs Medical Center, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, California, USA
- Molecular Biology Institute, University of California, Los Angeles, California, USA
- California NanoSystems Institute, University of California, Los Angeles, California, USA
| |
Collapse
|
37
|
Madaudo C, Coppola G, Parlati ALM, Corrado E. Discovering Inflammation in Atherosclerosis: Insights from Pathogenic Pathways to Clinical Practice. Int J Mol Sci 2024; 25:6016. [PMID: 38892201 PMCID: PMC11173271 DOI: 10.3390/ijms25116016] [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: 04/28/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
This comprehensive review explores the various scenarios of atherosclerosis, a systemic and chronic arterial disease that underlies most cardiovascular disorders. Starting from an overview of its insidious development, often asymptomatic until it reaches advanced stages, the review delves into the pathophysiological evolution of atherosclerotic lesions, highlighting the central role of inflammation. Insights into clinical manifestations, including heart attacks and strokes, highlight the disease's significant burden on global health. Emphasis is placed on carotid atherosclerosis, clarifying its epidemiology, clinical implications, and association with cognitive decline. Prevention strategies, lifestyle modifications, risk factor management, and nuanced antithrombotic treatment considerations are critical to managing cardiovascular complications, thus addressing a crucial aspect of cardiovascular health.
Collapse
Affiliation(s)
- Cristina Madaudo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| | - Giuseppe Coppola
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| | | | - Egle Corrado
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| |
Collapse
|
38
|
Obare LM, Temu T, Mallal SA, Wanjalla CN. Inflammation in HIV and Its Impact on Atherosclerotic Cardiovascular Disease. Circ Res 2024; 134:1515-1545. [PMID: 38781301 PMCID: PMC11122788 DOI: 10.1161/circresaha.124.323891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8+ and CD4+ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| | - Tecla Temu
- Department of Pathology, Harvard Medical School, Boston, MA (T.T.)
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia (S.A.M.)
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| |
Collapse
|
39
|
Blachutzik F, Meier S, Blachutzik M, Schlattner S, Gori T, Ullrich-Daub H, Gaede L, Achenbach S, Möllmann H, Chitic B, Aksoy A, Nickenig G, Weferling M, Dörr O, Boeder N, Bayer M, Hamm C, Nef H. Comparison of interventional treatment options for coronary calcified nodules: A sub-analysis of the ROTA.shock trial. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2024:S1553-8389(24)00507-4. [PMID: 38796321 DOI: 10.1016/j.carrev.2024.05.030] [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: 02/15/2024] [Revised: 03/17/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND The optimal treatment for coronary calcified nodules (CNs) is still unclear. The aim of this study was to compare the modification of these lesions by coronary intravascular lithotripsy (IVL) and rotational atherectomy (RA) using optical coherence tomography (OCT). METHODS ROTA.shock was a 1:1 randomized, prospective, double-arm multi-center non-inferiority trial that compared the use of IVL and RA with percutaneous coronary intervention (PCI) in severely calcified lesions. In 19 of the patients out of this study CNs were detected by OCT in the target lesion and were treated by either IVL or RA. RESULTS The mean angle of CNs was significantly larger in final OCT scans than before RA (92 ± 17° vs. 68 ± 7°; p = 0.01) and IVL (89 ± 18° vs. 60 ± 10°; p = 0.03). The CNs were thinner upon final scans than in initial native scans (RA: 17.8 ± 7.8 mm vs. 38.6 ± 13.1 mm; p = 0.02; IVL: 16.5 ± 9.0 mm vs. 37.2 ± 14.3 mm; p = 0.02). Nodule volume did not differ significantly between native and final OCT scans (RA: 0.66 ± 0.12 mm3 vs. 0.61 ± 0.33 mm3; p = 0.68; IVL: 0.64 ± 0.19 mm3 vs. 0.68 ± 0.22 mm3; p = 0.74). Final stent eccentricity was high with 0.62 ± 0.10 after RA and 0.61 ± 0.09 after IVL. CONCLUSION RA or IVL are unable to reduce the volume of the calcified plaque. CN modulation seems to be mainly induced by the stent implantation and not by RA or IVL.
Collapse
Affiliation(s)
- Florian Blachutzik
- Justus Liebig Universität Giessen, Medizinische Klinik 1, Giessen, Germany; Kardiocentrum Frankfurt an der Klinik Rotes Kreuz, Frankfurt am Main, Germany.
| | - Sophie Meier
- Justus Liebig Universität Giessen, Medizinische Klinik 1, Giessen, Germany
| | - Melissa Blachutzik
- Kerckhoff-Klinik, Department of Cardiology; German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Bad Nauheim, Germany
| | - Sophia Schlattner
- Justus Liebig Universität Giessen, Medizinische Klinik 1, Giessen, Germany; Kerckhoff-Klinik, Department of Cardiology; German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Bad Nauheim, Germany
| | - Tommaso Gori
- Universitätsmedizin Mainz, Kardiologie 1, Mainz, Germany
| | | | - Luise Gaede
- Universitätsklinikum Erlangen, Medizinische Klinik 2, Erlangen, Germany
| | - Stephan Achenbach
- Universitätsklinikum Erlangen, Medizinische Klinik 2, Erlangen, Germany
| | - Helge Möllmann
- St. Johannes-Hospital, Innere Medizin 1, Dortmund, Germany
| | - Bogdan Chitic
- St. Johannes-Hospital, Innere Medizin 1, Dortmund, Germany
| | - Adem Aksoy
- Universitätsklinikum Bonn, Medizinische Klinik 2, Bonn, Germany
| | - Georg Nickenig
- Universitätsklinikum Bonn, Medizinische Klinik 2, Bonn, Germany
| | - Maren Weferling
- Kerckhoff-Klinik, Department of Cardiology; German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Bad Nauheim, Germany
| | - Oliver Dörr
- Justus Liebig Universität Giessen, Medizinische Klinik 1, Giessen, Germany
| | - Niklas Boeder
- Justus Liebig Universität Giessen, Medizinische Klinik 1, Giessen, Germany
| | - Matthias Bayer
- Justus Liebig Universität Giessen, Medizinische Klinik 1, Giessen, Germany
| | - Christian Hamm
- Justus Liebig Universität Giessen, Medizinische Klinik 1, Giessen, Germany; Kerckhoff-Klinik, Department of Cardiology; German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Bad Nauheim, Germany
| | - Holger Nef
- Justus Liebig Universität Giessen, Medizinische Klinik 1, Giessen, Germany; Segeberger Kliniken GmbH, Klinik für Kardiologie und Angiologie, Bad Segeberg, Germany
| |
Collapse
|
40
|
Qi J, Zhou S, Wang G, Hua R, Wang X, He J, Wang Z, Zhu Y, Luo J, Shi W, Luo Y, Chen X. The Antioxidant Dendrobium officinale Polysaccharide Modulates Host Metabolism and Gut Microbiota to Alleviate High-Fat Diet-Induced Atherosclerosis in ApoE -/- Mice. Antioxidants (Basel) 2024; 13:599. [PMID: 38790704 PMCID: PMC11117934 DOI: 10.3390/antiox13050599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/01/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND The discovery of traditional plants' medicinal and nutritional properties has opened up new avenues for developing pharmaceutical and dietary strategies to prevent atherosclerosis. However, the effect of the antioxidant Dendrobium officinale polysaccharide (DOP) on atherosclerosis is still not elucidated. PURPOSE This study aims to investigate the inhibitory effect and the potential mechanism of DOP on high-fat diet-induced atherosclerosis in Apolipoprotein E knockout (ApoE-/-) mice. STUDY DESIGN AND METHODS The identification of DOP was measured by high-performance gel permeation chromatography (HPLC) and Fourier transform infrared spectroscopy (FTIR). We used high-fat diet (HFD)-induced atherosclerosis in ApoE-/- mice as an animal model. In the DOP intervention stage, the DOP group was treated by gavage with 200 μL of 200 mg/kg DOP at regular times each day and continued for eight weeks. We detected changes in serum lipid profiles, inflammatory factors, anti-inflammatory factors, and antioxidant capacity to investigate the effect of the DOP on host metabolism. We also determined microbial composition using 16S rRNA gene sequencing to investigate whether the DOP could improve the structure of the gut microbiota in atherosclerotic mice. RESULTS DOP effectively inhibited histopathological deterioration in atherosclerotic mice and significantly reduced serum lipid levels, inflammatory factors, and malondialdehyde (F/B) production. Additionally, the levels of anti-inflammatory factors and the activity of antioxidant enzymes, including superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX), were significantly increased after DOP intervention. Furthermore, we found that DOP restructures the gut microbiota composition by decreasing the Firmicutes/Bacteroidota (F/B) ratio. The Spearman's correlation analysis indicated that serum lipid profiles, antioxidant activity, and pro-/anti-inflammatory factors were associated with Firmicutes, Bacteroidota, Allobaculum, and Coriobacteriaceae_UCG-002. CONCLUSIONS This study suggests that DOP has the potential to be developed as a food prebiotic for the treatment of atherosclerosis in the future.
Collapse
Affiliation(s)
- Jingyi Qi
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (J.Q.); (S.Z.); (Z.W.); (Y.Z.); (J.L.)
| | - Shuaishuai Zhou
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (J.Q.); (S.Z.); (Z.W.); (Y.Z.); (J.L.)
| | - Guisheng Wang
- Department of Radiology, The Third Medical Centre, Chinese PLA General Hospital, Beijing 100039, China; (G.W.); (R.H.)
| | - Rongrong Hua
- Department of Radiology, The Third Medical Centre, Chinese PLA General Hospital, Beijing 100039, China; (G.W.); (R.H.)
| | - Xiaoping Wang
- Zhejiang Medicine Co., Ltd., Shaoxing 312366, China;
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot 010110, China;
| | - Zi Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (J.Q.); (S.Z.); (Z.W.); (Y.Z.); (J.L.)
| | - Yinhua Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (J.Q.); (S.Z.); (Z.W.); (Y.Z.); (J.L.)
| | - Junjie Luo
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (J.Q.); (S.Z.); (Z.W.); (Y.Z.); (J.L.)
| | - Wenbiao Shi
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (J.Q.); (S.Z.); (Z.W.); (Y.Z.); (J.L.)
| | - Yongting Luo
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (J.Q.); (S.Z.); (Z.W.); (Y.Z.); (J.L.)
| | - Xiaoxia Chen
- Department of Radiology, The Third Medical Centre, Chinese PLA General Hospital, Beijing 100039, China; (G.W.); (R.H.)
| |
Collapse
|
41
|
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
|
42
|
Lu H, Xu Y, Zhao H, Xu X. A novel rabbit model of atherosclerotic vulnerable plaque established by cryofluid-induced endothelial injury. Sci Rep 2024; 14:9447. [PMID: 38658774 PMCID: PMC11043414 DOI: 10.1038/s41598-024-60287-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024] Open
Abstract
Acute thrombosis secondary to atherosclerotic plaque rupture is the main cause of acute cardiac and cerebral ischemia. An animal model of unstable atherosclerotic plaques is highly important for investigating the mechanism of plaque rupture and thrombosis. However, current animal models involve complex operations, are costly, and have plaque morphologies that are different from those of humans. We aimed to establish a simple animal model of vulnerable plaques similar to those of humans. Rabbits were randomly divided into three groups. Group A was given a normal formula diet for 13 weeks. Group C underwent surgery on the intima of the right carotid artery with - 80 °C cryofluid-induced injury after 1 week of a high-fat diet and further feeding a 12-week high-fat diet. Group B underwent the same procedure as Group C but without the - 80 °C cryofluid. Serum lipid levels were detected via ELISA. The plaque morphology, stability and degree of stenosis were evaluated through hematoxylin-eosin (HE) staining, Masson trichrome staining, Elastica van Gieson staining (EVG), and oil red O staining. Macrophages and inflammatory factors in the plaques were assessed via immunohistochemical analysis. The serum low-density lipoprotein cholesterol (LDL-C), triglyceride (TG), and total cholesterol (TC) levels in groups B and C were significantly greater than those in group A. No plaque formation was observed in group A. The plaques in group B were very small. In group C, obvious plaques were observed in the blood vessels, and the plaques exhibited a thin fibrous cap, a large lipid core, and partially visible neovascularization, which is consistent with the characteristics of vulnerable plaques. In the plaques of group C, a large number of macrophages were present, and matrix metalloproteinase 9 (MMP-9) and lectin-like oxidized LDL receptor 1 (LOX-1) were abundantly expressed. We successfully established a rabbit model of vulnerable carotid plaque similar to that of humans through the combination of cryofluid-induced endothelial injury and a high-fat diet, which is feasible and cost effective.
Collapse
Affiliation(s)
- Huaizhi Lu
- Department of Cardiovascular Medicine, First People's Hospital of Shangqiu, Kaixuan South Road 292, Shangqiu, 476000, China.
| | - Yiran Xu
- The Second Naval Hospital of Southern Theater Command of PLA, Sanya, 572029, China
| | - Hui Zhao
- Department of Cardiovascular Medicine, First People's Hospital of Shangqiu, Kaixuan South Road 292, Shangqiu, 476000, China
| | - Xuesheng Xu
- Department of Cardiovascular Medicine, First People's Hospital of Shangqiu, Kaixuan South Road 292, Shangqiu, 476000, China
| |
Collapse
|
43
|
Meloni A, Maffei E, Clemente A, De Gori C, Occhipinti M, Positano V, Berti S, La Grutta L, Saba L, Cau R, Bossone E, Mantini C, Cavaliere C, Punzo B, Celi S, Cademartiri F. Spectral Photon-Counting Computed Tomography: Technical Principles and Applications in the Assessment of Cardiovascular Diseases. J Clin Med 2024; 13:2359. [PMID: 38673632 PMCID: PMC11051476 DOI: 10.3390/jcm13082359] [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: 03/16/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Spectral Photon-Counting Computed Tomography (SPCCT) represents a groundbreaking advancement in X-ray imaging technology. The core innovation of SPCCT lies in its photon-counting detectors, which can count the exact number of incoming x-ray photons and individually measure their energy. The first part of this review summarizes the key elements of SPCCT technology, such as energy binning, energy weighting, and material decomposition. Its energy-discriminating ability represents the key to the increase in the contrast between different tissues, the elimination of the electronic noise, and the correction of beam-hardening artifacts. Material decomposition provides valuable insights into specific elements' composition, concentration, and distribution. The capability of SPCCT to operate in three or more energy regimes allows for the differentiation of several contrast agents, facilitating quantitative assessments of elements with specific energy thresholds within the diagnostic energy range. The second part of this review provides a brief overview of the applications of SPCCT in the assessment of various cardiovascular disease processes. SPCCT can support the study of myocardial blood perfusion and enable enhanced tissue characterization and the identification of contrast agents, in a manner that was previously unattainable.
Collapse
Affiliation(s)
- Antonella Meloni
- Bioengineering Unit, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (V.P.)
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| | - Erica Maffei
- Department of Radiology, Istituto di Ricovero e Cura a Carattere Scientifico SYNLAB SDN, 80131 Naples, Italy; (E.M.); (C.C.); (B.P.)
| | - Alberto Clemente
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| | - Carmelo De Gori
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| | - Mariaelena Occhipinti
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| | - Vicenzo Positano
- Bioengineering Unit, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (V.P.)
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| | - Sergio Berti
- Diagnostic and Interventional Cardiology Department, Fondazione G. Monasterio CNR-Regione Toscana, 54100 Massa, Italy;
| | - Ludovico La Grutta
- Department of Radiology, University Hospital “P. Giaccone”, 90127 Palermo, Italy;
| | - Luca Saba
- Department of Radiology, University Hospital of Cagliari, 09042 Monserrato (CA), Italy; (L.S.); (R.C.)
| | - Riccardo Cau
- Department of Radiology, University Hospital of Cagliari, 09042 Monserrato (CA), Italy; (L.S.); (R.C.)
| | - Eduardo Bossone
- Department of Cardiology, Ospedale Cardarelli, 80131 Naples, Italy;
| | - Cesare Mantini
- Department of Radiology, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Carlo Cavaliere
- Department of Radiology, Istituto di Ricovero e Cura a Carattere Scientifico SYNLAB SDN, 80131 Naples, Italy; (E.M.); (C.C.); (B.P.)
| | - Bruna Punzo
- Department of Radiology, Istituto di Ricovero e Cura a Carattere Scientifico SYNLAB SDN, 80131 Naples, Italy; (E.M.); (C.C.); (B.P.)
| | - Simona Celi
- BioCardioLab, Fondazione G. Monasterio CNR-Regione Toscana, 54100 Massa, Italy;
| | - Filippo Cademartiri
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| |
Collapse
|
44
|
Li X, Yang Y, Wang Z, Lin X, Fu X, He X, Liu M, Wang JX, Yu T, Sun P. CircHIPK3 targets DRP1 to mediate hydrogen peroxide-induced necroptosis of vascular smooth muscle cells and atherosclerotic vulnerable plaque formation. J Adv Res 2024:S2090-1232(24)00154-1. [PMID: 38621622 DOI: 10.1016/j.jare.2024.04.011] [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: 12/28/2023] [Revised: 03/21/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024] Open
Abstract
INTRODUCTION Necroptosis triggered by H2O2 is hypothesized to be a critical factor in the rupture of atherosclerotic plaques, which may precipitate acute cardiovascular events. Nevertheless, the specific regulatory molecules of this development remain unclear. We aims to elucidate a mechanism from the perspective of circular RNA. OBJECTIVES There are few studies on circRNA in VSMCs necroptosis. The objective of our research is to shed light on the intricate roles that circHIPK3 plays in the process of necroptosis in VSMCs and the development of atherosclerotic plaques that are prone to rupture. Our study elucidates the specific molecular mechanisms by which circHIPK3 regulates necroptosis and atherosclerotic vulnerable plaque formation through targeted proteins. Identifying this mechanism at the cellular level offers a molecular framework for understanding plaque progression and stability regulation, as well as a potential biomarker for the prognosis of susceptible atherosclerotic plaques. METHODS We collected clinical vascular tissue for HE staining and Masson staining to determine the presence and stability of plaques. Then, NCBI database was used to screen out circRNA with elevated expression level in plaque tissue, and the up-regulated circRNA, circHIPK3, was verified by qRT-PCR and FISH. Further, we synthesized circHIPK3's small interference sequence and overexpressed plasmid in vitro, and verified its regulation effect on necroptosis of VSMCs under physiological and pathological conditions by WB, qRT-PCR and PI staining. Through RNA pull down, mass spectrometry and RNA immunoprecipitation, DRP1 was identified as circHIPK3 binding protein and was positively regulated by circHIPK3. Meanwhile, on the basis of silencing of DRP1, the regulation of circHIPK3 on necroptosis is verified to be mediated by DRP1. Finally, we validated the regulation of circHIPK3 on vulnerable plaque formation in ApoE-/- mice. RESULTS We investigated that circHIPK3 was highly expressed in vulnerable plaques, and the increase in expression level promoted H2O2 induced necroptosis of VSMCs. CircHIPK3 targeted the protein DRP1, leading to an elevation in mitochondrial division rate, resulting in increased reactive oxygen species and impaired mitochondrial function, ultimately leading to necroptosis of VSMCs and vulnerable plaque formation. CONCLUSION CircHIPK3 interact with DRP1 involve in H2O2 induced Mitochondrial damage and necroptosis of VSMCs, and Silencing circHIPK3 in vivo can reduce atherosclerotic vulnerable plaque formation. Our research findings may have applications in providing diagnostic biomarkers for vulnerable plaques.
Collapse
Affiliation(s)
- Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao 266071, People's Republic of China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China
| | - Xiaotong Lin
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao 266011, People's Republic of China
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China
| | - Xiangqin He
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China
| | - Meixin Liu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China
| | - Jian-Xun Wang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao 266071, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China; Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China.
| | - Pin Sun
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China.
| |
Collapse
|
45
|
Homma S, Kato K. Validity of Atherosclerotic Calcified Lesions Observed on Low-Dose Computed Tomography and Cardio-Ankle Vascular Index as Surrogate Markers of Atherosclerosis Progression. Angiology 2024; 75:349-358. [PMID: 36787785 DOI: 10.1177/00033197231155963] [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: 02/16/2023]
Abstract
The significance of atherosclerotic calcified lesions observed on low-dose computed tomography (LDCT) performed during general checkups was investigated. The coronary arteries (CA), ascending aorta and aortic arch (AAAA), descending thoracic aorta (DTA), and abdominal aorta (AA) were examined. Semiquantitative calcified index analysis of the DTA and AA in terms of atherosclerosis risk factors and cardio-ankle vascular index (CAVI) measurements was also performed. We included 1594 participants (mean age: 59.2 years; range: 31-91 years). The prevalence of calcified lesions was 71.0%, 66.6%, 57.2%, and 37.9% in the AA, CA, AAAA, and DTA, respectively. Age-related advances in calcification among participants with no major risk factors, revealed that calcification appeared earliest in the AA, followed by the CA, AAAA, and DTA. Participants with calcified lesions in all arteries had a significantly greater CAVI than those without calcification. The CAVI was negatively correlated with low-density lipoprotein cholesterol levels, particularly in participants without calcified lesions in the DTA. Calcified lesions on LDCT could indicate the end stage of atherosclerotic lesions. The CAVI can be used to assess atherosclerotic changes at all stages of disease progression. A combination of LDCT and CAVI could be used as a routine non-invasive assessment of atherosclerosis.
Collapse
Affiliation(s)
- Satoki Homma
- Health Care Center in Saitama Medical Center of the Japan Community Health Care Organization, Saitama, Japan
- Faculty of Nursing and Medical Care, Keio University & Keio Research Institute at SFC (Shonan Fujisawa Campus), Fujisawa, Japan
| | - Kiyoe Kato
- Center of General Health Check-Up, Saiseikai Central Hospital, Tokyo, Japan
| |
Collapse
|
46
|
Jansen I, Cahalane R, Hengst R, Akyildiz A, Farrell E, Gijsen F, Aikawa E, van der Heiden K, Wissing T. The interplay of collagen, macrophages, and microcalcification in atherosclerotic plaque cap rupture mechanics. Basic Res Cardiol 2024; 119:193-213. [PMID: 38329498 PMCID: PMC11008085 DOI: 10.1007/s00395-024-01033-5] [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: 08/21/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
The rupture of an atherosclerotic plaque cap overlying a lipid pool and/or necrotic core can lead to thrombotic cardiovascular events. In essence, the rupture of the plaque cap is a mechanical event, which occurs when the local stress exceeds the local tissue strength. However, due to inter- and intra-cap heterogeneity, the resulting ultimate cap strength varies, causing proper assessment of the plaque at risk of rupture to be lacking. Important players involved in tissue strength include the load-bearing collagenous matrix, macrophages, as major promoters of extracellular matrix degradation, and microcalcifications, deposits that can exacerbate local stress, increasing tissue propensity for rupture. This review summarizes the role of these components individually in tissue mechanics, along with the interplay between them. We argue that to be able to improve risk assessment, a better understanding of the effect of these individual components, as well as their reciprocal relationships on cap mechanics, is required. Finally, we discuss potential future steps, including a holistic multidisciplinary approach, multifactorial 3D in vitro model systems, and advancements in imaging techniques. The obtained knowledge will ultimately serve as input to help diagnose, prevent, and treat atherosclerotic cap rupture.
Collapse
Affiliation(s)
- Imke Jansen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rachel Cahalane
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ranmadusha Hengst
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ali Akyildiz
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Frank Gijsen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kim van der Heiden
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamar Wissing
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
47
|
Bec J, Zhou X, Villiger M, Southard JA, Bouma B, Marcu L. Dual modality intravascular catheter system combining pulse-sampling fluorescence lifetime imaging and polarization-sensitive optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2024; 15:2114-2132. [PMID: 38633060 PMCID: PMC11019710 DOI: 10.1364/boe.516515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 04/19/2024]
Abstract
The clinical management of coronary artery disease and the prevention of acute coronary syndromes require knowledge of the underlying atherosclerotic plaque pathobiology. Hybrid imaging modalities capable of comprehensive assessment of biochemical and morphological plaques features can address this need. Here we report the first implementation of an intravascular catheter system combining fluorescence lifetime imaging (FLIm) with polarization-sensitive optical coherence tomography (PSOCT). This system provides multi-scale assessment of plaque structure and composition via high spatial resolution morphology from OCT, polarimetry-derived tissue microstructure, and biochemical composition from FLIm, without requiring any molecular contrast agent. This result was achieved with a low profile (2.7 Fr) double-clad fiber (DCF) catheter and high speed (100 fps B-scan rate, 40 mm/s pullback speed) console. Use of a DCF and broadband rotary junction required extensive optimization to mitigate the reduction in OCT performance originating from additional reflections and multipath artifacts. This challenge was addressed by the development of a broad-band (UV-visible-IR), high return loss (47 dB) rotary junction. We demonstrate in phantoms, ex vivo swine coronary specimens and in vivo swine heart (percutaneous coronary access) that the FLIm-PSOCT catheter system can simultaneously acquire co-registered FLIm data over four distinct spectral bands (380/20 nm, 400/20 nm, 452/45 nm, 540/45 nm) and PSOCT backscattered intensity, birefringence, and depolarization. The unique ability to collect complementary information from tissue (e.g., morphology, extracellular matrix composition, inflammation) with a device suitable for percutaneous coronary intervention offers new opportunities for cardiovascular research and clinical diagnosis.
Collapse
Affiliation(s)
- Julien Bec
- Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Xiangnan Zhou
- Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Martin Villiger
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey A. Southard
- Division of Cardiovascular Medicine, UC Davis Health System, University of California-Davis, Sacramento, CA 95817, USA
| | - Brett Bouma
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Laura Marcu
- Biomedical Engineering, University of California, Davis, CA 95616, USA
| |
Collapse
|
48
|
Almeida AG, Grapsa J, Gimelli A, Bucciarelli-Ducci C, Gerber B, Ajmone-Marsan N, Bernard A, Donal E, Dweck MR, Haugaa KH, Hristova K, Maceira A, Mandoli GE, Mulvagh S, Morrone D, Plonska-Gosciniak E, Sade LE, Shivalkar B, Schulz-Menger J, Shaw L, Sitges M, von Kemp B, Pinto FJ, Edvardsen T, Petersen SE, Cosyns B. Cardiovascular multimodality imaging in women: a scientific statement of the European Association of Cardiovascular Imaging of the European Society of Cardiology. Eur Heart J Cardiovasc Imaging 2024; 25:e116-e136. [PMID: 38198766 DOI: 10.1093/ehjci/jeae013] [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: 12/28/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024] Open
Abstract
Cardiovascular diseases (CVD) represent an important cause of mortality and morbidity in women. It is now recognized that there are sex differences regarding the prevalence and the clinical significance of the traditional cardiovascular (CV) risk factors as well as the pathology underlying a range of CVDs. Unfortunately, women have been under-represented in most CVD imaging studies and trials regarding diagnosis, prognosis, and therapeutics. There is therefore a clear need for further investigation of how CVD affects women along their life span. Multimodality CV imaging plays a key role in the diagnosis of CVD in women as well as in prognosis, decision-making, and monitoring of therapeutics and interventions. However, multimodality imaging in women requires specific consideration given the differences in CVD between the sexes. These differences relate to physiological changes that only women experience (e.g. pregnancy and menopause) as well as variation in the underlying pathophysiology of CVD and also differences in the prevalence of certain conditions such as connective tissue disorders, Takotsubo, and spontaneous coronary artery dissection, which are all more common in women. This scientific statement on CV multimodality in women, an initiative of the European Association of Cardiovascular Imaging of the European Society of Cardiology, reviews the role of multimodality CV imaging in the diagnosis, management, and risk stratification of CVD, as well as highlights important gaps in our knowledge that require further investigation.
Collapse
Affiliation(s)
- Ana G Almeida
- Heart and Vessels Department, University Hospital Santa Maria, CAML, CCUL, Faculty of Medicine of Lisbon University, Lisbon, Portugal
| | - Julia Grapsa
- Cardiology Department, Guys and St Thomas NHS Trust, London, UK
| | - Alessia Gimelli
- Imaging Department, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Chiara Bucciarelli-Ducci
- Department of Cardiology, Royal Brompton and Harefield Hospitals, Guys' and St Thomas NHS Hospitals, London, UK
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Bernhard Gerber
- Service de Cardiologie, Département Cardiovasculaire, Cliniques Universitaires St. Luc, UCLouvain, Brussels, Belgium
- Division CARD, Institut de Recherche Expérimental et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Nina Ajmone-Marsan
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne Bernard
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, Tours, France
- Service de Cardiologie, CHRU de Tours, Tours, France
| | - Erwan Donal
- CHU Rennes, Inserm, LTSI-UMR 1099, University of Rennes, Rennes, France
| | - Marc R Dweck
- Centre for Cardiovascular Science, Chancellors Building, Little France Crescent, Edinburgh, UK
| | - Kristina H Haugaa
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- ProCardio Center for Innovation, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Krassimira Hristova
- Center for Cardiovascular Diseases, Faculty of Medicine, Sofia University, Sofia, Bulgaria
| | - Alicia Maceira
- Ascires Biomedical Group, Valencia, Spain
- Department of Medicine, Health Sciences School, UCH-CEU University, Valencia, Spain
| | - Giulia Elena Mandoli
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - Sharon Mulvagh
- Division of Cardiology, Dalhousie University, Halifax, NS, Canada
| | - Doralisa Morrone
- Division of Cardiology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | | | - Leyla Elif Sade
- Cardiology Department, University of Baskent, Ankara, Turkey
- UPMC Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jeanette Schulz-Menger
- Charité ECRC Medical Faculty of the Humboldt University Berlin and Helios-Clinics, Berlin, Germany
- DZHK, Partner site Berlin, Berlin, Germany
| | - Leslee Shaw
- Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Marta Sitges
- Cardiovascular Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Institut Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- CIBERCV, Barcelona, Spain
| | - Berlinde von Kemp
- Cardiology, Centrum voor Hart en Vaatziekten (CHVZ), Universitair Ziejkenhuis Brussel (UZB), Vrij Universiteit Brussel (VUB), Brussels, Belgium
| | - Fausto J Pinto
- Heart and Vessels Department, University Hospital Santa Maria, CAML, CCUL, Faculty of Medicine of Lisbon University, Lisbon, Portugal
| | - Thor Edvardsen
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- ProCardio Center for Innovation, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Steffen E Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Bernard Cosyns
- Cardiology, Centrum voor Hart en Vaatziekten (CHVZ), Universitair Ziejkenhuis Brussel (UZB), Vrij Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
49
|
Makuch M, Stepanechko M, Bzowska M. The dance of macrophage death: the interplay between the inevitable and the microenvironment. Front Immunol 2024; 15:1330461. [PMID: 38576612 PMCID: PMC10993711 DOI: 10.3389/fimmu.2024.1330461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/26/2024] [Indexed: 04/06/2024] Open
Abstract
Macrophages are highly plastic cells ubiquitous in various tissues, where they perform diverse functions. They participate in the response to pathogen invasion and inflammation resolution following the immune response, as well as the maintenance of homeostasis and proper tissue functions. Macrophages are generally considered long-lived cells with relatively strong resistance to numerous cytotoxic factors. On the other hand, their death seems to be one of the principal mechanisms by which macrophages perform their physiological functions or can contribute to the development of certain diseases. In this review, we scrutinize three distinct pro-inflammatory programmed cell death pathways - pyroptosis, necroptosis, and ferroptosis - occurring in macrophages under specific circumstances, and explain how these cells appear to undergo dynamic yet not always final changes before ultimately dying. We achieve that by examining the interconnectivity of these cell death types, which in macrophages seem to create a coordinated and flexible system responding to the microenvironment. Finally, we discuss the complexity and consequences of pyroptotic, necroptotic, and ferroptotic pathway induction in macrophages under two pathological conditions - atherosclerosis and cancer. We summarize damage-associated molecular patterns (DAMPs) along with other microenvironmental factors, macrophage polarization states, associated mechanisms as well as general outcomes, as such a comprehensive look at these correlations may point out the proper methodologies and potential therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Małgorzata Bzowska
- Department of Immunology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
50
|
Zhang J, Li S, Wu L, Wang H, Wang C, Zhou Y, Sui B, Zhao X. Application of Dual-Layer Spectral-Detector Computed Tomography Angiography in Identifying Symptomatic Carotid Atherosclerosis: A Prospective Observational Study. J Am Heart Assoc 2024; 13:e032665. [PMID: 38497470 PMCID: PMC11010034 DOI: 10.1161/jaha.123.032665] [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: 09/13/2023] [Accepted: 02/20/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Dual-layer spectral-detector dual-energy computed tomography angiography (DLCTA) can distinguish components of carotid plaques. Data on identifying symptomatic carotid plaques in patients using DLCTA are not available. METHODS AND RESULTS In this prospective observational study, patients with carotid plaques were enrolled and received DLCTA. The attenuation for both polyenergetic image and virtual monoenergetic images (40, 70, 100, and 140 keV), as well as Z-effective value, were recorded in the noncalcified regions of plaques. Logistic regression models were used to assess the association between attenuations of DLCTA and the presence of symptomatic carotid plaques. In total, 100 participants (mean±SD age, 64.37±8.31 years; 82.0% were men) were included, and 36% of the cases were identified with the symptomatic group. DLCTA parameters were different between 2 groups (symptomatic versus asymptomatic: computed tomography [CT] 40 keV, 152.63 [interquartile range (IQR), 70.22-259.78] versus 256.78 [IQR, 150.34-408.13]; CT 70 keV, 81.28 [IQR, 50.13-119.33] versus 108.87 [IQR, 77.01-165.88]; slope40-140 keV, 0.91 [IQR, 0.35-1.87] versus 1.92 [IQR, 0.96-3.00]; Z-effective value, 7.92 [IQR, 7.53-8.46] versus 8.41 [IQR, 7.94-8.92]), whereas no difference was found in conventional polyenergetic images. The risk of symptomatic plaque was lower in the highest tertiles of attenuations in CT 40 keV (adjusted odds ratio [OR], 0.243 [95% CI, 0.078-0.754]), CT 70 keV (adjusted OR, 0.313 [95% CI, 0.104-0.940]), Z-effective values (adjusted OR, 0.138 [95% CI, 0.039-0.490]), and slope40-140 keV (adjusted OR, 0.157 [95% CI, 0.046-0.539]), with all P values and P trends <0.05. The areas under the curve for CT 40 keV, CT 70 keV, slope 40 to 140 keV, and Z-effective values were 0.64, 0.61, 0.64, and 0.63, respectively. CONCLUSIONS Parameters of DLCTA might help assist in distinguishing symptomatic carotid plaques. Further studies with a larger sample size may address the overlap and improve the diagnostic accuracy.
Collapse
Affiliation(s)
- Jia Zhang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Sijia Li
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Lei Wu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Haoyuan Wang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Chuanying Wang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Yinan Zhou
- CT Clinical SpecialistPhilips HealthcareBeijingChina
| | - Binbin Sui
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- Tiantan Neuroimaging Center of ExcellenceChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Xingquan Zhao
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- Research Unit of Artificial Intelligence in Cerebrovascular DiseaseChinese Academy of Medical SciencesBeijingChina
| |
Collapse
|