51
|
Lee JE, Park HM, Lim Y, Jeong WGI, Kim YH. Pathophysiology and Role of Coronary CT Angiography in Stable Angina. JOURNAL OF THE KOREAN SOCIETY OF RADIOLOGY 2022; 83:42-53. [PMID: 36237352 PMCID: PMC9238201 DOI: 10.3348/jksr.2021.0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/11/2021] [Accepted: 12/24/2021] [Indexed: 11/28/2022]
Abstract
관상동맥질환의 임상적인 개념은 지난 10년 동안 패러다임의 변화를 보여왔다. 관상동맥질환은 대부분 진행성 질환이며, 관상동맥질환 환자는 질병 진행의 어느 시점에서도 급성 관상동맥 증후군에 직면할 수 있다. 이러한 임상적 맥락에서 2019년 유럽심장학회 가이드라인에서는 조기 진단과 꾸준한 관리가 필요한 관상동맥질환의 임상적인 중요성을 반영하여 “만성 관상동맥증후군”이라는 용어의 사용을 발표하였다. 최근 관상동맥 전산화단층촬영 혈관조영술을 이용한 관상동맥질환의 평가는 많은 발전을 이루었고, 안정형 협심증 또는 만성 관상동맥증후군 환자에서 관상동맥 전산화단층촬영 혈관조영술의 임상적 유용성은 초기 무증상 관상동맥질환의 발견에서부터 죽상경화판의 특성 분석 및 관상동맥질환의 치료 전략 결정에 도움을 주는 역할까지 관상동맥질환의 다양한 단계에 걸쳐 입증되고 있다. 이 종설에서는 안정형 협심증 환자의 이해를 돕는 병태생리를 설명하고 이에 대한 관상동맥 전산화단층촬영의 임상적 적용과 역할에 대해 알아보고자 한다.
Collapse
Affiliation(s)
- Jong Eun Lee
- Department of Radiology, Chonnam National University Hospital, Gwangju, Korea
| | - Hye Mi Park
- Department of Radiology, Chonnam National University Hospital, Gwangju, Korea
| | - Yongwhan Lim
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea
| | - Won GI Jeong
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Yun-Hyeon Kim
- Department of Radiology, Chonnam National University Hospital, Gwangju, Korea
| |
Collapse
|
52
|
Ruan W, Wang M, Sun C, Yao J, Ma Y, Ma H, Ding J, Lian X. Correlation between neutrophil-to-lymphocyte ratio and stability of carotid plaques. Clin Neurol Neurosurg 2021; 212:107055. [PMID: 34844159 DOI: 10.1016/j.clineuro.2021.107055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/05/2021] [Accepted: 11/16/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE The neutrophil-to-lymphocyte ratio (NLR) has been proved to be a strong predictor of carotid atherosclerotic plaque, but the correlation between NLR and the stability of carotid plaque is not clear. Thus we conducted a study to evaluate the correlation between NLR and the stability of carotid atherosclerotic plaque, and to develop a new evaluation scale for rapid clinical evaluation of carotid plaque stability. METHODS We recruited 528 patients with acute anterior circulation ischemic stroke who were in accordance with extracranial and intracranial large artery atherosclerosis of Chinese ischemic stroke subtype. Blood routine examination and carotid ultrasound examination were performed on admission. According to the ultrasonic characteristics, the patients were divided into plaque stabilization group and plaque instability group. RESULTS There was significant difference in NLR between plaque stability and instability groups (P < 0.001). The risk of plaque instability increased with the increase of NLR (odds ratio (OR), 4.737; 95% confidence interval (CI), 3.404-6.592; P < 0.001). Receiver operating characteristic (ROC) curve showed that the critical point of NLR is 2.55 and the area under the curve (AUC) was 0.782 (95%CI, 0.740-0.823; P < 0.001). The best cut-off value of the evaluation scale was ≥ 4 points (sensitivity, 0.77; specificity, 0.75; accuracy, 0.76). CONCLUSION There is a correlation between NLR and carotid plaque instability. NLR may be useful as a potential inflammation biomarker indicating the risk of unstable carotid plaques. The new scoring scale is a reliable index to predict the stability of carotid plaque.
Collapse
Affiliation(s)
- Wang Ruan
- Department of Neurology, Third Affiliated Hospital, Soochow University, Changzhou 213003, China
| | - Mengmeng Wang
- Department of Neurology, Third Affiliated Hospital, Soochow University, Changzhou 213003, China
| | - Chuxue Sun
- Department of Ultrasonography, Third Affiliated Hospital, Soochow University, Changzhou 213003, China
| | - Jianrong Yao
- Department of Neurology, Third Affiliated Hospital, Soochow University, Changzhou 213003, China
| | - Yazhou Ma
- Department of Neurology, Third Affiliated Hospital, Soochow University, Changzhou 213003, China
| | - Huayi Ma
- Department of Neurology, Shantou Central Hospital, Shantou 515000, China
| | - Jia Ding
- Department of Neurology, Third Affiliated Hospital, Soochow University, Changzhou 213003, China
| | - Xuegan Lian
- Department of Neurology, Third Affiliated Hospital, Soochow University, Changzhou 213003, China.
| |
Collapse
|
53
|
Plaque Character and Progression According to the Location of Coronary Atherosclerotic Plaque. Am J Cardiol 2021; 158:15-22. [PMID: 34465463 DOI: 10.1016/j.amjcard.2021.07.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 01/08/2023]
Abstract
Although acute coronary syndrome culprit lesions occur more frequently in the proximal coronary artery, whether the proximal clustering of high-risk plaque is reflected in earlier-stage atherosclerosis remains unclarified. We evaluated the longitudinal distribution of stable atherosclerotic lesions on coronary computed tomography angiography (CCTA) in 1,478 patients (mean age, 61 years; men, 58%) enrolled from a prospective multinational registry of consecutive patients undergoing serial CCTA. Of 3,202 coronary artery lesions identified, 2,140 left lesions were classified (based on the minimal lumen diameter location) into left main (LM, n = 128), proximal (n = 739), and other (n = 1,273), and 1,062 right lesions were classified into proximal (n = 355) and other (n = 707). Plaque volume (PV) was the highest in proximal lesions (median, 26.1 mm3), followed by LM (20.6 mm3) and other lesions (15.0 mm3, p <0.001), for left lesions, and was lager in proximal (25.8 mm3) than in other lesions (15.2 mm3, p <0.001) for right lesions. On both sides, proximally located lesions tended to have greater necrotic core and fibrofatty components than other lesions (left: LM, 10.6%; proximal, 5.8%; other, 3.4% of the total PV, p <0.001; right: proximal, 8.4%; other 3.1%, p <0.001), with less calcified plaque component (left: LM, 18.3%; proximal, 30.3%; other, 37.7%, p <0.001; right: proximal, 23.3%, other, 36.6%, p <0.001), and tended to progress rapidly (adjusted odds ratios: left: LM, reference; proximal, 0.95, p = 0.803; other, 0.64, p = 0.017; right: proximal, reference; other, 0.52, p <0.001). Proximally located plaques were larger, with more risky composition, and progressed more rapidly.
Collapse
|
54
|
Lu G, Ye W, Ou J, Li X, Tan Z, Li T, Liu H. Coronary Computed Tomography Angiography Assessment of High-Risk Plaques in Predicting Acute Coronary Syndrome. Front Cardiovasc Med 2021; 8:743538. [PMID: 34660742 PMCID: PMC8517134 DOI: 10.3389/fcvm.2021.743538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/07/2021] [Indexed: 01/07/2023] Open
Abstract
Coronary computed tomography angiography (CCTA) is a comprehensive, non-invasive and cost-effective imaging assessment approach, which can provide the ability to identify the characteristics and morphology of high-risk atherosclerotic plaques associated with acute coronary syndrome (ACS). The development of CCTA and latest advances in emerging technologies, such as computational fluid dynamics (CFD), have made it possible not only to identify the morphological characteristics of high-risk plaques non-invasively, but also to assess the hemodynamic parameters, the environment surrounding coronaries and so on, which may help to predict the risk of ACS. In this review, we present how CCTA was used to characterize the composition and morphology of high-risk plaques prone to ACS and the current role of CCTA, including emerging CCTA technologies, advanced analysis, and characterization techniques in prognosticating the occurrence of ACS.
Collapse
Affiliation(s)
- Guanyu Lu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,College of Medicine, Shantou University, Shantou, China
| | - Weitao Ye
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jiehao Ou
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xinyun Li
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zekun Tan
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Tingyu Li
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hui Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,College of Medicine, Shantou University, Shantou, China
| |
Collapse
|
55
|
Dual-energy CT plaque characteristics of post mortem thin-cap fibroatheroma in comparison to infarct-related culprit lesions. Heart Vessels 2021; 37:400-410. [PMID: 34608510 DOI: 10.1007/s00380-021-01942-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 09/10/2021] [Indexed: 10/20/2022]
Abstract
Improvement of non-invasive identification of high-risk plaque may increase the preventive options of acute coronary syndrome. To describe the characteristics of thin-cap fibroatheroma (TCFA) in a post mortem model in comparison to characteristics of culprit lesions in patients with non-ST-elevation-myocardial-infarction (NSTEMI) using the dual energy computed tomography (DECT). Three post mortem hearts were prepared with iodine-contrast, inserted in a Kyoto phantom and scanned by DECT. Six TCFA were identified using histopathological analysis (cap thickness < 65 μm and necrotic core > 10% of the plaque area). In the NSTEMI group, 29 patients were scheduled to DECT prior to coronary angiography and invasive treatment. Culprit lesions were identified blinded for the patient history by two independent invasive cardiologists using the coronary angiography. The DECT analysis of TCFA and culprit lesions was performed retrospectively with determination of effective atomic number (Effective-Z), Hounsfield Unit (HU), plaque type (non-calcified, predominantly non-calcified, predominantly calcified or calcified), spotty calcification,, plaque length, plaque volume and plaque burden and the remodeling index. The Effective-Z, HU and plaqueburden were significantly different between TCFA and culprit lesions (P < 0.05).The TCFA plaques were more calcified in comparison to culprit lesions (P < 0.05). No significant difference in the other plaque characteristics was observed. The use of DECT demonstrated different Effective-Z values and different characteristics of post mortem TCFA in comparison to in vivo culprit lesions. This finding may highlight, that not all TCFA should be considered as vulnerable.
Collapse
|
56
|
Imai M, Kawamura M, Kochi I, Matsuoka T, Kihara S, Yamamoto H. Anti-Apo B-100 Autoantibody is a Marker of Unstable Coronary Plaque. J Atheroscler Thromb 2021; 28:1025-1034. [PMID: 33191364 PMCID: PMC8560848 DOI: 10.5551/jat.58784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/23/2020] [Indexed: 01/14/2023] Open
Abstract
AIMS Cardiovascular diseases (CVD) are a global leading cause of mortality. However, few biomarkers are available to predict future coronary plaque rupture. We have recently demonstrated that low levels of anti-apolipoprotein B-100 autoantibody (anti-apo B-100 Ab) correlated with an increased CVD risk in Japanese patients with diabetes. In the present study, we examined the relationship between serum anti-apo B-100 Ab levels and coronary plaque characteristics in patients undergoing elective percutaneous coronary intervention (PCI). METHODS We conducted iMAP®-intravascular ultrasound (IVUS) in 88 Japanese male patients undergoing elective PCI, and the five consecutive slices of IVUS images at the center of the most stenotic culprit lesion were used for identifying the plaque characteristics. The serum levels of anti-apo B-100 Ab against synthetic peptides (p45 or p210) were measured using a homemade enzyme-linked immunosorbent assay. RESULTS Serum IgG levels of anti-apo B-100 Ab against both native p45 and p210 (IgG N-p45 and IgGN-p210) and malondialdehyde (MDA)-modified p45 and p210 (IgGMDA-p45 or IgGMDA-p210) showed a negative correlation with plaque burden in total male patients undergoing elective PCI. Additionally, both IgGN-p45 and IgGN-p210, but neither IgGMDA-p45 nor IgGMDA-p210, correlated negatively with necrotic and positively with fibrotic components of iMAP®-IVUS plaque characteristics in the patients with <1 month statin treatment before elective PCI ("statin-untreated" group). There was no significant correlation between anti-apo B-100 Ab and any plaque characteristics in the patients with statin treatment for 1 month or more before elective PCI ("statin-treated" group). CONCLUSION Measuring serum levels of anti-apo B-100 Ab might be helpful in the evaluation of unstable coronary plaque in male CVD patients without statin treatment.
Collapse
Affiliation(s)
- Minami Imai
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine
| | - Mari Kawamura
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine
| | - Ikoi Kochi
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine
| | - Tetsuro Matsuoka
- Department of Cardiology, Hyogo Prefectural Nishinomiya Hospital
| | - Shinji Kihara
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine
| | - Hiroyasu Yamamoto
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine
| |
Collapse
|
57
|
Aguirre AD, Arbab-Zadeh A, Soeda T, Fuster V, Jang IK. Optical Coherence Tomography of Plaque Vulnerability and Rupture: JACC Focus Seminar Part 1/3. J Am Coll Cardiol 2021; 78:1257-1265. [PMID: 34531027 PMCID: PMC9851427 DOI: 10.1016/j.jacc.2021.06.050] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/22/2021] [Indexed: 01/21/2023]
Abstract
Plaque rupture is the most common cause of acute coronary syndromes and sudden cardiac death. Characteristics and pathobiology of vulnerable plaques prone to plaque rupture have been studied extensively over 2 decades in humans using optical coherence tomography (OCT), an intravascular imaging technique with micron scale resolution. OCT studies have identified key features of plaque vulnerability and described the in vivo characteristics and spatial distribution of thin cap fibroatheromas as major precursors to plaque rupture. In addition, OCT data supports the evolving understanding of coronary heart disease as a panvascular process associated with inflammation. In the setting of high atherosclerotic burden, plaque ruptures often occur at multiple sites in the coronary arteries, and plaque progression and healing are dynamic processes modulated by systemic risk factors. This review details major investigations with intravascular OCT into the biology and clinical implications of plaque vulnerability and plaque rupture.
Collapse
Affiliation(s)
- Aaron D. Aguirre
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA,Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA,Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Armin Arbab-Zadeh
- Department of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Tsunenari Soeda
- Department of Cardiology, Nara Medical University, Nara, Japan
| | - Valentin Fuster
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA,Kyung Hee University, Seoul, South Korea
| |
Collapse
|
58
|
Gao F, Wang ZJ, Ma XT, Shen H, Yang LX, Zhou YJ. Effect of alirocumab on coronary plaque in patients with coronary artery disease assessed by optical coherence tomography. Lipids Health Dis 2021; 20:106. [PMID: 34511134 PMCID: PMC8436513 DOI: 10.1186/s12944-021-01528-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/23/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Proprotein convertase subtilisin kexin type 9 (PCSK9) inhibitors have been demonstrated to produce significantly greater reduction in LDL cholesterol levels and cardiovascular events than standard statin therapy. However, evidence on the impact of PCSK9 inhibitors on coronary plaque composition and morphology is limited. METHODS In this open-label randomized study, eligible patients with intermediate coronary lesions and elevated LDL cholesterol values were randomized to either alirocumab 75 mg Q2W plus statin (atorvastatin 20 mg/day or rosuvastatin 10 mg/day) therapy or standard care. Optical coherence tomography (OCT) assessments for target lesions were obtained at baseline and at 36 weeks of follow-up. RESULTS LDL cholesterol levels were significantly decreased in both the alirocumab and standard care arms, whereas the absolute reduction in LDL cholesterol was significantly greater in patients treated with alirocumab (1.72 ± 0.51 vs. 0.96 ± 0.59, P < 0.0001). Compared with standard care, the addition of alirocumab to statins was associated with significantly greater increases in minimum fibrous cap thickness (18.0 [10.8-29.2] μm vs 13.2 [7.4-18.6] μm; P = 0.029), greater increases in minimum lumen area (0.20[0.10-0.33] mm2 vs 0.13 [0.12-0.24] mm2; P = 0.006) and a greater diminution in maximum lipid arc (15.1̊ [7.8-24.5] vs. 8.4̊ [2.0-10.5]; P = 0.008). CONCLUSIONS The addition of alirocumab to statins can not only provide additional LDL cholesterol lowering effects but also have a potential role in promoting a more stable plaque phenotype. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04851769 . Registered 2 Mar 2019.
Collapse
MESH Headings
- Aged
- Antibodies, Monoclonal, Humanized/therapeutic use
- Anticholesteremic Agents/therapeutic use
- Atorvastatin/therapeutic use
- Cholesterol, LDL/antagonists & inhibitors
- Cholesterol, LDL/blood
- Coronary Artery Disease/blood
- Coronary Artery Disease/diagnostic imaging
- Coronary Artery Disease/drug therapy
- Coronary Artery Disease/pathology
- Drug Synergism
- Drug Therapy, Combination
- Female
- Follow-Up Studies
- Gene Expression
- Humans
- Male
- Middle Aged
- PCSK9 Inhibitors/therapeutic use
- Plaque, Atherosclerotic/blood
- Plaque, Atherosclerotic/diagnostic imaging
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/pathology
- Proprotein Convertase 9/blood
- Proprotein Convertase 9/genetics
- Rosuvastatin Calcium/therapeutic use
- Tomography, Optical Coherence
Collapse
Affiliation(s)
- Fei Gao
- Department of Cardiology, An Zhen Hospital, Capital Medical University, Anzhenli avenue, Chao Yang district, Beijing, 100029, China.
| | - Zhi Jian Wang
- Department of Cardiology, An Zhen Hospital, Capital Medical University, Anzhenli avenue, Chao Yang district, Beijing, 100029, China
| | - Xiao Teng Ma
- Department of Cardiology, An Zhen Hospital, Capital Medical University, Anzhenli avenue, Chao Yang district, Beijing, 100029, China
| | - Hua Shen
- Department of Cardiology, An Zhen Hospital, Capital Medical University, Anzhenli avenue, Chao Yang district, Beijing, 100029, China
| | - Li Xia Yang
- Department of Cardiology, An Zhen Hospital, Capital Medical University, Anzhenli avenue, Chao Yang district, Beijing, 100029, China
| | - Yu Jie Zhou
- Department of Cardiology, An Zhen Hospital, Capital Medical University, Anzhenli avenue, Chao Yang district, Beijing, 100029, China.
| |
Collapse
|
59
|
Computed tomography of coronary artery atherosclerosis: A review. J Med Imaging Radiat Sci 2021; 52:S19-S39. [PMID: 34479831 DOI: 10.1016/j.jmir.2021.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/29/2021] [Accepted: 08/06/2021] [Indexed: 11/23/2022]
Abstract
Coronary artery atherosclerosis resulting in ischemic cardiac disease is the leading cause of mortality in the United States. In symptomatic patients, invasive diagnostic methods like catheter angiography, intravascular ultrasound, or vascular endoscopy may be used. However, for primary prevention of atherosclerotic coronary artery disease in asymptomatic patients, non-invasive methods are more commonly utilized like stress imaging, single-photon emission computed tomography (SPECT) and coronary artery calcification scoring. Coronary computed tomographic angiography (CCTA) is an excellent diagnostic tool for detection of coronary artery plaque and ability to identify resultant stenoses with an excellent negative predictive value which can potentially result in optimal exclusion of the presence of coronary artery disease. Long term follow up after a negative CCTA has repeatedly demonstrated very low incidence of future adverse coronary events, attesting its predictive value. CCTA based management is associated with improved CAD outcome in stable angina. Coronary CTA is valuable in acute chest pain evaluation in the emergency department helping in better triage. CT perfusion and CT-FFR are both very promising tools for assessment of hemodynamic significance of coronary artery stenosis.
Collapse
|
60
|
Görich HM, Buss SJ, Emami M, Seitz S, Lossnitzer D, Fortner P, Baumann S, Brado M, Gückel F, Sokiranski R, Sommer A, Görich J, Andre F. Short-Time Changes in Coronary Artery Plaques Assessed by Follow-Up Coronary CT Angiography-Characteristics and Impact on Patient Management. Front Cardiovasc Med 2021; 8:691665. [PMID: 34434975 PMCID: PMC8380958 DOI: 10.3389/fcvm.2021.691665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/14/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Coronary artery disease (CAD) shows a chronic but heterogeneous clinical course. Coronary CT angiography (CTA) allows for the visualization of the entire coronary tree and the detection of early stages of CAD. The aim of this study was to assess short-time changes in non-calcified and mixed plaques and their clinical impact using coronary CTA in a real-world setting. Methods: Between 11/2014 and 07/2019, 6,701 patients had a coronary CTA with a third-generation dual-source CT, of whom 77 patients (57 males, 63.8 ± 10.8 years) with a chronic CAD received clinically indicated follow-up CTA. Non-calcified and mixed plaques were analyzed in 1,211 coronary segments. Patients were divided into groups: stable, progressive, or regressive plaques. Results: Within the follow-up period of 22.3 ± 10.4 months, 44 patients (58%) showed stable plaques, 27 (36%) showed progression, 5 (7%) showed regression. One patient was excluded due to an undetermined CAD course showing both, progressive and regressive plaques. Age did not differ significantly between groups. Patients with plaque regression were predominantly female (80 vs. 20%), whereas patients showing progression were mainly male (85 vs. 15%; p < 0.01 for both). Regression was only observed in patients with mild CAD or one-vessel disease. The follow-up CTA led to changes in patient management in the majority of subjects (n = 50; 66%). Conclusions: Changes in coronary artery plaques can be observed within a short period resulting in an adjustment of the clinical management in the majority of CAD patients. Follow-up coronary CTA renders the non-invasive assessment of plaque development possible and allows for an individualized diagnostics and therapy optimization.
Collapse
Affiliation(s)
- Hanna Maria Görich
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany.,Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sebastian J Buss
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany
| | - Mostafa Emami
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany
| | - Sebastian Seitz
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany
| | - Dirk Lossnitzer
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany.,First Department of Medicine - Cardiology, University Medical Center Mannheim, Mannheim, Germany
| | - Philipp Fortner
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany
| | - Stefan Baumann
- First Department of Medicine - Cardiology, University Medical Center Mannheim, Mannheim, Germany
| | - Matthias Brado
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany
| | - Friedemann Gückel
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany
| | - Roman Sokiranski
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany
| | - André Sommer
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany
| | - Johannes Görich
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany
| | - Florian Andre
- Radiology Center Sinsheim-Eberbach-Erbach-Walldorf-Heidelberg, Heidelberg, Germany.,Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
61
|
Lv R, Maehara A, Matsumura M, Wang L, Zhang C, Huang M, Guo X, Samady H, Giddens DP, Zheng J, Mintz GS, Tang D. Using Optical Coherence Tomography and Intravascular Ultrasound Imaging to Quantify Coronary Plaque Cap Stress/Strain and Progression: A Follow-Up Study Using 3D Thin-Layer Models. Front Bioeng Biotechnol 2021; 9:713525. [PMID: 34497800 PMCID: PMC8419245 DOI: 10.3389/fbioe.2021.713525] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
Accurate plaque cap thickness quantification and cap stress/strain calculations are of fundamental importance for vulnerable plaque research. To overcome uncertainties due to intravascular ultrasound (IVUS) resolution limitation, IVUS and optical coherence tomography (OCT) coronary plaque image data were combined together to obtain accurate and reliable cap thickness data, stress/strain calculations, and reliable plaque progression predictions. IVUS, OCT, and angiography baseline and follow-up data were collected from nine patients (mean age: 69; m: 5) at Cardiovascular Research Foundation with informed consent obtained. IVUS and OCT slices were coregistered and merged to form IVUS + OCT (IO) slices. A total of 114 matched slices (IVUS and OCT, baseline and follow-up) were obtained, and 3D thin-layer models were constructed to obtain stress and strain values. A generalized linear mixed model (GLMM) and least squares support vector machine (LSSVM) method were used to predict cap thickness change using nine morphological and mechanical risk factors. Prediction accuracies by all combinations (511) of those predictors with both IVUS and IO data were compared to identify optimal predictor(s) with their best accuracies. For the nine patients, the average of minimum cap thickness from IVUS was 0.17 mm, which was 26.08% lower than that from IO data (average = 0.23 mm). Patient variations of the individual errors ranged from ‒58.11 to 20.37%. For maximum cap stress between IO and IVUS, patient variations of the individual errors ranged from ‒30.40 to 46.17%. Patient variations of the individual errors of maximum cap strain values ranged from ‒19.90 to 17.65%. For the GLMM method, the optimal combination predictor using IO data had AUC (area under the ROC curve) = 0.926 and highest accuracy = 90.8%, vs. AUC = 0.783 and accuracy = 74.6% using IVUS data. For the LSSVM method, the best combination predictor using IO data had AUC = 0.838 and accuracy = 75.7%, vs. AUC = 0.780 and accuracy = 69.6% using IVUS data. This preliminary study demonstrated improved plaque cap progression prediction accuracy using accurate cap thickness data from IO slices and the differences in cap thickness, stress/strain values, and prediction results between IVUS and IO data. Large-scale studies are needed to verify our findings.
Collapse
Affiliation(s)
- Rui Lv
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Akiko Maehara
- The Cardiovascular Research Foundation, Columbia University, New York, NY, United States
| | - Mitsuaki Matsumura
- The Cardiovascular Research Foundation, Columbia University, New York, NY, United States
| | - Liang Wang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Caining Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Mengde Huang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Xiaoya Guo
- School of Science, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Habib Samady
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Don. P. Giddens
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Jie Zheng
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, United States
| | - Gary S. Mintz
- The Cardiovascular Research Foundation, Columbia University, New York, NY, United States
| | - Dalin Tang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Mathematical Sciences Department, Worcester Polytechnic Institute, Worcester, MA, United States
| |
Collapse
|
62
|
Refahiyat L, Madder RD. In Search of a Blood Marker for Vulnerable Plaque. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2021; 32:11-12. [PMID: 34412990 DOI: 10.1016/j.carrev.2021.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Lahdan Refahiyat
- Frederik Meijer Heart & Vascular Institute, Spectrum Health, Grand Rapids, MI, United States of America.
| | - Ryan D Madder
- Frederik Meijer Heart & Vascular Institute, Spectrum Health, Grand Rapids, MI, United States of America
| |
Collapse
|
63
|
Kurihara O, Takano M, Miyauchi Y, Mizuno K, Shimizu W. Vulnerable atherosclerotic plaque features: findings from coronary imaging. J Geriatr Cardiol 2021; 18:577-584. [PMID: 34404993 PMCID: PMC8352771 DOI: 10.11909/j.issn.1671-5411.2021.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Pathological studies have suggested that features of vulnerable atherosclerotic plaques likely to progress and lead to acute cardiovascular events have specific characteristics. Given the progress of intravascular coronary imaging technology, some large prospective studies have detected features of vulnerable atherosclerotic plaques using these imaging modalities. However, the rate of cardiovascular events, such as acute coronary syndrome, has been found to be considerably reduced in the limited follow-up period available in the statin era. Additionally, not all disrupted plaques lead to thrombus formation with clinical presentation. If sub-occlusive or occlusive thrombus formation does not occur, a thrombus on a disrupted plaque will organize without any symptoms, forming a "healed plaque". Although vulnerable plaque detection using intracoronary imaging is focused on "thin-cap fibroatheroma" leading to plaque rupture, superficial plaque erosion is increasingly recognized; however, the underlying mechanism of thrombus formation on eroded plaques is not well understood. One of intravascular imaging, optical coherence tomography (OCT) has the highest image resolution and has enabled detailed characterization of the plaque in vivo. Here, we reviewed the status and limitations of intravascular imaging in terms of detecting vulnerable plaque through mainly OCT studies. We suggested that vulnerable plaque should be reconsidered in terms of eroded plaque and healed plaque and that both plaque and circulating blood should be assessed in greater detail accordingly.
Collapse
Affiliation(s)
- Osamu Kurihara
- Cardiovascular Center, Chiba Hokusoh Hospital, Nippon Medical School, Chiba, Japan
| | - Masamichi Takano
- Cardiovascular Center, Chiba Hokusoh Hospital, Nippon Medical School, Chiba, Japan
| | - Yasushi Miyauchi
- Cardiovascular Center, Chiba Hokusoh Hospital, Nippon Medical School, Chiba, Japan
| | | | - Wataru Shimizu
- Division of Cardiology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
64
|
Mozzini C, Pagani M. Cardiovascular Diseases: Consider Netosis. Curr Probl Cardiol 2021; 47:100929. [PMID: 34315622 DOI: 10.1016/j.cpcardiol.2021.100929] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 06/21/2021] [Indexed: 01/18/2023]
Abstract
Neutrophil extracellular traps (NETs) are net-like chromatin fibers that are released from dying neutrophils during infections. NETs are a sort of scaffold, ideal to retain microbes. The main function of NETs is the trapping and killing pathogens, as such as bacteria, fungi, viruses (including SARS-CoV-2) and protozoa. The death of neutrophils via NETs formation is called "NETosis." Nevertheless, recent studies suggest that NETosis is involved in several diseases, other than infections. Very recently, it has been shown that NETs formation contributes to venous thromboembolism but also to atherosclerosis progression, creating a link between venous and arterial thrombosis. The presence of NETs in the luminal portion of human atherosclerotic vessels and coronary specimens obtained from patients after acute myocardial infarction has been detected. This review provides evidence of the most important updates about the role of NETs in myocardial infarction, in heart failure and in the process of atherosclerosis itself. The prognostic significance of NETs-related markers in cardiovascular diseases will be discussed, in order to assess targeted therapeutic strategies.
Collapse
Affiliation(s)
- Chiara Mozzini
- Department of Medicine, Section of Internal Medicine, Carlo Poma Hospital, Mantova Italy.
| | - Mauro Pagani
- Department of Medicine, Section of Internal Medicine, Carlo Poma Hospital, Mantova Italy
| |
Collapse
|
65
|
Krittanawong C, Hahn J, Kayani W, Jneid H. Fibrinolytic Therapy in Patients with Acute ST-elevation Myocardial Infarction. Interv Cardiol Clin 2021; 10:381-390. [PMID: 34053624 DOI: 10.1016/j.iccl.2021.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fibrinolytic agents provide an important alternative therapeutic strategy in individuals presenting with ST-elevation myocardial infarction (STEMI). Ultimately, primary percutaneous coronary intervention (PCI) is the preferred reperfusion strategy for most patients with STEMI, including elderly patients and patients with coronavirus disease 2019 (COVID-19) infection. Fibrinolytic therapy should always be considered when timely primary PCI cannot be delivered appropriately. Clinicians should promptly recognize the signs of fibrinolytic therapy failure and consider rescue PCI. When fibrinolytics are used, coronary angiography and revascularization should not be conducted within the initial 3 hours after fibrinolytic administration.
Collapse
Affiliation(s)
- Chayakrit Krittanawong
- Section of Cardiology, Baylor College of Medicine, 7200 Cambridge Street, Houston, TX 77030, USA
| | - Joshua Hahn
- Section of Cardiology, Baylor College of Medicine, 7200 Cambridge Street, Houston, TX 77030, USA
| | - Waleed Kayani
- Section of Cardiology, Baylor College of Medicine, 7200 Cambridge Street, Houston, TX 77030, USA
| | - Hani Jneid
- Section of Cardiology, Baylor College of Medicine, 7200 Cambridge Street, Houston, TX 77030, USA; Interventional Cardiology Fellowship Program, Interventional Cardiology Research, Baylor College of Medicine, Interventional Cardiology, The Michael E. DeBakey VA Medical Center, MEDVAMC - 2002 Holcombe Boulevard, Cardiology 3C-320C, Houston, TX 77030, USA.
| |
Collapse
|
66
|
Abstract
The endothelium is a crucial regulator of vascular homeostasis by controlling barrier integrity as well acting as an important signal transducer, thereby illustrating that endothelial cells are not inert cells. In the context of atherosclerosis, this barrier function is impaired and endothelial cells become activated, resulting in the upregulation of adhesion molecules, secretion of cytokines and chemokines and internalization of integrins. Finally, this leads to increased vessel permeability, thereby facilitating leukocyte extravasation as well as fostering a pro-inflammatory environment. Additionally, activated endothelial cells can form migrating tip cells and proliferative stalk cells, resulting in the formation of new blood vessels. Emerging evidence has accumulated indicating that cellular metabolism is crucial in fueling these pro-atherosclerotic processes, including neovascularization and inflammation, thereby contributing to plaque progression and altering plaque stability. Therefore, further research is necessary to unravel the complex mechanisms underlying endothelial cell metabolic changes, and exploit this knowledge for finding and developing potential future therapeutic strategies. In this review we discuss the metabolic alterations endothelial cells undergo in the context of inflammation and atherosclerosis and how this relates to changes in endothelial functioning. Finally, we will describe several metabolic targets that are currently being used for therapeutic interventions.
Collapse
|
67
|
Gao B, Xu J, Zhou J, Zhang H, Yang R, Wang H, Huang J, Yan F, Luo Y. Multifunctional pathology-mapping theranostic nanoplatforms for US/MR imaging and ultrasound therapy of atherosclerosis. NANOSCALE 2021; 13:8623-8638. [PMID: 33929480 DOI: 10.1039/d1nr01096d] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Atherosclerotic thrombosis is the leading cause of most life-threatening cardiovascular diseases (CVDs), particularly as a result of rupture or erosion of vulnerable plaques. Rupture or erosion-prone plaques are quite different in cellular composition and immunopathology, requiring different treatment strategies. The current imaging technology cannot distinguish the types of vulnerable plaques, and thus empirical treatment is still applied to all without a tailored and precise treatment. Herein, we propose a novel strategy called "Multifunctional Pathology-mapping Theranostic Nanoplatform (MPmTN)" for the tailored treatment of plaques based on the pathological classification. MPmTNs are made up of poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs), containing contrast imaging materials Fe3O4 and perfluoropentane (PFP), and coated with specific plaque-targeted peptides PP1 and cyclic RGD. The PFP encapsulated inside the MPmTN can undergo a phase change from nanodroplets to gas microbubbles under therapeutic ultrasound (TUS) exposure. The acoustic and biological effects induced by TUS and disruption of microbubbles may further promote therapeutic effects. Hypothetically, MPmTN NPs can target the rupture-prone plaque via the binding of PP1 to class A scavenger receptors (SR-A) on macrophages, induce the apoptosis due to TUS exposure and thus reduce the chronic soakage of inflammatory cells. The MPmTN NPs can also target the erosion-prone plaque through the binding of cRGD to glycoprotein (GP) IIb/IIIa on activated platelets and promote platelet disaggregation under TUS exposure. Therefore, MPmTNs may work as a multifunctional pathology-mapping therapeutic agent. Our in vitro results show that the MPmTN with PP1 and cRGD peptides had a high binding affinity both for activated macrophages and blood clots. Under TUS exposure, the MPmTN could effectively induce macrophage apoptosis, destroy thrombus and exhibit good imaging properties for ultrasound (US) and MRI. In apoE-/- mice, MPmTNs can selectively accumulate at the plaque site and reduce the T2-weighted signal. The apoptosis of macrophages and disaggregation of activated platelets on the plaques were also confirmed in vivo. In summary, this study provides a potential strategy for a tailored treatment of vulnerable plaques based on their pathological nature and a multimodal imaging tool for the risk stratification and assessment of therapeutic efficacy.
Collapse
Affiliation(s)
- Binyang Gao
- Department of Ultrasound, Laboratory of Ultrasound Imaging and Drug, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Li J, Montarello NJ, Hoogendoorn A, Verjans JW, Bursill CA, Peter K, Nicholls SJ, McLaughlin RA, Psaltis PJ. Multimodality Intravascular Imaging of High-Risk Coronary Plaque. JACC Cardiovasc Imaging 2021; 15:145-159. [PMID: 34023267 DOI: 10.1016/j.jcmg.2021.03.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/01/2021] [Accepted: 03/22/2021] [Indexed: 01/13/2023]
Abstract
The majority of coronary atherothrombotic events presenting as myocardial infarction (MI) occur as a result of plaque rupture or erosion. Understanding the evolution from a stable plaque into a life-threatening, high-risk plaque is required for advancing clinical approaches to predict atherothrombotic events, and better treat coronary atherosclerosis. Unfortunately, none of the coronary imaging approaches used in clinical practice can reliably predict which plaques will cause an MI. Currently used imaging techniques mostly identify morphological features of plaques, but are not capable of detecting essential molecular characteristics known to be important drivers of future risk. To address this challenge, engineers, scientists, and clinicians have been working hand-in-hand to advance a variety of multimodality intravascular imaging techniques, whereby 2 or more complementary modalities are integrated into the same imaging catheter. Some of these have already been tested in early clinical studies, with other next-generation techniques also in development. This review examines these emerging hybrid intracoronary imaging techniques and discusses their strengths, limitations, and potential for clinical translation from both an engineering and clinical perspective.
Collapse
Affiliation(s)
- Jiawen Li
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, Australia; Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
| | - Nicholas J Montarello
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide, Australia
| | - Ayla Hoogendoorn
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, Australia; Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia; Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Johan W Verjans
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, Australia; Department of Cardiology, Central Adelaide Local Health Network, Adelaide, Australia; Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Christina A Bursill
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, Australia; Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia; Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | | | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Robert A McLaughlin
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, Australia; Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, Australia
| | - Peter J Psaltis
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Department of Cardiology, Central Adelaide Local Health Network, Adelaide, Australia; Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.
| |
Collapse
|
69
|
Shimamura K, Kubo T, Akasaka T. Evaluation of coronary plaques and atherosclerosis using optical coherence tomography. Expert Rev Cardiovasc Ther 2021; 19:379-386. [PMID: 33823735 DOI: 10.1080/14779072.2021.1914588] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Coronary angiography (CAG) is the standard modality for assessing coronary stenosis; however, it has limitations in assessing coronary plaque morphology. Optical coherence tomography (OCT) is a high-resolution (10-20 μm) light-based intravascular imaging technique that can identify more detailed coronary plaque morphology compared to other intravascular imaging modalities. OCT is remarkable for characterizing fibrous, fibrocalcific, and lipid-rich plaques. The capabilities of OCT are well suited for discriminating three types of unstable plaque morphologies underlying coronary thrombosis, such as plaque rupture, erosion, and calcified nodules. The high resolution of OCT makes it possible to identify important features of vulnerable plaques, such as thin-cap (<65 μm thick) fibroatheroma, macrophages, vasa vasorum, and cholesterol crystals.Areas covered: This review summarizes the clinical impact of OCT and its efficacy in identifying plaque components and morphological features associated with plaque vulnerability.Expertopinion: The unique properties of OCT as a tool for investigating high-risk lesions have greatly contributed to a better understanding of plaque vulnerability. Consequently, OCT has led to significant changes in medical treatment and percutaneous coronary intervention strategies for acute coronary syndrome. Further development and investigation of OCT are necessary to better predict and manage acute coronary events in the future.
Collapse
Affiliation(s)
- Kunihiro Shimamura
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takashi Kubo
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takashi Akasaka
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
70
|
Zhang JY, Chen QQ, Li J, Zhang L, Qi LW. Neuraminidase 1 and its Inhibitors from Chinese Herbal Medicines: An Emerging Role for Cardiovascular Diseases. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:843-862. [PMID: 33827385 DOI: 10.1142/s0192415x21500403] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Neuraminidase, also known as sialidase, is ubiquitous in animals and microorganisms. It is predominantly distributed in the cell membrane, cytoplasmic vesicles, and lysosomes. Neuraminidase generally recognizes the sialic acid glycosidic bonds at the ends of glycoproteins or glycolipids and enzymatically removes sialic acid. There are four types of neuraminidases, named as Neu1, Neu2, Neu3, and Neu4. Among them, Neu1 is the most abundant in mammals. Recent studies have revealed the involvement of Neu1 in several diseases, including cardiovascular diseases, diabetes, cancers, and neurological disorders. In this review, we center the attention to the role of Neu1 in cardiovascular diseases, including atherosclerosis, ischemic myocardial injury, cerebrovascular disease, congenital heart disease, and pulmonary embolism. We also summarize inhibitors from Chinese herbal medicines (CHMs) in inhibiting virus neuraminidase or human Neu1. Many Chinese herbs and Chinese herb preparations, such as Lonicerae Japonicae Flos, Scutellariae Radix, Yupingfeng San, and Huanglian Jiedu Decoction, have neuraminidase inhibitory activity. We hope to highlight the emerging role of Neu1 in humans and potentially titillate interest for further studies in this area.
Collapse
Affiliation(s)
- Jun-Yuan Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Qian-Qian Chen
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jia Li
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Lei Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Lian-Wen Qi
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China.,Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
71
|
Guo X, Maehara A, Matsumura M, Wang L, Zheng J, Samady H, Mintz GS, Giddens DP, Tang D. Predicting plaque vulnerability change using intravascular ultrasound + optical coherence tomography image-based fluid-structure interaction models and machine learning methods with patient follow-up data: a feasibility study. Biomed Eng Online 2021; 20:34. [PMID: 33823858 PMCID: PMC8025351 DOI: 10.1186/s12938-021-00868-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/13/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Coronary plaque vulnerability prediction is difficult because plaque vulnerability is non-trivial to quantify, clinically available medical image modality is not enough to quantify thin cap thickness, prediction methods with high accuracies still need to be developed, and gold-standard data to validate vulnerability prediction are often not available. Patient follow-up intravascular ultrasound (IVUS), optical coherence tomography (OCT) and angiography data were acquired to construct 3D fluid-structure interaction (FSI) coronary models and four machine-learning methods were compared to identify optimal method to predict future plaque vulnerability. METHODS Baseline and 10-month follow-up in vivo IVUS and OCT coronary plaque data were acquired from two arteries of one patient using IRB approved protocols with informed consent obtained. IVUS and OCT-based FSI models were constructed to obtain plaque wall stress/strain and wall shear stress. Forty-five slices were selected as machine learning sample database for vulnerability prediction study. Thirteen key morphological factors from IVUS and OCT images and biomechanical factors from FSI model were extracted from 45 slices at baseline for analysis. Lipid percentage index (LPI), cap thickness index (CTI) and morphological plaque vulnerability index (MPVI) were quantified to measure plaque vulnerability. Four machine learning methods (least square support vector machine, discriminant analysis, random forest and ensemble learning) were employed to predict the changes of three indices using all combinations of 13 factors. A standard fivefold cross-validation procedure was used to evaluate prediction results. RESULTS For LPI change prediction using support vector machine, wall thickness was the optimal single-factor predictor with area under curve (AUC) 0.883 and the AUC of optimal combinational-factor predictor achieved 0.963. For CTI change prediction using discriminant analysis, minimum cap thickness was the optimal single-factor predictor with AUC 0.818 while optimal combinational-factor predictor achieved an AUC 0.836. Using random forest for predicting MPVI change, minimum cap thickness was the optimal single-factor predictor with AUC 0.785 and the AUC of optimal combinational-factor predictor achieved 0.847. CONCLUSION This feasibility study demonstrated that machine learning methods could be used to accurately predict plaque vulnerability change based on morphological and biomechanical factors from multi-modality image-based FSI models. Large-scale studies are needed to verify our findings.
Collapse
Affiliation(s)
- Xiaoya Guo
- School of Science, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China.
- Department of Mathematics, Southeast University, Nanjing, 210096, China.
| | - Akiko Maehara
- The Cardiovascular Research Foundation, Columbia University, New York, NY, 10022, USA
| | - Mitsuaki Matsumura
- The Cardiovascular Research Foundation, Columbia University, New York, NY, 10022, USA
| | - Liang Wang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jie Zheng
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, 63110, USA
| | - Habib Samady
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30307, USA
| | - Gary S Mintz
- The Cardiovascular Research Foundation, Columbia University, New York, NY, 10022, USA
| | - Don P Giddens
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30307, USA
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Dalin Tang
- Department of Mathematics, Southeast University, Nanjing, 210096, China.
- Mathematical Sciences Department, Worcester Polytechnic Institute, Worcester, MA, 01609, USA.
| |
Collapse
|
72
|
Flouda K, Mercer J, Davies MJ, Hawkins CL. Role of myeloperoxidase-derived oxidants in the induction of vascular smooth muscle cell damage. Free Radic Biol Med 2021; 166:165-177. [PMID: 33631301 DOI: 10.1016/j.freeradbiomed.2021.02.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/14/2021] [Indexed: 01/12/2023]
Abstract
Myeloperoxidase (MPO) is released by activated immune cells and forms the oxidants hypochlorous acid (HOCl) and hypothiocyanous acid (HOSCN) from the competing substrates chloride and thiocyanate. MPO and the overproduction of HOCl are strongly linked with vascular cell dysfunction and inflammation in atherosclerosis. HOCl is highly reactive and causes marked cell dysfunction and death, whereas data with HOSCN are conflicting, and highly dependent on the nature of the cell type. In this study we have examined the reactivity of HOCl and HOSCN with human coronary artery smooth muscle cells (HCASMC), given the key role of this cell type in maintaining vascular function. HOCl reacts rapidly with the cells, resulting in extensive cell death by both necrosis and apoptosis, and increased levels of intracellular calcium. In contrast, HOSCN reacts more slowly, with cell death occurring only after prolonged incubation, and in the absence of the accumulation of intracellular calcium. Exposure of HCASMC to HOCl also influences mitochondrial respiration, decreases glycolysis, lactate release, the production of ATP, cellular thiols and glutathione levels. These changes occurred to varying extents on exposure of the cells to HOSCN, where evidence was also obtained for the reversible modification of cellular thiols. HOCl also induced alterations in the mRNA expression of multiple inflammatory and phenotypic genes. Interestingly, the extent and nature of these changes was highly dependent on the specific cell donor used, with more marked effects observed in cells isolated from diseased compared to healthy vessels. Overall, these data provide new insight into pathways promoting vascular dysfunction during chronic inflammation, support the use of thiocyanate as a means to modulate MPO-induced cellular damage in atherosclerosis.
Collapse
Affiliation(s)
- Konstantina Flouda
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - John Mercer
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, United Kingdom
| | - Michael J Davies
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark.
| |
Collapse
|
73
|
Mozzini C, Girelli D, Cominacini L, Soresi M. An Exploratory Look at Bicuspid Aortic Valve (Bav) Aortopathy: Focus on Molecular and Cellular Mechanisms. Curr Probl Cardiol 2021; 46:100425. [PMID: 31097209 DOI: 10.1016/j.cpcardiol.2019.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 01/12/2023]
Abstract
Bicuspid aortic valve (BAV) is the most common congenital heart malformation. BAV patients are at increased risk for aortic valve disease (stenosis/regurgitation), infective endocarditis, thrombi formation and, in particular, aortic dilatation, aneurysm and dissection. This review aims at exploring the possible interplay among genetics, extracellular matrix remodeling, abnormal signaling pathways, oxidative stress and inflammation in contributing to BAV-associated aortopathy (BAV-A-A). Novel circulating biomarkers have been proposed as diagnostic tools able to improve risk stratification in BAV-A-A. However, to date, the precise molecular and cellular mechanisms that lead to BAV-A-A remain unknown. Genetic, hemodynamic and cardiovascular risk factors have been implicated in the development and progression of BAV-A-A. Oxidative stress may also play a role, similarly to what observed in atherosclerosis and vulnerable plaque formation. The identification of common pathways between these 2 conditions may provide a platform for future therapeutic solutions.
Collapse
|
74
|
Characteristics of culprit lesion in patients with non-ST-elevation myocardial infarction and improvement of diagnostic utility using dual energy cardiac CT. Int J Cardiovasc Imaging 2021; 37:1781-1788. [PMID: 33502653 DOI: 10.1007/s10554-020-02141-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
AIMS The aim of the study was to identify the characteristics of the culprit lesions compared to non-culprit lesions in patients with non-ST-elevation-myocardial infarction using dual energy computed tomography (DECT). METHODS AND RESULTS In 29 patients, we identified 29 culprit lesions and 227 non-culprit lesions. Quantitative values such as the effective atomic number (effective-Z) and Hounsfield Units (HU) values were measured. Furthermore, all the lesions were characterised using characteristics such as composition (non-calcified, predominantly-non-calcified, predominantly-calcified, or calcified), presence of spotty calcification, remodelling index, and napkin ring sign. The mean effective-Z and HU values were significantly lower in culprit lesions than in non-culprit lesions (8.99 ± 1.21 vs 9.79 ± 1.52; p = 0.0066 and 87.41 ± 84.97 vs. 154.45 ± 176.13; p = 0.0447). The culprit lesions had a higher frequency of non-calcified plaques and predominantly non-calcified plaques, and were with a greater presence of napkin ring signs in comparison with non-culprit lesions. There were no differences in the presence of spotty calcification or remodelling index. By adding effective-Z to plaque characteristics such as non-calcified, positive remodelling, spotty calcification, and napkin rings we observed a significant increased sensitivity of detecting culprit lesions (65.5% vs.44.8%), but no significant changes in area under curve (AUC). CONCLUSION The use of DECT adds new information of the plaque composition expressed by the effective-Z, which differs significantly in culprit lesions in comparison with non-culprit lesions. The use of the effective-Z improves the diagnostic sensitivity in detection of culprit lesions.
Collapse
|
75
|
Merkulova IN, Shariya MA, Mironov VM, Shabanova MS, Veselova TN, Gaman SA, Barysheva NA, Shakhnovich RM, Zhukova NI, Sukhinina TS, Staroverov II, Ternovoy SK. [Computed Tomography Coronary Angiography Possibilities in "High Risk" Plaque Identification in Patients with non-ST-Elevation Acute Coronary Syndrome: Comparison with Intravascular Ultrasound]. ACTA ACUST UNITED AC 2021; 60:64-75. [PMID: 33522469 DOI: 10.18087/cardio.2020.12.n1304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/26/2020] [Indexed: 11/18/2022]
Abstract
Aim To evaluate structural characteristics of atherosclerotic plaques (ASP) by coronary computed tomography arteriography (CCTA) and intravascular ultrasound (IVUS).Material and methods This study included 37 patients with acute coronary syndrome (ACS). 64-detector-row CCTA, coronarography, and grayscale IVUS were performed prior to coronary stenting. The ASP length and burden, remodeling index (RI), and known CT signs of unstable ASP (presence of dot calcification, positive remodeling of the artery in the ASP area, irregular plaque contour, presence of a peripheral high-density ring and a low-density patch in the ASP). The ASP type and signs of rupture or thrombosis were determined by IVUS.Results The IVUS study revealed 45 unstable ASP (UASP), including 25 UASP with rupture and 20 thin-cap fibroatheromas (TCFA), and 13 stable ASP (SASP). No significant differences were found between distribution of TCFA and ASP with rupture among symptom-associated plaques (SAP, n=28) and non-symptom-associated plaques (NSAP, n=30). They were found in 82.1 and 73.3 % of cases, respectively (p>0.05), which indicated generalization of the ASP destabilization process in the coronary circulation. However, the incidence of mural thrombus was higher for SAP (53.5 and 16.6 % of ASP, respectively; p<0.001). There was no difference between UASP and SASP in the incidence of qualitative ASP characteristics or in values of quantitative ASP characteristics, including known signs of instability, except for the irregular contour, which was observed in 92.9 % of UASP and 46.1 % of SASP (p=0.0007), and patches with X-ray density ≤46 HU, which were detected in 83.3 % of UASP and 46.1 % of SASP (р=0.01). The presence of these CT criteria 11- and 7-fold increased the likelihood of unstable ASP (odd ratio (OR), 11.1 at 95 % confidence interval (CI), from 2.24 to 55.33 and OR, 7.0 at 95 % CI, from 5.63 to 8.37 for the former and the latter criterion, respectively).Conclusion According to IVUS data, two X-ray signs are most characteristic for UASP, the irregular contour and a patch with X-ray density ≤46 HU. The presence of these signs 11- and 7-fold, respectively, increases the likelihood of unstable ASP.
Collapse
Affiliation(s)
- I N Merkulova
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - M A Shariya
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - V M Mironov
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - M S Shabanova
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - T N Veselova
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - S A Gaman
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - N A Barysheva
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - R M Shakhnovich
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - N I Zhukova
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - T S Sukhinina
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - I I Staroverov
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| | - S K Ternovoy
- Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Healthcare Russian Federation, Moscow
| |
Collapse
|
76
|
Li Z, Wang Y, Wu X, Liu X, Huang S, He Y, Liu S, Ren L. Studying the Factors of Human Carotid Atherosclerotic Plaque Rupture, by Calculating Stress/Strain in the Plaque, Based on CEUS Images: A Numerical Study. Front Neuroinform 2020; 14:596340. [PMID: 33324188 PMCID: PMC7721669 DOI: 10.3389/fninf.2020.596340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 09/11/2020] [Indexed: 01/08/2023] Open
Abstract
Carotid plaque neovascularization is one of the major factors for the classification of vulnerable plaque, but the axial force effects of the pulsatile blood flow on the plaque with neovessel and intraplaque hemorrhage was unclear. Together with the severity of stenosis, the fibrous cap thickness, large lipid core, and the neovascularization followed by intraplaque hemorrhage (IPH) have been regarded as high-risk features of plaque rupture. In this work, the effects of these factors were evaluated on the progression and rupture of the carotid atherosclerotic plaques. Five geometries of carotid artery plaque were developed based on contrast-enhanced ultrasound (CEUS) images, which contain two types of neovessel and IPH, and geometry without neovessel and IPH. A one-way fluid-structure interaction model was applied to compute the maximum principal stress and strain in the plaque. For that hyper-elastic and non-linear material, Yeoh 3rd Order strain energy density function was used for components of the plaque. The simulation results indicated that the maximum principal stress of plaque in the carotid artery was higher when the degree of the luminal stenosis increased and the thickness of the fibrous cap decreased. The neovessels within the plaque could introduce a 2.5% increments of deformation in the plaque under the pulsatile blood flow pressure. The IPH also contributed to the increased risk of plaque rupture that a gain of stress was 8.983, 14.526, and 34.47 kPa for the plaque with 50, 65, and 75%, respectively, when comparing stress in the plaque with IPH distributed at the middle to the shoulder of the plaque. In conclusion, neovascularization in the plaque could reduce the stability of the plaque by increasing the stress within the plaque. Also, the risk of plaque rupture increased when large luminal stenosis, thin fibrous cap, and IPH were observed.
Collapse
Affiliation(s)
- Zhenzhou Li
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yongfeng Wang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, China
| | - Xinyin Wu
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Xin Liu
- Guangdong Academy Research on Virtual Reality (VR) Industry, Foshan University, Foshan, China
| | - Shanshan Huang
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yi He
- Department of Neurosurgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Shuyu Liu
- School of Pharmacy, Sun Yat-sen University, Guangzhou, China
| | - Lijie Ren
- Department of Neurology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
77
|
Majeed K, Bellinge JW, Butcher SC, Alcock R, Spiro J, Playford D, Hillis GS, Newby DE, Mori TA, Francis R, Schultz CJ. Coronary 18F-sodium fluoride PET detects high-risk plaque features on optical coherence tomography and CT-angiography in patients with acute coronary syndrome. Atherosclerosis 2020; 319:142-148. [PMID: 33358367 DOI: 10.1016/j.atherosclerosis.2020.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/08/2020] [Accepted: 12/11/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS 18F-Sodium Fluoride Positron Emission Tomography (18F-NaF PET) non-invasively detects micro-calcification activity, the earliest stage of atherosclerotic arterial calcification. We studied the association between coronary 18F-NaF uptake and high-risk plaque features on intra-coronary optical coherence tomography (OCT) and CT-angiography (CTCA) and the potential application to patient-level risk stratification. METHODS Sixty-two prospectively recruited patients with acute coronary syndrome (ACS) underwent multi-vessel OCT, 18F-NaF PET and CTCA. The maximum tissue to background ratio (TBRmax = standardised uptake value (SUV)max/SUVbloodpool) was measured in each coronary segment on 18F-NaF PET scans. High-risk plaque features on OCT and CTCA were compared in matched coronary segments. The number of patients testing positive (>2SD above the normal range) for micro-calcification activity was determined. RESULTS In 62 patients (age, mean ± standard deviation (SD) = 61 ± 9 years, 85% male) the coronary segments with elevated 18F-NaF uptake had higher lipid arc (LA) (median [25th-75th centile]: 74° [35°-117°] versus 48° [15°-83°], p=0.021), higher prevalence of macrophages [n(%): 37 (62%) versus 89 (39%), p=0.008] and lower plaque free wall (PFW) (50° [7°-110°] versus 94° [34°-180°], p=0.027) on OCT, and a higher total plaque burden (p=0.011) and higher dense calcified plaque burden (p= 0.001) on CTCA, when compared with 18F-NaF negative segments. Patients grouped by increasing number of coronary lesions positive for microcalcification activity (0,1, ≥2) showed decreasing plaque free wall, increasing calcification and increasing macrophages on OCT (respectively p=0.008, p < 0.001 and p=0.028). CONCLUSIONS 18F-NaF uptake is associated with high-risk plaque features on OCT and CTCA in a per-segment and per-patient analysis in subjects hospitalized for ACS.
Collapse
Affiliation(s)
- Kamran Majeed
- Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia; Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Jamie W Bellinge
- Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia; Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Steele C Butcher
- School of Medicine, University of Notre Dame, Fremantle, Western Australia, Australia
| | - Richard Alcock
- Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Jon Spiro
- Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - David Playford
- School of Medicine, University of Notre Dame, Fremantle, Western Australia, Australia
| | - Graham S Hillis
- Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia; Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - David E Newby
- Centre for Cardiovascular Science, Clinical Research Imaging Centre, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Trevor A Mori
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Roslyn Francis
- Medical School, University of Western Australia, Perth, Western Australia, Australia; Nuclear Medicine Department, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Carl J Schultz
- Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia; Medical School, University of Western Australia, Perth, Western Australia, Australia.
| |
Collapse
|
78
|
Lv R, Maehara A, Matsumura M, Wang L, Wang Q, Zhang C, Guo X, Samady H, Giddens DP, Zheng J, Mintz GS, Tang D. Using optical coherence tomography and intravascular ultrasound imaging to quantify coronary plaque cap thickness and vulnerability: a pilot study. Biomed Eng Online 2020; 19:90. [PMID: 33256759 PMCID: PMC7706023 DOI: 10.1186/s12938-020-00832-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/17/2020] [Indexed: 11/11/2022] Open
Abstract
Background Detecting coronary vulnerable plaques in vivo and assessing their vulnerability have been great challenges for clinicians and the research community. Intravascular ultrasound (IVUS) is commonly used in clinical practice for diagnosis and treatment decisions. However, due to IVUS limited resolution (about 150–200 µm), it is not sufficient to detect vulnerable plaques with a threshold cap thickness of 65 µm. Optical Coherence Tomography (OCT) has a resolution of 15–20 µm and can measure fibrous cap thickness more accurately. The aim of this study was to use OCT as the benchmark to obtain patient-specific coronary plaque cap thickness and evaluate the differences between OCT and IVUS fibrous cap quantifications. A cap index with integer values 0–4 was also introduced as a quantitative measure of plaque vulnerability to study plaque vulnerability. Methods Data from 10 patients (mean age: 70.4; m: 6; f: 4) with coronary heart disease who underwent IVUS, OCT, and angiography were collected at Cardiovascular Research Foundation (CRF) using approved protocol with informed consent obtained. 348 slices with lipid core and fibrous caps were selected for study. Convolutional Neural Network (CNN)-based and expert-based data segmentation were performed using established methods previously published. Cap thickness data were extracted to quantify differences between IVUS and OCT measurements. Results For the 348 slices analyzed, the mean value difference between OCT and IVUS cap thickness measurements was 1.83% (p = 0.031). However, mean value of point-to-point differences was 35.76%. Comparing minimum cap thickness for each plaque, the mean value of the 20 plaque IVUS-OCT differences was 44.46%, ranging from 2.36% to 91.15%. For cap index values assigned to the 348 slices, the disagreement between OCT and IVUS assignments was 25%. However, for the OCT cap index = 2 and 3 groups, the disagreement rates were 91% and 80%, respectively. Furthermore, the observation of cap index changes from baseline to follow-up indicated that IVUS results differed from OCT by 80%. Conclusions These preliminary results demonstrated that there were significant differences between IVUS and OCT plaque cap thickness measurements. Large-scale patient studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Rui Lv
- School of Biological Science and Medical Engineering, Southeast University, #2 SiPailou, Nanjing, China
| | - Akiko Maehara
- The Cardiovascular Research Foundation, Columbia University, New York, USA
| | - Mitsuaki Matsumura
- The Cardiovascular Research Foundation, Columbia University, New York, USA
| | - Liang Wang
- School of Biological Science and Medical Engineering, Southeast University, #2 SiPailou, Nanjing, China
| | - Qingyu Wang
- School of Biological Science and Medical Engineering, Southeast University, #2 SiPailou, Nanjing, China
| | - Caining Zhang
- School of Biological Science and Medical Engineering, Southeast University, #2 SiPailou, Nanjing, China
| | - Xiaoya Guo
- School of Science, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Habib Samady
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Don P Giddens
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.,The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jie Zheng
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Gary S Mintz
- The Cardiovascular Research Foundation, Columbia University, New York, USA
| | - Dalin Tang
- School of Biological Science and Medical Engineering, Southeast University, #2 SiPailou, Nanjing, China. .,Mathematical Sciences Department, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA.
| |
Collapse
|
79
|
Feng Z, Zhu Y, Zhang J, Yang W, Chen Z, Li B. Hsa-circ_0010283 Regulates Oxidized Low-Density Lipoprotein-Induced Proliferation and Migration of Vascular Smooth Muscle Cells by Targeting the miR-133a-3p/Pregnancy-Associated Plasma Protein A Axis. Circ J 2020; 84:2259-2269. [PMID: 33162460 DOI: 10.1253/circj.cj-20-0345] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The dysfunction of vascular smooth muscle cells (VSMCs) contributes to the development of atherosclerosis. This study aimed to investigate the role of circular RNA-0010283 (circ_0010283) in oxidized low-density lipoprotein (ox-LDL)-treated VSMCs and the associated action mechanism. METHODS AND RESULTS The expression of circ_0010283 was investigated using quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation was monitored by using a 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. Cell apoptosis was detected by using flow cytometry assay. A transwell assay was performed to observe migration and invasion, and a scratch assay was implemented to test migration. The expression of proliferation, apoptosis and migration/invasion-related proteins was measured by using a western blot. The targeted relationship was predicted by using a bioinformatics tool (Starbase) and verified by using a dual-luciferase reporter assay, a RNA immunoprecipitation (RIP) assay and a RNA pull-down assay. circ_0010283 was highly expressed in serum samples from atherosclerosis patients and ox-LDL-treated human VSMCs (HVSMCs). circ_0010283 knockdown suppressed ox-LDL-induced proliferation, migration and invasion in HVSMCs. MicroRNA-133a-3p (miR-133a-3p) was confirmed as a target of circ_0010283, and miR-133a-3p deficiency reversed the effects of circ_0010283 knockdown. Moreover, pregnancy-associated plasma protein A (PAPPA) was targeted by miR-133a-3p, and PAPPA overexpression reversed the effects of miR-133a-3p restoration. Interestingly, circ_0010283 could regulate PAPPA expression by mediating miR-133a-3p. CONCLUSIONS circ_0010283 participated in ox-LDL-induced dysfunctions of HVSMCs by modulating the miR-133a-3p/PAPPA pathway, suggesting that circ_0010283 might be associated with atherosclerosis pathogenesis.
Collapse
Affiliation(s)
- Zibo Feng
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Youpeng Zhu
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Jing Zhang
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Wenbo Yang
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Zhimin Chen
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Binghui Li
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| |
Collapse
|
80
|
Montarello NJ, Nelson AJ, Verjans J, Nicholls SJ, Psaltis PJ. The role of intracoronary imaging in translational research. Cardiovasc Diagn Ther 2020; 10:1480-1507. [PMID: 33224769 DOI: 10.21037/cdt-20-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Atherosclerotic cardiovascular disease is a key public health concern worldwide and leading cause of morbidity, mortality and health economic costs. Understanding atherosclerotic plaque microstructure in relation to molecular mechanisms that underpin its initiation and progression is needed to provide the best chance of combating this disease. Evolving vessel wall-based, endovascular coronary imaging modalities, including intravascular ultrasound (IVUS), optical coherence tomography (OCT) and near-infrared spectroscopy (NIRS), used in isolation or as hybrid modalities, have been advanced to allow comprehensive visualization of the pathological substrate of coronary atherosclerosis and accurately measure temporal changes in both the vessel wall and plaque characteristics. This has helped further our appreciation of the natural history of coronary artery disease (CAD) and the risk for major adverse cardiovascular events (MACE), evaluate the responsiveness to conventional and experimental therapeutic interventions, and assist in guiding percutaneous coronary intervention (PCI). Here we review the use of different imaging modalities for these purposes and the lessons they have provided thus far.
Collapse
Affiliation(s)
- Nicholas J Montarello
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide, Australia
| | - Adam J Nelson
- Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Duke Clinical Research Institute, Durham, NC, USA
| | - Johan Verjans
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Clayton, Australia
| | - Peter J Psaltis
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| |
Collapse
|
81
|
Zhang TY, Zhao Q, Liu ZS, Zhang CY, Yang J, Meng K. Relationship between monocyte/lymphocyte ratio and non-culprit plaque vulnerability in patients with acute coronary syndrome: An optical coherence tomography study. Medicine (Baltimore) 2020; 99:e21562. [PMID: 33031255 PMCID: PMC7544167 DOI: 10.1097/md.0000000000021562] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The importance of monocyte/lymphocyte ratio (MLR) has been indicated in the initiation and progression of coronary artery disease. However, few previous researches demonstrated the relationship between MLR and plaque vulnerability. We aimed to investigate coronary non-culprit plaque vulnerability in patients with acute coronary syndrome (ACS) by optical coherence tomography (OCT).A total of 72 ACS patients who underwent coronary angiography and OCT test in Beijing Anzhen Hospital were included in this retrospective study. The plaque vulnerability and plaque morphology were assessed by OCT.The non-culprit plaque in high MLR group exhibited more vulnerable features, characterizing as thinner thickness of fibrous cap (P = .013), greater maximum lipid core angle (P = .010) and longer lipid plaque length (P = .041). A prominently negative liner relation was found between MLR and thickness of fibrous cap (R = -0.225, P = .005). Meanwhile, the proportion of OCT-detected thin cap fibro-atheroma (TCFA) (P = .014) and plaque rupture (P = .017) were higher in high MLR group. Most importantly, multivariable logistic regression analysis showed MLR level was identified as an independent contributor to the presence of TCFA (OR:3.316, 95%: 1.448-7.593, P = .005). MLR could differentiate TCFA with a sensitivity of 60.0% and a specificity of 85.1%.Circulating MLR level has potential value in identifying the presence of vulnerable plaque in patients with ACS. MLR, as a non- invasive biomarker of inflammation, may be valuable in revealing plaque vulnerability.
Collapse
Affiliation(s)
- Ting-yu Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Qi Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Ze-sen Liu
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University
| | - Chao-yi Zhang
- Department of Cardiology, Beijing Zhongguanchun Hospital, Beijing, China
| | - Jie Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Kang Meng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| |
Collapse
|
82
|
Cheng Q, Zhang M, Zhang M, Ning L, Chen D. Long non-coding RNA LOC285194 regulates vascular smooth muscle cell apoptosis in atherosclerosis. Bioengineered 2020; 11:53-60. [PMID: 31884873 PMCID: PMC6961585 DOI: 10.1080/21655979.2019.1705054] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) recently have been implicated in many biological processes and diseases. Atherosclerosis is a major risk factor for cardiovascular disease. However, the functional role of lncRNAs in atherosclerosis is largely unknown. Here we identified LOC285194 as a key regulator of cell proliferation and apoptosis during atherosclerosis. The expression of LOC285194 was dramatically down-regulated in a aortic atherosclerotic plaques of well-defined model of apolipoprotein-E knockout (ApoE−/-) mice. Moreover, we found that targeting LOC285194 results in neointimal hyperplasia in vivo in carotid artery injury model. We also showed that targeting LOC285194 promotes cell proliferation and inhibits apoptosis in vascular smooth muscle cells (VSMCs) in vitro, and vice versa. In addition, targeting LOC285194 promotes cell invasion and migration in vitro. Our studies identify LOC285194 as a novel regulator of cell proliferation and apoptosis and suggest that this lncRNA could serve as a therapeutic target to treat atherosclerosis and related cardiovascular disorders.
Collapse
Affiliation(s)
- Qiushi Cheng
- Department of Health Care, Qingdao Municipal Hospital (East), Qingdao, Shandong, China
| | - Min Zhang
- Department of General surgery, The affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Maoshen Zhang
- Department of General surgery, The affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liang Ning
- Department of General surgery, The affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Dong Chen
- Department of General surgery, The affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
83
|
Abdelrahman KM, Chen MY, Dey AK, Virmani R, Finn AV, Khamis RY, Choi AD, Min JK, Williams MC, Buckler AJ, Taylor CA, Rogers C, Samady H, Antoniades C, Shaw LJ, Budoff MJ, Hoffmann U, Blankstein R, Narula J, Mehta NN. Coronary Computed Tomography Angiography From Clinical Uses to Emerging Technologies: JACC State-of-the-Art Review. J Am Coll Cardiol 2020; 76:1226-1243. [PMID: 32883417 PMCID: PMC7480405 DOI: 10.1016/j.jacc.2020.06.076] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/08/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Evaluation of coronary artery disease (CAD) using coronary computed tomography angiography (CCTA) has seen a paradigm shift in the last decade. Evidence increasingly supports the clinical utility of CCTA across various stages of CAD, from the detection of early subclinical disease to the assessment of acute chest pain. Additionally, CCTA can be used to noninvasively quantify plaque burden and identify high-risk plaque, aiding in diagnosis, prognosis, and treatment. This is especially important in the evaluation of CAD in immune-driven conditions with increased cardiovascular disease prevalence. Emerging applications of CCTA based on hemodynamic indices and plaque characterization may provide personalized risk assessment, affect disease detection, and further guide therapy. This review provides an update on the evidence, clinical applications, and emerging technologies surrounding CCTA as highlighted at the 2019 National Heart, Lung and Blood Institute CCTA Summit.
Collapse
Affiliation(s)
- Khaled M Abdelrahman
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Marcus Y Chen
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Amit K Dey
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Renu Virmani
- Department of Pathology, CVPath Institute, Gaithersburg, Maryland
| | - Aloke V Finn
- Department of Pathology, CVPath Institute, Gaithersburg, Maryland
| | - Ramzi Y Khamis
- Vascular Sciences Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Andrew D Choi
- Division of Cardiology and Department of Radiology, The George Washington University School of Medicine, Washington, DC
| | - James K Min
- Department of Radiology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, New York
| | - Michelle C Williams
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; Edinburgh Imaging, Queen's Medical Research Institute University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | - Habib Samady
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Leslee J Shaw
- Department of Radiology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, New York
| | - Matthew J Budoff
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California
| | - Udo Hoffmann
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ron Blankstein
- Departments of Medicine (Cardiovascular Division) and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jagat Narula
- Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josée and Henry R. Kravis Center for Cardiovascular Health Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, New York, New York
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
84
|
Pirri D, Fragiadaki M, Evans PC. Diabetic atherosclerosis: is there a role for the hypoxia-inducible factors? Biosci Rep 2020; 40:BSR20200026. [PMID: 32816039 PMCID: PMC7441368 DOI: 10.1042/bsr20200026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 07/28/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is a major cause of mortality worldwide and is driven by multiple risk factors, including diabetes. Diabetes is associated with either an insulin deficiency in its juvenile form or with insulin resistance and obesity in Type 2 diabetes mellitus, and the latter is clustered with other comorbidities to define the metabolic syndrome. Diabetes and metabolic syndrome are complex pathologies and are associated with cardiovascular risk via vascular inflammation and other mechanisms. Several transcription factors are activated upon diabetes-driven endothelial dysfunction and drive the progression of atherosclerosis. In particular, the hypoxia-inducible factor (HIF) transcription factor family is a master regulator of endothelial biology and is raising interest in the field of atherosclerosis. In this review, we will present an overview of studies contributing to the understanding of diabetes-driven atherosclerosis, integrating the role of HIF in this disease with the knowledge of its functions in metabolic syndrome and diabetic scenario.
Collapse
Affiliation(s)
- Daniela Pirri
- Department of Infection, Immunity and Cardiovascular disease, The University of Sheffield, U.K
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Maria Fragiadaki
- Department of Infection, Immunity and Cardiovascular disease, The University of Sheffield, U.K
| | - Paul C. Evans
- Department of Infection, Immunity and Cardiovascular disease, The University of Sheffield, U.K
| |
Collapse
|
85
|
Association of systemic inflammatory biomarkers with morphological characteristics of coronary atherosclerotic plaque by intravascular optical coherence tomography. Hellenic J Cardiol 2020; 62:101-106. [PMID: 32628997 DOI: 10.1016/j.hjc.2020.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Despite significant advances in preventive, medical, and interventional management, coronary artery disease remains the leading cause of death worldwide. We now know that in the majority of acute coronary syndromes, a thrombotic event is triggered either by the rupture or erosion of the so-called high-risk or 'vulnerable' plaque. However, accurately identifying the individual who is at significant risk of acute event remains the holy grail of preventive cardiology. To better stratify an individual's risk of developing and suffering a cardiovascular event, biomarkers are needed that can accurately predict coronary events and, if possible, monitor disease activity in response to medical or interventional therapies. In order to be able to understand the association of these biomarkers with the morphological substrate of high-risk plaques, intravascular imaging modalities can provide invaluable assistance. Novel imaging tools such as optical coherence tomography (OCT) have not only helped in identifying atherosclerotic plaque characteristics that are unstable but also in estimating global plaque burden. In this study, we provide an overview of our current knowledge of association of various inflammatory markers with atherosclerotic plaque characteristics seen on OCT.
Collapse
|
86
|
Fedak A, Ciuk K, Urbanik A. Ultrasonography of vulnerable atherosclerotic plaque in the carotid arteries: B-mode imaging. J Ultrason 2020; 20:e135-e145. [PMID: 32609972 PMCID: PMC7418858 DOI: 10.15557/jou.2020.0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/30/2020] [Indexed: 11/22/2022] Open
Abstract
The most common type of stroke, i.e. ischemic stroke, is a great challenge for contemporary medicine as it poses both diagnostic and therapeutic difficulties. Atherosclerosis, which is rapidly beginning to affect more and more social groups, is the main cause of cerebrovascular accidents. Atherosclerosis is currently defined as a generalized, dynamic and heterogeneous inflammatory and immune process affecting arterial walls. Atherosclerotic plaque is the emanation of this disease. As the paradigm of the diagnosis of atherosclerosis has changed, it has become crucial to properly identify plaque instability within the carotid arteries by evaluating parameters and phenomena that signify a developing cascade of complications, eventually leading to stroke. Irrespective of the ultrasound technique employed, proper morphological evaluation of atherosclerotic plaque, involving observation of its echogenicity, i.e. subjective analysis of its structure, with the classification to Gray-Weale–Nicolaides types as well as assessment of the integrity of its surface, makes it possible to roughly evaluate plaque morphology and thereby its stability. This enables treatment planning and therapy monitoring. This evaluation should be a prelude to further diagnostic work-up, which involves non-invasive examinations that enable unambiguous assessment of plaque stability. These examinations include contrast-enhanced ultrasound to assess progression or recession of inflammation, which presents as plaque neovascularization, or shear wave elastography to objectively define tissue stiffness, and thereby its mineralization.
Collapse
Affiliation(s)
- Andrzej Fedak
- Department of Radiology, Jagiellonian University Medical College , Krakow , Poland
| | - Katarzyna Ciuk
- Students' Scientific Group at the Department of Radiology, Jagiellonian University Medical College , Krakow , Poland
| | - Andrzej Urbanik
- Department of Radiology, Jagiellonian University Medical College , Krakow , Poland
| |
Collapse
|
87
|
From CT to artificial intelligence for complex assessment of plaque-associated risk. Int J Cardiovasc Imaging 2020; 36:2403-2427. [PMID: 32617720 DOI: 10.1007/s10554-020-01926-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023]
Abstract
The recent technological developments in the field of cardiac imaging have established coronary computed tomography angiography (CCTA) as a first-line diagnostic tool in patients with suspected coronary artery disease (CAD). CCTA offers robust information on the overall coronary circulation and luminal stenosis, also providing the ability to assess the composition, morphology, and vulnerability of atherosclerotic plaques. In addition, the perivascular adipose tissue (PVAT) has recently emerged as a marker of increased cardiovascular risk. The addition of PVAT quantification to standard CCTA imaging may provide the ability to extract information on local inflammation, for an individualized approach in coronary risk stratification. The development of image post-processing tools over the past several years allowed CCTA to provide a significant amount of data that can be incorporated into machine learning (ML) applications. ML algorithms that use radiomic features extracted from CCTA are still at an early stage. However, the recent development of artificial intelligence will probably bring major changes in the way we integrate clinical, biological, and imaging information, for a complex risk stratification and individualized therapeutic decision making in patients with CAD. This review aims to present the current evidence on the complex role of CCTA in the detection and quantification of vulnerable plaques and the associated coronary inflammation, also describing the most recent developments in the radiomics-based machine learning approach for complex assessment of plaque-associated risk.
Collapse
|
88
|
Kuku KO, Singh M, Ozaki Y, Dan K, Chezar-Azerrad C, Waksman R, Garcia-Garcia HM. Near-Infrared Spectroscopy Intravascular Ultrasound Imaging: State of the Art. Front Cardiovasc Med 2020; 7:107. [PMID: 32695796 PMCID: PMC7338425 DOI: 10.3389/fcvm.2020.00107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022] Open
Abstract
Acute coronary syndromes (ACS) secondary to coronary vessel plaques represent a major cause of cardiovascular morbidity and mortality worldwide. Advancements in imaging technology over the last 3 decades have continuously enabled the study of coronary plaques via invasive imaging methods like intravascular ultrasound (IVUS) and optical coherence tomography (OCT). The introduction of near-infrared spectroscopy (NIRS) as a modality that could detect the lipid (cholesterol) content of atherosclerotic plaques in the early nineties, opened the potential of studying “vulnerable” or rupture-prone, lipid-rich coronary plaques in ACS patients. Most recently, the ability of NIRS-IVUS to identify patients at risk of future adverse events was shown in a prospective multicenter trial, the Lipid-Rich-plaque Study. Intracoronary NIRS-IVUS imaging offers a unique method of coronary lipid-plaque characterization and could become a valuable clinical diagnostic and treatment monitoring tool.
Collapse
Affiliation(s)
- Kayode O Kuku
- MedStar Cardiovascular Research Network, MedStar Washington Hospital Center, MedStar Health Research Institute, Washington, DC, United States.,Section of Interventional Cardiology MedStar Washington Hospital Center, MedStar Heart and Vascular Institute, Washington, DC, United States.,Georgetown University Department of Medicine, Washington, DC, United States
| | - Manavotam Singh
- Section of Interventional Cardiology MedStar Washington Hospital Center, MedStar Heart and Vascular Institute, Washington, DC, United States.,Georgetown University Department of Medicine, Washington, DC, United States
| | - Yuichi Ozaki
- Section of Interventional Cardiology MedStar Washington Hospital Center, MedStar Heart and Vascular Institute, Washington, DC, United States.,Georgetown University Department of Medicine, Washington, DC, United States
| | - Kazuhiro Dan
- Section of Interventional Cardiology MedStar Washington Hospital Center, MedStar Heart and Vascular Institute, Washington, DC, United States.,Georgetown University Department of Medicine, Washington, DC, United States
| | - Chava Chezar-Azerrad
- Section of Interventional Cardiology MedStar Washington Hospital Center, MedStar Heart and Vascular Institute, Washington, DC, United States.,Georgetown University Department of Medicine, Washington, DC, United States
| | - Ron Waksman
- MedStar Cardiovascular Research Network, MedStar Washington Hospital Center, MedStar Health Research Institute, Washington, DC, United States.,Section of Interventional Cardiology MedStar Washington Hospital Center, MedStar Heart and Vascular Institute, Washington, DC, United States.,Georgetown University Department of Medicine, Washington, DC, United States
| | - Hector M Garcia-Garcia
- MedStar Cardiovascular Research Network, MedStar Washington Hospital Center, MedStar Health Research Institute, Washington, DC, United States.,Section of Interventional Cardiology MedStar Washington Hospital Center, MedStar Heart and Vascular Institute, Washington, DC, United States.,Georgetown University Department of Medicine, Washington, DC, United States
| |
Collapse
|
89
|
Di Gregoli K, Somerville M, Bianco R, Thomas AC, Frankow A, Newby AC, George SJ, Jackson CL, Johnson JL. Galectin-3 Identifies a Subset of Macrophages With a Potential Beneficial Role in Atherosclerosis. Arterioscler Thromb Vasc Biol 2020; 40:1491-1509. [PMID: 32295421 PMCID: PMC7253188 DOI: 10.1161/atvbaha.120.314252] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 04/06/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Galectin-3 (formerly known as Mac-2), encoded by the LGALS3 gene, is proposed to regulate macrophage adhesion, chemotaxis, and apoptosis. We investigated the role of galectin-3 in determining the inflammatory profile of macrophages and composition of atherosclerotic plaques. Approach and Results: We observed increased accumulation of galectin-3-negative macrophages within advanced human, rabbit, and mouse plaques compared with early lesions. Interestingly, statin treatment reduced galectin-3-negative macrophage accrual in advanced plaques within hypercholesterolemic (apolipoprotein E deficient) Apoe-/- mice. Accordingly, compared with Lgals3+/+:Apoe-/- mice, Lgals3-/-:Apoe-/- mice displayed altered plaque composition through increased macrophage:smooth muscle cell ratio, reduced collagen content, and increased necrotic core area, characteristics of advanced plaques in humans. Additionally, macrophages from Lgals3-/- mice exhibited increased invasive capacity in vitro and in vivo. Furthermore, loss of galectin-3 in vitro and in vivo was associated with increased expression of proinflammatory genes including MMP (matrix metalloproteinase)-12, CCL2 (chemokine [C-C motif] ligand 2), PTGS2 (prostaglandin-endoperoxide synthase 2), and IL (interleukin)-6, alongside reduced TGF (transforming growth factor)-β1 expression and consequent SMAD signaling. Moreover, we found that MMP12 cleaves macrophage cell-surface galectin-3 resulting in the appearance of a 22-kDa fragment, whereas plasma levels of galectin-3 were reduced in Mmp12-/-:Apoe-/- mice, highlighting a novel mechanism where MMP12-dependent cleavage of galectin-3 promotes proinflammatory macrophage polarization. Moreover, galectin-3-positive macrophages were more abundant within plaques of Mmp12-/-:Apoe-/- mice compared with Mmp12+/+:Apoe-/- animals. CONCLUSIONS This study reveals a prominent protective role for galectin-3 in regulating macrophage polarization and invasive capacity and, therefore, delaying plaque progression.
Collapse
Affiliation(s)
- Karina Di Gregoli
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Michelle Somerville
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Rosaria Bianco
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Anita C. Thomas
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Aleksandra Frankow
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Andrew C. Newby
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Sarah J. George
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Christopher L. Jackson
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| | - Jason L. Johnson
- From the Laboratory of Cardiovascular Pathology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, England
| |
Collapse
|
90
|
Ning K, Wang MJ, Lin G, Zhang YL, Li MY, Yang BF, Chen Y, Huang Y, Li ZM, Huang YJ, Zhu L, Liang K, Yu B, Zhu YZ, Zhu YC. eNOS-Nitric Oxide System Contributes to a Novel Antiatherogenic Effect of Leonurine via Inflammation Inhibition and Plaque Stabilization. J Pharmacol Exp Ther 2020; 373:463-475. [PMID: 32238453 DOI: 10.1124/jpet.119.264887] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/18/2020] [Indexed: 01/08/2023] Open
Abstract
Leonurine (LEO) is a bioactive small molecular compound that has protective effects on the cardiovascular system and prevents the early progression of atherosclerosis; however, it is not clear whether LEO is effective for plaque stability. A novel mouse atherosclerosis model involving tandem stenosis (TS) of the right carotid artery combined with western diet (WD) feeding was used. Apolipoprotein E gene-deficient mice were fed with a WD and received LEO administration daily for 13 weeks. TS was introduced 6 weeks after the onset of experiments. We found that LEO enhanced plaque stability by increasing fibrous cap thickness and collagen content while decreasing the population of CD68-positive cells. Enhanced plaque stability by LEO was associated with the nitric oxide synthase (NOS)-nitric oxide (NO) system. LEO restored the balance between endothelial NOS(E)- and inducible NOS(iNOS)-derived NO production; suppressed the NF-κB signaling pathway; reduced the level of the inflammatory infiltration in plaque, including cytokine interleukin 6; and downregulated the expression of adhesion molecules. These findings support the distinct role of LEO in plaque stabilization. In vitro studies with oxidized low-density lipoprotein-challenged human umbilical vein endothelial cells revealed that LEO balanced NO production and inhibited NF-κB/P65 nuclear translocation, thus mitigating inflammation. In conclusion, the restored balance of the NOS-NO system and mitigated inflammation contribute to the plaque-stabilizing effect of LEO. SIGNIFICANCE STATEMENT: LEO restored the balance between endothelial NOS and inducible NOS in NO production and inhibited excessive inflammation in atherosclerotic "unstable" and rupture-prone plaques in apolipoprotein E gene-deficient mice. The protective effect of LEO for stabilizing atherosclerotic plaques was due to improved collagen content, increased fibrous cap thickness, and decreased accumulation of macrophages/foam cells. So far, LEO has passed the safety and feasibility test of phase I clinical trial.
Collapse
Affiliation(s)
- Ke Ning
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Ming-Jie Wang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Ge Lin
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Yi-Lin Zhang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Meng-Yao Li
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Bao-Feng Yang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Ying Chen
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Yong Huang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Zhi-Ming Li
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Yi-Jun Huang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Lei Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Kun Liang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Bo Yu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Yi-Zhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Yi-Chun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| |
Collapse
|
91
|
Daghem M, Bing R, Fayad ZA, Dweck MR. Noninvasive Imaging to Assess Atherosclerotic Plaque Composition and Disease Activity: Coronary and Carotid Applications. JACC Cardiovasc Imaging 2020; 13:1055-1068. [PMID: 31422147 PMCID: PMC10661368 DOI: 10.1016/j.jcmg.2019.03.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/07/2019] [Accepted: 03/24/2019] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease is one of the leading causes of mortality and morbidity worldwide. Atherosclerosis imaging has traditionally focused on detection of obstructive luminal stenoses or measurements of plaque burden. However, with advances in imaging technology it has now become possible to noninvasively interrogate plaque composition and disease activity, thereby differentiating stable from unstable patterns of disease and potentially improving risk stratification. This manuscript reviews multimodality imaging in this field, focusing on carotid and coronary atherosclerosis and how these novel techniques have the potential to complement current imaging assessments and improve clinical decision making.
Collapse
Affiliation(s)
- Marwa Daghem
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Rong Bing
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
92
|
Do We See "Mona Lisa Smiles" in Different Angles by Pathology, Angiography, and OCT? JACC Cardiovasc Imaging 2020; 13:2000-2001. [PMID: 32912473 DOI: 10.1016/j.jcmg.2020.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 11/21/2022]
|
93
|
Clarke JRD, Duarte Lau F, Zarich SW. Determining the Significance of Coronary Plaque Lesions: Physiological Stenosis Severity and Plaque Characteristics. J Clin Med 2020; 9:jcm9030665. [PMID: 32131474 PMCID: PMC7141262 DOI: 10.3390/jcm9030665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 01/10/2023] Open
Abstract
The evaluation of coronary lesions has evolved in recent years. Physiologic-guided revascularization (particularly with pressure-derived fractional flow reserve (FFR)) has led to superior outcomes compared to traditional angiographic assessment. A greater importance, therefore, has been placed on the functional significance of an epicardial lesion. Despite the improvements in the limitations of angiography, insights into the relationship between hemodynamic significance and plaque morphology at the lesion level has shown that determining the implications of epicardial lesions is rather complex. Investigators have sought greater understanding by correlating ischemia quantified by FFR with plaque characteristics determined on invasive and non-invasive modalities. We review the background of the use of these diagnostic tools in coronary artery disease and discuss the implications of analyzing physiological stenosis severity and plaque characteristics concurrently.
Collapse
Affiliation(s)
- John-Ross D. Clarke
- Department of Internal Medicine, Yale-New Haven Health/Bridgeport Hospital, Bridgeport, CT 06610, USA;
- Correspondence: or ; Tel.: +1-203-260-4510
| | - Freddy Duarte Lau
- Department of Internal Medicine, Yale-New Haven Health/Bridgeport Hospital, Bridgeport, CT 06610, USA;
| | - Stuart W. Zarich
- The Heart and Vascular Institute, Yale-New Haven Health/Bridgeport Hospital, Bridgeport, CT 06610, USA;
| |
Collapse
|
94
|
Finck T, Stojanovic A, Will A, Hendrich E, Martinoff S, Hausleiter J, Hadamitzky M. Long-term prognostic value of morphological plaque features on coronary computed tomography angiography. Eur Heart J Cardiovasc Imaging 2020; 21:237-248. [PMID: 31578556 DOI: 10.1093/ehjci/jez238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/03/2019] [Accepted: 08/31/2019] [Indexed: 01/24/2023] Open
Abstract
AIMS To investigate the incremental prognostic value of morphological plaque features beyond clinical risk and coronary stenosis levels. Although associated with the degree of coronary stenosis, most cardiac events occur on the basis of ruptured non-obstructive plaques and consecutive vessel thrombosis. As such, identification of vulnerable plaques is paramount for cardiovascular risk prediction and treatment decisions. METHODS AND RESULTS A total of 1615 patients with suspected but not previously diagnosed coronary artery disease (CAD) were examined by coronary computed tomography angiography and morphological plaque features were assessed. Mean follow-up was 10.5 (interquartile range 9.2-11.4) years. Cox proportional hazards analysis was used for the composite endpoint of cardiac death and non-fatal myocardial infarction. The study endpoint was reached in 51 patients (36 cardiac deaths, 15 non-fatal myocardial infarctions). In addition to quantitative parameters (presence of any calcified/non-calcified plaque or elevated plaque load), morphologic plaque features such as a spotty or gross calcification pattern and napkin-ring sign (NRS) were predictive for events. However, only spotty calcified plaques and NRS could confer additive prognostic value beyond clinical risk and coronary stenosis level. In a stepwise approach, endpoint prediction beyond clinical risk (Morise score) could be improved by inclusion of CAD severity (χ2 of 27.5, P < 0.001) and further discrimination for spotty calcified plaques (χ2 of 3.89, P = 0.049). CONCLUSION Improved cardiovascular risk prediction beyond clinical risk and coronary stenosis levels can be made by discriminating for the presence of spotty calcified plaques. Thus, an intensified prophylactic anti-atherosclerotic treatment appears to be warranted in patients with coronary plaques that show spotty calcifications.
Collapse
Affiliation(s)
- Tom Finck
- Institut für Radiologie und Nuklearmedizin, Deutsches Herzzentrum München, Klinik an der Technische Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Antonija Stojanovic
- Institut für Radiologie und Nuklearmedizin, Deutsches Herzzentrum München, Klinik an der Technische Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Albrecht Will
- Institut für Radiologie und Nuklearmedizin, Deutsches Herzzentrum München, Klinik an der Technische Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Eva Hendrich
- Institut für Radiologie und Nuklearmedizin, Deutsches Herzzentrum München, Klinik an der Technische Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Stefan Martinoff
- Institut für Radiologie und Nuklearmedizin, Deutsches Herzzentrum München, Klinik an der Technische Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Jörg Hausleiter
- Medizinische Klinik und Poliklinik I, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Heart Alliance at DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung E.V.), Munich, Germany
| | - Martin Hadamitzky
- Institut für Radiologie und Nuklearmedizin, Deutsches Herzzentrum München, Klinik an der Technische Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| |
Collapse
|
95
|
Bajraktari A, Bytyçi I, Henein MY. The Relationship between Coronary Artery Wall Shear Strain and Plaque Morphology: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2020; 10:diagnostics10020091. [PMID: 32046306 PMCID: PMC7168174 DOI: 10.3390/diagnostics10020091] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 11/26/2022] Open
Abstract
Background and Aim: Arterial wall shear strain (WSS) has been proposed to impact the features of atherosclerotic plaques. The aim of this meta-analysis was to assess the impact of different types of WSS on plaque features in coronary artery disease (CAD). Methods: We systematically searched PubMed-Medline, EMBASE, Scopus, Google Scholar, and the Cochrane Central Registry, from 1989 up to January 2020 and selected clinical trials and observational studies which assessed the relationship between WSS, measured by intravascular ultrasound (IVUS), and plaque morphology in patients with CAD. Results: In four studies, a total of 72 patients with 13,098 coronary artery segments were recruited, with mean age 57.5 ± 9.5 years. The pooled analysis showed that low WSS was associated with larger baseline lumen area (WMD 2.55 [1.34 to 3.76, p < 0.001]), smaller plaque area (WMD −1.16 [−1.84 to −0.49, p = 0.0007]), lower plaque burden (WMD −12.7 [−21.4 to −4.01, p = 0.04]), and lower necrotic core area (WMD −0.32 [−0.78 to 0.14, p = 0.04]). Low WSS also had smaller fibrous area (WMD −0.79 [−1.88 to 0.30, p = 0.02]) and smaller fibro-fatty area (WMD −0.22 [−0.57 to 0.13, p = 0.02]), compared with high WSS, but the dense calcium score was similar between the two groups (WMD −0.17 [−0.47 to 0.13, p = 0.26]). No differences were found between intermediate and high WSS. Conclusions: High WSS is associated with signs of plaque instability such as higher necrotic core, higher calcium score, and higher plaque burden compared with low WSS. These findings highlight the role of IVUS in assessing plaque vulnerability.
Collapse
Affiliation(s)
- Artan Bajraktari
- Institute of Public Health and Clinical Medicine, Umeå University, 90187 Umeå, Sweden; (A.B.); (I.B.)
| | - Ibadete Bytyçi
- Institute of Public Health and Clinical Medicine, Umeå University, 90187 Umeå, Sweden; (A.B.); (I.B.)
- Clinic of Cardiology, University Clinical Centre of Kosovo, Prishtina 10000, Kosovo
| | - Michael Y. Henein
- Institute of Public Health and Clinical Medicine, Umeå University, 90187 Umeå, Sweden; (A.B.); (I.B.)
- Institute of Environment & Health and Societies, Brunel University, Middlesex UB8 3PH, UK
- Molecular and Clinic Research Institute, St George University, London SW17 0RE, UK
- Correspondence: ; Tel.: +46-90-785-1431
| |
Collapse
|
96
|
Daghem M, Newby DE. Detecting unstable plaques in humans using cardiac CT: Can it guide treatments? Br J Pharmacol 2020; 178:2204-2217. [PMID: 31596945 DOI: 10.1111/bph.14896] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/15/2019] [Accepted: 09/27/2019] [Indexed: 12/15/2022] Open
Abstract
Advances in imaging technology have driven the rapid expansion in the use of CT in the assessment of coronary atherosclerotic plaque. Based on a rapidly growing evidence base, current guidelines recommend coronary CT angiography as the first-line diagnostic test for patients presenting with stable chest pain. There is a growing need to refine current methods for diagnosis and risk stratification to improve the individualisation of preventative therapies. Imaging assessments of high-risk plaque with CT can be used to differentiate stable from unstable patterns of coronary atherosclerosis and potentially to improve patient risk stratification. This review will focus on coronary imaging with CT with a specific focus on the detection of coronary atherosclerosis, high-risk plaque features, and the implications for patient management.
Collapse
Affiliation(s)
- Marwa Daghem
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
97
|
Henein MY, Vancheri S, Bajraktari G, Vancheri F. Coronary Atherosclerosis Imaging. Diagnostics (Basel) 2020; 10:65. [PMID: 31991633 PMCID: PMC7168918 DOI: 10.3390/diagnostics10020065] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 02/05/2023] Open
Abstract
Identifying patients at increased risk of coronary artery disease, before the atherosclerotic complications become clinically evident, is the aim of cardiovascular prevention. Imaging techniques provide direct assessment of coronary atherosclerotic burden and pathological characteristics of atherosclerotic lesions which may predict the progression of disease. Atherosclerosis imaging has been traditionally based on the evaluation of coronary luminal narrowing and stenosis. However, the degree of arterial obstruction is a poor predictor of subsequent acute events. More recent techniques focus on the high-resolution visualization of the arterial wall and the coronary plaques. Most acute coronary events are triggered by plaque rupture or erosion. Hence, atherosclerotic plaque imaging has generally focused on the detection of vulnerable plaque prone to rupture. However, atherosclerosis is a dynamic process and the plaque morphology and composition may change over time. Most vulnerable plaques undergo progressive transformation from high-risk to more stable and heavily calcified lesions, while others undergo subclinical rupture and healing. Although extensive plaque calcification is often associated with stable atherosclerosis, the extent of coronary artery calcification strongly correlates with the degree of atherosclerosis and with the rate of future cardiac events. Inflammation has a central role in atherogenesis, from plaque formation to rupture, hence in the development of acute coronary events. Morphologic plaque assessment, both invasive and non-invasive, gives limited information as to the current activity of the atherosclerotic disease. The addition of nuclear imaging, based on radioactive tracers targeted to the inflammatory components of the plaques, provides a highly sensitive assessment of coronary disease activity, thus distinguishing those patients who have stable disease from those with active plaque inflammation.
Collapse
Affiliation(s)
- Michael Y. Henein
- Institute of Public Health and Clinical Medicine, Umea University, SE-90187 Umea, Sweden; (M.Y.H.); (G.B.)
- Departments of Fluid Mechanics, Brunel University, Middlesex, London UB8 3PH, UK
- Molecular and Nuclear Research Institute, St George’s University, London SW17 0RE, UK
| | - Sergio Vancheri
- Radiology Department, I.R.C.C.S. Policlinico San Matteo, 27100 Pavia, Italy;
| | - Gani Bajraktari
- Institute of Public Health and Clinical Medicine, Umea University, SE-90187 Umea, Sweden; (M.Y.H.); (G.B.)
- Medical Faculty, University of Prishtina, 10000 Prishtina, Kosovo
- Clinic of Cardiology, University Clinical Centre of Kosova, 10000 Prishtina, Kosovo
| | | |
Collapse
|
98
|
Berchiolli R, Erba PA, Slart RHJA. Hunting the Carotid Culprit: An Intriguing Game. Stroke 2020; 51:701-702. [PMID: 31948358 DOI: 10.1161/strokeaha.119.027945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Raffaella Berchiolli
- From the Vascular Surgery Unit, Cardiothoracic and Vascular Department (R.B.), University of Pisa, Italy
| | - Paola A Erba
- Department of Nuclear Medicine (P.A.E.), University of Pisa, Italy.,Department of Translational Research and New Technology in Medicine (P.A.E.), University of Pisa, Italy.,Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, the Netherlands (P.A.E., R.H.J.A.S.)
| | - Riemer H J A Slart
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, the Netherlands (P.A.E., R.H.J.A.S.).,Department of Biomedical Photonic Imaging, TechMed Centre, University of Twente, Enschede, the Netherlands (R.H.J.A.S.)
| |
Collapse
|
99
|
Horman S, Dechamps M, Octave M, Lepropre S, Bertrand L, Beauloye C. Platelet Function and Coronary Microvascular Dysfunction. Microcirculation 2020. [DOI: 10.1007/978-3-030-28199-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
100
|
Waksman R, Di Mario C, Torguson R, Ali ZA, Singh V, Skinner WH, Artis AK, Cate TT, Powers E, Kim C, Regar E, Wong SC, Lewis S, Wykrzykowska J, Dube S, Kazziha S, van der Ent M, Shah P, Craig PE, Zou Q, Kolm P, Brewer HB, Garcia-Garcia HM. Identification of patients and plaques vulnerable to future coronary events with near-infrared spectroscopy intravascular ultrasound imaging: a prospective, cohort study. Lancet 2019; 394:1629-1637. [PMID: 31570255 DOI: 10.1016/s0140-6736(19)31794-5] [Citation(s) in RCA: 286] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Near-infrared spectroscopy (NIRS) intravascular ultrasound imaging can detect lipid-rich plaques (LRPs). LRPs are associated with acute coronary syndromes or myocardial infarction, which can result in revascularisation or cardiac death. In this study, we aimed to establish the relationship between LRPs detected by NIRS-intravascular ultrasound imaging at unstented sites and subsequent coronary events from new culprit lesions. METHODS In this prospective, cohort study (LRP), patients from 44 medical centres were enrolled in Italy, Latvia, Netherlands, Slovakia, UK, and the USA. Patients with suspected coronary artery disease who underwent cardiac catheterisation with possible ad hoc percutaneous coronary intervention were eligible to be enrolled. Enrolled patients underwent scanning of non-culprit segments using NIRS-intravascular ultrasound imaging. The study had two hierarchal primary hypotheses, patient and plaque, each testing the association between maximum 4 mm Lipid Core Burden Index (maxLCBI4mm) and non-culprit major adverse cardiovascular events (NC-MACE). Enrolled patients with large LRPs (≥250 maxLCBI4mm) and a randomly selected half of patients with small LRPs (<250 maxLCBI4mm) were followed up for 24 months. This study is registered with ClinicalTrials.gov, NCT02033694. FINDINGS Between Feb 21, 2014, and March 30, 2016, 1563 patients were enrolled. NIRS-intravascular ultrasound device-related events were seen in six (0·4%) patients. 1271 patients (mean age 64 years, SD 10, 883 [69%] men, 388 [31%]women) with analysable maxLCBI4mm were allocated to follow-up. The 2-year cumulative incidence of NC-MACE was 9% (n=103). Both hierarchical primary hypotheses were met. On a patient level, the unadjusted hazard ratio (HR) for NC-MACE was 1·21 (95% CI 1·09-1·35; p=0·0004) for each 100-unit increase maxLCBI4mm) and adjusted HR 1·18 (1·05-1·32; p=0·0043). In patients with a maxLCBI4mm more than 400, the unadjusted HR for NC-MACE was 2·18 (1·48-3·22; p<0·0001) and adjusted HR was 1·89 (1·26-2·83; p=0·0021). At the plaque level, the unadjusted HR was 1·45 (1·30-1·60; p<0·0001) for each 100-unit increase in maxLCBI4mm. For segments with a maxLCBI4mm more than 400, the unadjusted HR for NC-MACE was 4·22 (2·39-7·45; p<0·0001) and adjusted HR was 3·39 (1·85-6·20; p<0·0001). INTERPRETATION NIRS imaging of non-obstructive territories in patients undergoing cardiac catheterisation and possible percutaneous coronary intervention was safe and can aid in identifying patients and segments at higher risk for subsequent NC-MACE. NIRS-intravascular ultrasound imaging adds to the armamentarium as the first diagnostic tool able to detect vulnerable patients and plaques in clinical practice. FUNDING Infraredx.
Collapse
Affiliation(s)
- Ron Waksman
- MedStar Washington Hospital Center, Washington, DC, USA.
| | | | | | - Ziad A Ali
- New York Presbyterian/Columbia University Medical Center, New York, NY & Cardiovascular Research Foundation, New York, NY, USA
| | | | | | | | | | - Eric Powers
- Medical University of South Carolina Hospital, Charleston, SC, USA
| | | | | | - S Chiu Wong
- NewYork-Presbyterian/Weill Cornell Medical Center, New York, NY, USA
| | | | | | - Sandeep Dube
- Community Heart and Vascular, Indianapolis, IN, USA
| | | | | | | | - Paige E Craig
- MedStar Washington Hospital Center, Washington, DC, USA
| | - Quan Zou
- MedStar Washington Hospital Center, Washington, DC, USA
| | - Paul Kolm
- MedStar Washington Hospital Center, Washington, DC, USA
| | | | | |
Collapse
|