1
|
Ueki Y, Itagaki T, Kuwahara K. Lipid-lowering Therapy and Coronary Plaque Regression. J Atheroscler Thromb 2024; 31:1479-1495. [PMID: 39111840 PMCID: PMC11537793 DOI: 10.5551/jat.rv22024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 11/06/2024] Open
Abstract
Lipid-lowering therapy plays a central role in reducing cardiovascular events. Over the past few decades, clinical trials utilizing several imaging techniques have consistently shown that lipid-lowering therapy can reduce the coronary plaque burden and improve plaque composition. Although intravascular ultrasound has been the most extensively used modality to assess plaque burden, other invasive modalities, such as optical coherence tomography and near-infrared spectroscopy, provide relevant data on plaque vulnerability, and computed tomography angiography detects both plaque volume and characteristics non-invasively. A large body of evidence supports the notion that reducing low-density lipoprotein cholesterol using statins combined with ezetimibe and proprotein convertase subtillisin/kexin type 9 inhibitors consistently shows improvements in plaque burden and favorable morphological changes. This review summarizes previously obtained data on the impact of lipid-lowering treatment strategies on atherosclerotic plaque regression, as assessed using several imaging modalities.
Collapse
Affiliation(s)
- Yasushi Ueki
- Department of Cardiovascular Medicine, Shinshu University Hospital, Nagano, Japan
| | - Tadashi Itagaki
- Department of Cardiovascular Medicine, Shinshu University Hospital, Nagano, Japan
| | - Koichiro Kuwahara
- Department of Cardiovascular Medicine, Shinshu University Hospital, Nagano, Japan
| |
Collapse
|
2
|
Yang Y, Pan J, Wang A, Ma Y, Liu Y, Jiang W. A novel method for the diagnosis of atherosclerosis based on nanotechnology. J Mater Chem B 2024; 12:9144-9154. [PMID: 39177217 DOI: 10.1039/d4tb00900b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Cardiovascular disease (CVD) is a global health concern, presenting significant risks to human health. Atherosclerosis is among the most prevalent CVD, impacting the medium and large arteries in the kidneys, brain, heart, and other vital organs, as well as the lower limbs. As the disease progresses, arterial obstruction can result in heart attacks and strokes. Therefore, patients with atherosclerosis should receive accurate diagnosis and timely therapeutic intervention. With the advancements in nanomedicine, researchers have proposed new research strategies and methods for atherosclerosis imaging. This paper summarizes some current research findings on the use of nanomaterials in diagnosing atherosclerosis and offers insights for optimizing the imaging applications of nanomaterials in the future.
Collapse
Affiliation(s)
- Ying Yang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Jiangpeng Pan
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Aifeng Wang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Yongcheng Ma
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Ying Liu
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Wei Jiang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| |
Collapse
|
3
|
Barkas F, Sener YZ, Golforoush PA, Kheirkhah A, Rodriguez-Sanchez E, Novak J, Apellaniz-Ruiz M, Akyea RK, Bianconi V, Ceasovschih A, Chee YJ, Cherska M, Chora JR, D'Oria M, Demikhova N, Kocyigit Burunkaya D, Rimbert A, Macchi C, Rathod K, Roth L, Sukhorukov V, Stoica S, Scicali R, Storozhenko T, Uzokov J, Lupo MG, van der Vorst EPC, Porsch F. Advancements in risk stratification and management strategies in primary cardiovascular prevention. Atherosclerosis 2024; 395:117579. [PMID: 38824844 DOI: 10.1016/j.atherosclerosis.2024.117579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/29/2024] [Accepted: 05/14/2024] [Indexed: 06/04/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of morbidity and mortality worldwide, highlighting the urgent need for advancements in risk assessment and management strategies. Although significant progress has been made recently, identifying and managing apparently healthy individuals at a higher risk of developing atherosclerosis and those with subclinical atherosclerosis still poses significant challenges. Traditional risk assessment tools have limitations in accurately predicting future events and fail to encompass the complexity of the atherosclerosis trajectory. In this review, we describe novel approaches in biomarkers, genetics, advanced imaging techniques, and artificial intelligence that have emerged to address this gap. Moreover, polygenic risk scores and imaging modalities such as coronary artery calcium scoring, and coronary computed tomography angiography offer promising avenues for enhancing primary cardiovascular risk stratification and personalised intervention strategies. On the other hand, interventions aiming against atherosclerosis development or promoting plaque regression have gained attention in primary ASCVD prevention. Therefore, the potential role of drugs like statins, ezetimibe, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, omega-3 fatty acids, antihypertensive agents, as well as glucose-lowering and anti-inflammatory drugs are also discussed. Since findings regarding the efficacy of these interventions vary, further research is still required to elucidate their mechanisms of action, optimize treatment regimens, and determine their long-term effects on ASCVD outcomes. In conclusion, advancements in strategies addressing atherosclerosis prevention and plaque regression present promising avenues for enhancing primary ASCVD prevention through personalised approaches tailored to individual risk profiles. Nevertheless, ongoing research efforts are imperative to refine these strategies further and maximise their effectiveness in safeguarding cardiovascular health.
Collapse
Affiliation(s)
- Fotios Barkas
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece.
| | - Yusuf Ziya Sener
- Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | | | - Azin Kheirkhah
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elena Rodriguez-Sanchez
- Division of Cardiology, Department of Medicine, Department of Physiology, and Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Jan Novak
- 2(nd) Department of Internal Medicine, St. Anne's University Hospital in Brno and Faculty of Medicine of Masaryk University, Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Maria Apellaniz-Ruiz
- Genomics Medicine Unit, Navarra Institute for Health Research - IdiSNA, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Ralph Kwame Akyea
- Centre for Academic Primary Care, School of Medicine, University of Nottingham, United Kingdom
| | - Vanessa Bianconi
- Department of Medicine and Surgery, University of Perugia, Italy
| | - Alexandr Ceasovschih
- Internal Medicine Department, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Ying Jie Chee
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore
| | - Mariia Cherska
- Cardiology Department, Institute of Endocrinology and Metabolism, Kyiv, Ukraine
| | - Joana Rita Chora
- Unidade I&D, Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal; Universidade de Lisboa, Faculdade de Ciências, BioISI - Biosystems & Integrative Sciences Institute, Lisboa, Portugal
| | - Mario D'Oria
- Division of Vascular and Endovascular Surgery, Department of Medical Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Nadiia Demikhova
- Sumy State University, Sumy, Ukraine; Tallinn University of Technology, Tallinn, Estonia
| | | | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l'institut du Thorax, Nantes, France
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy
| | - Krishnaraj Rathod
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom; Barts Interventional Group, Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Vasily Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Svetlana Stoica
- "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania; Institute of Cardiovascular Diseases Timisoara, Timisoara, Romania
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Tatyana Storozhenko
- Cardiovascular Center Aalst, OLV Clinic, Aalst, Belgium; Department of Prevention and Treatment of Emergency Conditions, L.T. Malaya Therapy National Institute NAMSU, Kharkiv, Ukraine
| | - Jamol Uzokov
- Republican Specialized Scientific Practical Medical Center of Therapy and Medical Rehabilitation, Tashkent, Uzbekistan
| | | | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074, Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074, Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336, Munich, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074, Aachen, Germany
| | - Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Jia H, Chen X, Shen J, Liu R, Hou P, Yue S. Label-Free Fiber-Optic Raman Spectroscopy for Intravascular Coronary Atherosclerosis and Plaque Detection. ACS OMEGA 2024; 9:27789-27797. [PMID: 38973848 PMCID: PMC11223210 DOI: 10.1021/acsomega.4c01611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/15/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
The rupture of atherosclerotic plaques remains one of the leading causes of morbidity and mortality worldwide. The plaques have certain pathological characteristics including a fibrous cap, inflammation, and extensive lipid deposition in a lipid core. Various invasive and noninvasive imaging techniques can interrogate structural aspects of atheroma; however, the composition of the lipid core in coronary atherosclerosis and plaques cannot be accurately detected. Fiber-optic Raman spectroscopy has the capability of in vivo rapid and accurate biomarker detection as an emerging omics technology. Previous studies demonstrated that an intravascular Raman spectroscopic technique may assess and manage the therapeutic and medication strategies intraoperatively. The Raman spectral information identified plaque depositions consisting of lipids, triglycerides, and cholesterol esters as the major components by comparing normal region and early plaque formation region with histology. By focusing on the composition of plaques, we could identify the subgroups of plaques accurately and rapidly by Raman spectroscopy. Collectively, this fiber-optic Raman spectroscopy opens up new opportunities for coronary atherosclerosis and plaque detection, which would assist optimal surgical strategy and instant postoperative decision-making. In this paper, we will review the advancement of label-free fiber-optic Raman probe spectroscopy and its applications of coronary atherosclerosis and atherosclerotic plaque detection.
Collapse
Affiliation(s)
- Hao Jia
- Key
Laboratory of Biomechanics and Mechanobiology (Beihang University),
Ministry of Education, Institute of Medical Photonics, Beijing Advanced
Innovation Center for Biomedical Engineering, School of Biological
Science and Medical Engineering, Beihang
University, Beijing 100191, China
| | - Xun Chen
- Key
Laboratory of Biomechanics and Mechanobiology (Beihang University),
Ministry of Education, Institute of Medical Photonics, Beijing Advanced
Innovation Center for Biomedical Engineering, School of Biological
Science and Medical Engineering, Beihang
University, Beijing 100191, China
| | - Jianghao Shen
- Key
Laboratory of Biomechanics and Mechanobiology (Beihang University),
Ministry of Education, Institute of Medical Photonics, Beijing Advanced
Innovation Center for Biomedical Engineering, School of Biological
Science and Medical Engineering, Beihang
University, Beijing 100191, China
| | - Rujia Liu
- Key
Laboratory of Biomechanics and Mechanobiology (Beihang University),
Ministry of Education, Institute of Medical Photonics, Beijing Advanced
Innovation Center for Biomedical Engineering, School of Biological
Science and Medical Engineering, Beihang
University, Beijing 100191, China
| | - Peipei Hou
- Department
of Cardiology, The People’s Hospital
of China Medical University, Shenyang 110016, China
| | - Shuhua Yue
- Key
Laboratory of Biomechanics and Mechanobiology (Beihang University),
Ministry of Education, Institute of Medical Photonics, Beijing Advanced
Innovation Center for Biomedical Engineering, School of Biological
Science and Medical Engineering, Beihang
University, Beijing 100191, China
| |
Collapse
|
5
|
Sarraju A, Nissen SE. Atherosclerotic plaque stabilization and regression: a review of clinical evidence. Nat Rev Cardiol 2024; 21:487-497. [PMID: 38177454 DOI: 10.1038/s41569-023-00979-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 01/06/2024]
Abstract
Atherosclerotic plaque results from a complex interplay between lipid deposition, inflammatory changes, cell migration and arterial wall injury. Over the past two decades, clinical trials utilizing invasive arterial imaging modalities, such as intravascular ultrasonography, have shown that reducing levels of atherogenic lipoproteins, mainly serum LDL-cholesterol (LDL-C), to very low levels can safely reduce overall atherosclerotic plaque burden and favourably modify plaque composition. Classically, this outcome has been achieved with intensive statin therapy. Since 2016, newer and potent lipid-lowering strategies, such as proprotein convertase subtilisin-kexin type 9 inhibition, have shown incremental effects on plaque regression and risk of clinical events. Despite maximal reduction in plasma LDL-C levels, considerable residual cardiovascular risk remains in some patients. Therefore, there is a need to study therapeutic approaches that address residual risk beyond LDL-C reduction to promote plaque stabilization or regression. Contemporary imaging modalities, such as coronary computed tomography angiography, enable non-invasive assessment of the overall atherosclerotic plaque burden as well as of certain local plaque characteristics. This technology could allow further study of plaque stabilization and regression using novel therapeutic approaches. Non-invasive plaque assessment might also offer the potential to guide personalized management strategies if validated for this purpose.
Collapse
Affiliation(s)
- Ashish Sarraju
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Steven E Nissen
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
6
|
Aldana-Bitar J, Golub IS, Moore J, Krishnan S, Verghese D, Manubolu VS, Benzing T, Ichikawa K, Hamal S, Kianoush S, Anderson LR, Ramirez NR, Leipsic JA, Karlsberg RP, Budoff MJ. Colchicine and plaque: A focus on atherosclerosis imaging. Prog Cardiovasc Dis 2024; 84:68-75. [PMID: 38423236 DOI: 10.1016/j.pcad.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Colchicine is an anti-inflammatory medication, classically used to treat a wide spectrum of autoimmune diseases. More recently, colchicine has proven itself a key pharmacotherapy in cardiovascular disease (CVD) management, atherosclerotic plaque modification, and coronary artery disease (CAD) treatment. Colchicine acts on many anti-inflammatory pathways, which translates to cardiovascular event reduction, plaque transformation, and plaque reduction. With the FDA's 2023 approval of colchicine for reducing cardiovascular events, a novel clinical pathway opens. This advancement paves the route for CVD management that synergistically merges lipid lowering approaches with inflammation inhibition modalities. This pioneering moment spurs the need for this manuscript's comprehensive review. Hence, this paper synthesizes and surveys colchicine's new role as an atherosclerotic plaque modifier, to provide a framework for physicians in the clinical setting. We aim to improve understanding (and thereby application) of colchicine alongside existing mechanisms for CVD event reduction. This paper examines colchicine's anti-inflammatory mechanism, and reviews large cohort studies that evidence colchicine's blossoming role within CAD management. This paper also outlines imaging modalities for atherosclerotic analysis, reviews colchicine's mechanistic effect upon plaque transformation itself, and synthesizes trials which assess colchicine's nuanced effect upon atherosclerotic transformation.
Collapse
Affiliation(s)
- Jairo Aldana-Bitar
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA; Cardiovascular Research Foundation of Southern California, Beverly Hills, CA, USA.
| | - Ilana S Golub
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| | - Jeff Moore
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| | - Srikanth Krishnan
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA; Department of Medicine, Division of Cardiology, University of California Los Angeles, Westwood, CA, USA
| | - Dhiran Verghese
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| | - Venkat S Manubolu
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| | - Travis Benzing
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| | - Keshi Ichikawa
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| | - Sajad Hamal
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| | - Sina Kianoush
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| | - Lauren R Anderson
- Cardiovascular Research Foundation of Southern California, Beverly Hills, CA, USA
| | - Noah R Ramirez
- Cardiovascular Research Foundation of Southern California, Beverly Hills, CA, USA
| | - Jonathon A Leipsic
- Department of Medicine and Radiology, University of British Columbia, Canada
| | - Ronald P Karlsberg
- Cardiovascular Research Foundation of Southern California, Beverly Hills, CA, USA
| | - Matthew J Budoff
- Division of Cardiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| |
Collapse
|
7
|
Lin H, Zhang M, Hu M, Zhang Y, Jiang W, Tang W, Ouyang Y, Jiang L, Mi Y, Chen Z, He P, Zhao G, Ouyang X. Emerging applications of single-cell profiling in precision medicine of atherosclerosis. J Transl Med 2024; 22:97. [PMID: 38263066 PMCID: PMC10804726 DOI: 10.1186/s12967-023-04629-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 10/14/2023] [Indexed: 01/25/2024] Open
Abstract
Atherosclerosis is a chronic, progressive, inflammatory disease that occurs in the arterial wall. Despite recent advancements in treatment aimed at improving efficacy and prolonging survival, atherosclerosis remains largely incurable. In this review, we discuss emerging single-cell sequencing techniques and their novel insights into atherosclerosis. We provide examples of single-cell profiling studies that reveal phenotypic characteristics of atherosclerosis plaques, blood, liver, and the intestinal tract. Additionally, we highlight the potential clinical applications of single-cell analysis and propose that combining this approach with other techniques can facilitate early diagnosis and treatment, leading to more accurate medical interventions.
Collapse
Affiliation(s)
- Huiling Lin
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China
- Department of Physiology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Ming Zhang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, 511518, Guangdong, China
| | - Mi Hu
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China
| | - Yangkai Zhang
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China
| | - WeiWei Jiang
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wanying Tang
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China
| | - Yuxin Ouyang
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China
| | - Liping Jiang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yali Mi
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, 511518, Guangdong, China
| | - Zhi Chen
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Pingping He
- Department of Nursing, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China.
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, 511518, Guangdong, China.
| | - Xinping Ouyang
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China.
- Department of Physiology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China.
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, 410081, Hunan, Changsha, China.
- The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, School of Medicine, Hunan Normal University, 410081, Hunan, Changsha, China.
| |
Collapse
|
8
|
Johnsrud DO, Port SC. In 2023 invasive coronary angiography is preferred to PET myocardial perfusion imaging for the initial work-up of a symptomatic patient with a high coronary artery calcium score. J Nucl Cardiol 2023; 30:2269-2272. [PMID: 37845556 DOI: 10.1007/s12350-023-03385-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/06/2023] [Indexed: 10/18/2023]
|
9
|
Bass RD, García-García HM, Ueki Y, Holmvang L, Pedrazzini G, Roffi M, Koskinas KC, Shibutani H, Losdat S, Ziemer PGP, Blanco PJ, Levine MB, Bourantas CV, Räber L. Effect of High-Intensity Statin Therapy on Atherosclerosis (IBIS-4): Manual Versus Automated Methods of IVUS Analysis. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2023; 54:33-38. [PMID: 37087308 DOI: 10.1016/j.carrev.2023.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 04/24/2023]
Abstract
AIMS Standard manual analysis of IVUS to study the impact of anti-atherosclerotic therapies on the coronary vessel wall is done by a core laboratory (CL), the ground truth (GT). Automatic segmentation of IVUS with a machine learning (ML) algorithm has the potential to replace manual readings with an unbiased and reproducible method. The aim is to determine if results from a CL can be replicated with ML methods. METHODS This is a post-hoc, comparative analysis of the IBIS-4 (Integrated Biomarkers and Imaging Study-4) study (NCT00962416). The GT baseline and 13-month follow-up measurements of lumen and vessel area and percent atheroma volume (PAV) after statin induction were repeated by the ML algorithm. RESULTS The primary endpoint was change in PAV. PAV as measured by GT was 43.95 % at baseline and 43.02 % at follow-up with a change of -0.90 % (p = 0.007) while the ML algorithm measured 43.69 % and 42.41 % for baseline and follow-up, respectively, with a change of -1.28 % (p < 0.001). Along the most diseased 10 mm segments, GT-PAV was 52.31 % at baseline and 49.42 % at follow-up, with a change of -2.94 % (p < 0.001). The same segments measured by the ML algorithm resulted in PAV of 51.55 % at baseline and 47.81 % at follow-up with a change of -3.74 % (p < 0.001). CONCLUSIONS PAV, the most used endpoint in clinical trials, analyzed by the CL is closely replicated by the ML algorithm. ML automatic segmentation of lumen, vessel and plaque effectively reproduces GT and may be used in future clinical trials as the standard.
Collapse
Affiliation(s)
- Ronald D Bass
- School of Medicine, Georgetown University, Washington, DC, USA.
| | | | - Yasushi Ueki
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Lene Holmvang
- Cardiac Catheterization Laboratory, Rigshospitalet, Copenhagen, Denmark.
| | | | - Marco Roffi
- Division of Cardiology, University Hospital, Geneva, Switzerland.
| | | | - Hiroki Shibutani
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | | | - Paulo G P Ziemer
- National Laboratory for Scientific Computing, Petrópolis, Brazil
| | - Pablo J Blanco
- National Laboratory for Scientific Computing, Petrópolis, Brazil
| | - Molly B Levine
- Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA.
| | - Christos V Bourantas
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK; Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, UK
| | - Lorenz Räber
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
10
|
Wang L, Zhao Y, Zheng B, Huo Y, Fan Y, Ma D, Gu Y, Wang P. Ultrawide-bandwidth high-resolution all-optical intravascular ultrasound using miniaturized photoacoustic transducer. SCIENCE ADVANCES 2023; 9:eadg8600. [PMID: 37294755 PMCID: PMC10256152 DOI: 10.1126/sciadv.adg8600] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/05/2023] [Indexed: 06/11/2023]
Abstract
Conventional intravascular ultrasound (IVUS) uses piezoelectric transducers to electrically generate and receive ultrasound. However, it remains a challenge to achieve large bandwidth for high resolution without compromising imaging depth. We report an all-optical IVUS (AO-IVUS) imaging system using picosecond laser pulse-pumped carbon composite for ultrasound excitation and π-phase-shifted fiber Bragg gratings for ultrasound detection. Using this all-optical technique, we achieved ultrawide-bandwidth (147%) and high-resolution (18.6 micrometers) IVUS imaging, which is unattainable by conventional technique. Imaging performance has been characterized in phantoms, presenting 18.6-micrometer axial resolution, 124-micrometer lateral resolution, and 7-millimeter imaging depth. Rotational pullback imaging scans are performed in rabbit iliac artery, porcine coronary artery, and rabbit arteries with drug-eluting metal stents, in parallel with commercial intravenous ultrasound scans as reference. Results demonstrated the advantages of high-resolution AO-IVUS in delineating details in vascular structures, showing great potential in clinical applications.
Collapse
Affiliation(s)
- Lei Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yongwen Zhao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Bo Zheng
- Institute of Cardiovascular Disease, Peking University First Hospital, Beijing 100034, China
| | - Yong Huo
- Institute of Cardiovascular Disease, Peking University First Hospital, Beijing 100034, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Dinglong Ma
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Ying Gu
- Department of Laser Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Pu Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
11
|
Yao Y, Zhang P. Novel ultrasound techniques in the identification of vulnerable plaques-an updated review of the literature. Front Cardiovasc Med 2023; 10:1069745. [PMID: 37293284 PMCID: PMC10244552 DOI: 10.3389/fcvm.2023.1069745] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
Atherosclerosis is an inflammatory disease partly mediated by lipoproteins. The rupture of vulnerable atherosclerotic plaques and thrombosis are major contributors to the development of acute cardiovascular events. Despite various advances in the treatment of atherosclerosis, there has been no satisfaction in the prevention and assessment of atherosclerotic vascular disease. The identification and classification of vulnerable plaques at an early stage as well as research of new treatments remain a challenge and the ultimate goal in the management of atherosclerosis and cardiovascular disease. The specific morphological features of vulnerable plaques, including intraplaque hemorrhage, large lipid necrotic cores, thin fibrous caps, inflammation, and neovascularisation, make it possible to identify and characterize plaques with a variety of invasive and non-invasive imaging techniques. Notably, the development of novel ultrasound techniques has introduced the traditional assessment of plaque echogenicity and luminal stenosis to a deeper assessment of plaque composition and the molecular field. This review will discuss the advantages and limitations of five currently available ultrasound imaging modalities for assessing plaque vulnerability, based on the biological characteristics of the vulnerable plaque, and their value in terms of clinical diagnosis, prognosis, and treatment efficacy assessment.
Collapse
|
12
|
Association of Statins With Nonculprit Coronary Lesions and Adverse Events (from the LRP Study). Am J Cardiol 2023; 190:82-89. [PMID: 36571935 DOI: 10.1016/j.amjcard.2022.11.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/24/2022] [Accepted: 11/26/2022] [Indexed: 12/26/2022]
Abstract
Intravascular ultrasound and near-infrared spectroscopy can identify vulnerable coronary atherosclerotic plaques. In this LRP (Lipid-Rich Plaque) substudy, we evaluated the association of statins with nonculprit lesion arterial wall lipidic content and subsequent nonculprit major adverse cardiac events. Patients from the LRP study with known statin use were included. We divided the patients into 2 cohorts-"statin therapy" and "statin-naïve"-upon presentation and then described the intravascular ultrasound and near-infrared spectroscopy analysis based on maximum 4-mm lipid core burden index (maxLCBI4mm). At 2-year follow-up, the patients' clinical events were assessed based on their statin regimen change upon discharge. Finally, patients were stratified by statin intensity based on discharge regimen. Among the 1,526 patients, 1,120 were on a statin versus 396 who were statin-naive upon presentation. Patients on a statin at baseline had a statistically higher rate of cardiovascular risk factors, patients who were statin-naive were more likely to present with an acute coronary syndrome, and the maxLCBI4mm did not differ between the 2 groups (315.67 ± 181.36 vs 325.55 ± 192.16; p = 0.359). These findings were consistent in a secondary analysis evaluating statin intensity. Patients who were switched from no statin to a statin had improved outcomes (nonculprit major adverse cardiac events) compared with patients who were on a statin at baseline without change. In conclusion, despite having a higher burden of nonlipid-related cardiac co-morbidities, patients on a statin at baseline had similar maxLCBI4mm with patients who were statin-naive, regardless of intensity. Initiating a statin at discharge provides the most benefit for events related to nonculprit lesions.
Collapse
|
13
|
Joshi A, Bhambhani A, Barure R, Gonuguntla S, Sarathi V, Attia AM, Shrestha AB, Jaiswal V. Neutrophil-lymphocyte ratio and platelet-lymphocyte ratio as markers of stable ischemic heart disease in diabetic patients: An observational study. Medicine (Baltimore) 2023; 102:e32735. [PMID: 36749238 PMCID: PMC9902008 DOI: 10.1097/md.0000000000032735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Ischemic heart disease (IHD) is a pressing public health concern with high prevalence, mortality, and morbidity. Although the value of neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) as markers of the acute coronary syndrome are well recognized, there is a paucity of data deciphering their role in screening for stable ischemic heart disease (SIHD) in the presence of type 2 diabetes mellitus (T2DM). The present study investigates the value of NLR and PLR as markers of SIHD in T2DM. We evaluated the predictive value of NLR and PLR for SIHD by comparing T2DM patients having angiographically proven SIHD to T2DM patients without IHD at different cutoff levels by evaluating the area under the curve (AUC) obtained from receiver-operating-characteristic analysis. Raised NLR and PLR were significantly associated with SIHD ( P < .001 for each). On performing AUC-receiver-operating-characteristic analysis, NLR of > 2.39 and PLR of > 68.80 were associated with the highest prevalence of SIHD (NLR, AUC: 0.652 [0.605-0.699]; CI: 95%; P < .001, PLR, AUC: 0.623 [0.575-0.671] CI: 95%; P < .001). The sensitivities and specificities for these cutoff values were 50% and 73% for NLR and 73% and 46% for PLR, respectively. NLR and PLR were significantly higher in SIHD compared to those without; however, these markers had limited predictive potential in the setting of T2DM.
Collapse
Affiliation(s)
- Amey Joshi
- Department of Cardiology, Vydehi Institute of Medical Sciences and Research Center, Whitefield, Bangalore, Karnataka, India
| | - Anupam Bhambhani
- Department of Cardiology, Vydehi Institute of Medical Sciences and Research Center, Whitefield, Bangalore, Karnataka, India
| | - Ramdas Barure
- Department of Endocrinology, Vydehi Institute of Medical Sciences and Research Center, Whitefield, Bangalore, Karnataka, India
| | - Samhitha Gonuguntla
- Department of Cardiology, Vydehi Institute of Medical Sciences and Research Center, Whitefield, Bangalore, Karnataka, India
| | - Vijaya Sarathi
- Department of Endocrinology, Vydehi Institute of Medical Sciences and Research Center, Whitefield, Bangalore, Karnataka, India
| | | | - Abhigan Babu Shrestha
- Department of Medicine, M Abdur Rahim Medical College, Dinajpur, Bangladesh
- * Correspondence: Abhigan Babu Shrestha, Department of Medicine, M Abdur Rahim Medical College, Dinajpur 5200, Bangladesh (e-mail: )
| | | |
Collapse
|
14
|
Jiang J, Yuan C, Zhang J, Xie Z, Xiao J. Spectroscopic photoacoustic/ultrasound/optical-microscopic multimodal intrarectal endoscopy for detection of centimeter-scale deep lesions. Front Bioeng Biotechnol 2023; 11:1136005. [PMID: 36777250 PMCID: PMC9909099 DOI: 10.3389/fbioe.2023.1136005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
The inadequacy of existing colorectal imaging tools has significantly obstructed the efficient detection of colorectal cancer. To address this issue, this work presents the cross-scale endoscopic imaging of rectal tumors with a combined photoacoustic/ultrasound tomography system and wide-field optical microscopy. This multimodal system combines the merits of centimeter-scale deep penetration, multi-spectral imaging, cross-scale imaging ability, low system cost, and 360° view in a single modality. Results indicated that the proposed system could reliably depict the location of the cancer invasion depth spectroscopically with indocyanine green The tumor angiogenesis can be well identified in the wide-field optical imaging mode, which helps to localize the tumors and guide the following photoacoustic/ultrasound scan. This work may facilitate the accurate characterization of colorectal cancer and promote the clinical translation of photoacoustic-based colorectal endoscopy.
Collapse
|
15
|
Case BC, Torguson R, Mintz GS, Di Mario C, Medranda GA, Zhang C, Shea C, Garcia-Garcia HM, Waksman R. Additive Effect of Multiple High-Risk Coronary Artery Segments on Patient Outcomes: LRP Study Sub-Analysis. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2023; 46:38-43. [PMID: 36058829 DOI: 10.1016/j.carrev.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND The Lipid Rich Plaque (LRP) Study established the association between high volume of lipidic content (maximum Lipid Core Burden Index [maxLCBI4mm] >400) in the coronary arteries and subsequent non-culprit major adverse cardiac events (NC-MACE). This analysis sought to assess the clinical impact of more than one lipid-rich plaque in the coronary tree. METHODS The LRP patient population was divided into four cohorts: 1) patients with all segments with maxLCBI4mm = 0; 2) patients with all coronary segments maxLCBI4mm < 400, but >0; 3) patients with 1 segment maxLCBI4mm > 400; and 4) patients with 2+ coronary segments with maxLCBI4mm > 400. Baseline characteristics, plaque-level characteristics, and follow-up outcomes were described. RESULTS Among 1550 patients, only 3.2 % had all segments with maxLCBI4mm = 0; 65.1 % had segments with maxLCBI4mm > 0 but <400; 22.5 % had one segment with maxLCBI4mm > 400; and 9.5 % had 2+ coronary segments with maxLCBI4mm > 400. Distribution within the coronary tree (one versus multiple arteries) did not differ. Overall, 1269 patients were allocated to follow-up (per study design). The composite of all-cause death, cardiac death, any revascularization, and NC-MACE was statistically higher in patients with 1 segment maxLCBI4mm > 400 and numerically even higher in patients with 2+ segments with maxLCBI4mm > 400. Patients with maxLCBI4mm = 0 had no events within two years. CONCLUSION There is a stepwise increased risk of all-cause death, cardiac death, any revascularization, and NC-MACE according to the number of coronary segments with maxLCBI4mm > 400. In contrast, maxLCBI4mm = 0 results in a low event rate. CLINICAL TRIAL REGISTRATION The Lipid-Rich Plaque Study (LRP), https://clinicaltrials.gov/ct2/show/NCT02033694, NCT02033694.
Collapse
Affiliation(s)
- Brian C Case
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Rebecca Torguson
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Gary S Mintz
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Carlo Di Mario
- Structural Interventional Cardiology, Department of Clinical & Experimental Medicine, Careggi University Hospital, Florence, Italy
| | - Giorgio A Medranda
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Cheng Zhang
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Corey Shea
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Hector M Garcia-Garcia
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Ron Waksman
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA.
| |
Collapse
|
16
|
Vesterbekkmo EK, Aamot Aksetøy IL, Follestad T, Nilsen HO, Hegbom K, Wisløff U, Wiseth R, Madssen E. High intensity interval training induces beneficial effects on coronary atheromatous plaques - a randomized trial. Eur J Prev Cardiol 2022; 30:384-392. [PMID: 36562212 DOI: 10.1093/eurjpc/zwac309] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Coronary atheroma volume is associated with risk of coronary events in coronary artery disease (CAD). Exercise training is a cornerstone in primary and secondary prevention of CAD, but the effect of exercise on coronary atheromatous plaques is largely unknown. PURPOSE We assessed the effect of six months supervised high intensity interval training (HIIT) on coronary plaque geometry using intravascular ultrasound in patients with stable CAD following percutaneous coronary intervention (PCI). METHODS Sixty patients were randomized to two sessions of weekly supervised HIIT at 85-95% of peak heart rate (n = 30) or to follow contemporary preventive guidelines (control group, n = 30). The study endpoints were change in percent atheroma volume (PAV) and total atheroma volume (TAV) normalized for segment length (TAVnorm) at six-month follow-up. RESULTS The change in average PAV for matched coronary segments from baseline to follow-up showed a significant between-group difference (-1.4, 95% CI: -2.7 to -0.1, p = 0.036). There was a significant reduction in the HIIT group (-1.2, 95% CI: -2.1 to -0.2, p = 0.017) while not in the control group (0.2, 95% CI: -0.7 to 1.1, p = 0.616). TAVnorm was reduced (-9 mm3, 95% CI: -14.7 to -3.4, p = 0.002) after HIIT, with a significant between-group difference (-12.0 mm3, 95% CI: -19.9 to -4.2, p = 0.003). CONCLUSION In patients with established CAD, a regression of atheroma volume was observed in those undergoing six months of supervised HIIT compared with patients following contemporary preventive guidelines. Our study indicates that high intensity interval training counteracts atherosclerotic coronary disease progression and reduces atheroma volume in residual coronary atheromatous plaques following PCI.
Collapse
Affiliation(s)
- Elisabeth Kleivhaug Vesterbekkmo
- Clinic of Cardiology, St. Olavs University Hospital, Trondheim, Norway.,Department of Circulation and Medical Imaging at Norwegian University of Science and Technology, Trondheim, Norway.,National Advisory Unit on Exercise Training as Medicine for Cardiopulmonary Conditions, Trondheim, Norway
| | - Inger-Lise Aamot Aksetøy
- Clinic of Cardiology, St. Olavs University Hospital, Trondheim, Norway.,Department of Circulation and Medical Imaging at Norwegian University of Science and Technology, Trondheim, Norway.,National Advisory Unit on Exercise Training as Medicine for Cardiopulmonary Conditions, Trondheim, Norway
| | - Turid Follestad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Queensland, Australia
| | - Hans Olav Nilsen
- Clinic of Cardiology, St. Olavs University Hospital, Trondheim, Norway.,Department of Circulation and Medical Imaging at Norwegian University of Science and Technology, Trondheim, Norway
| | - Knut Hegbom
- Clinic of Cardiology, St. Olavs University Hospital, Trondheim, Norway
| | - Ulrik Wisløff
- Department of Circulation and Medical Imaging at Norwegian University of Science and Technology, Trondheim, Norway.,School of Human Movement and Nutrition Science, University of Queensland, Queensland, Australia
| | - Rune Wiseth
- Clinic of Cardiology, St. Olavs University Hospital, Trondheim, Norway.,Department of Circulation and Medical Imaging at Norwegian University of Science and Technology, Trondheim, Norway
| | - Erik Madssen
- Clinic of Cardiology, St. Olavs University Hospital, Trondheim, Norway.,Department of Circulation and Medical Imaging at Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
17
|
Kim K, Youm JY, Lee EH, Gulenko O, Kim M, Yoon BH, Jeon M, Kim TH, Ha YS, Yang JM. Tapered catheter-based transurethral photoacoustic and ultrasonic endoscopy of the urinary system. OPTICS EXPRESS 2022; 30:26169-26181. [PMID: 36236812 DOI: 10.1364/oe.461855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/13/2022] [Indexed: 06/16/2023]
Abstract
Early diagnosis is critical for treating bladder cancer, as this cancer is very aggressive and lethal if detected too late. To address this important clinical issue, a photoacoustic tomography (PAT)-based transabdominal imaging approach was suggested in previous reports, in which its in vivo feasibility was also demonstrated based on a small animal model. However, successful translation of this approach to real clinical settings would be challenging because the human bladder is located at a depth that far exceeds the typical penetration depth of PAT (∼3 cm for in vivo cases). In this study, we developed a tapered catheter-based, transurethral photoacoustic and ultrasonic endoscopic probe with a 2.8 mm outer diameter to investigate whether the well-known benefits of PAT can be harnessed to resolve unmet urological issues, including early diagnosis of bladder cancer. To demonstrate the in vivo imaging capability of the proposed imaging probe, we performed a rabbit model-based urinary system imaging experiment and acquired a 3D microvasculature map distributed in the wall of the urinary system, which is a first in PAT, to the best of our knowledge. We believe that the results strongly support the use of this transurethral imaging approach as a feasible strategy for addressing urological diagnosis issues.
Collapse
|
18
|
Bass RD, Garcia-Garcia HM, Sanz-Sánchez J, Ziemer PGP, Bulant CA, Kuku KK, Kahsay YA, Beyene S, Melaku G, Otsuka T, Choi J, Fernández-Peregrina E, Erdogan E, Gonzalo N, Bourantas CV, Blanco PJ, Räber L. Human vs. machine vs. core lab for the assessment of coronary atherosclerosis with lumen and vessel contour segmentation with intravascular ultrasound. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2022; 38:1431-1439. [PMID: 38819542 DOI: 10.1007/s10554-022-02563-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022]
Abstract
A machine learning (ML) algorithm for automatic segmentation of intravascular ultrasound was previously validated. It has the potential to improve efficiency, accuracy and precision of coronary vessel segmentation compared to manual segmentation by interventional cardiology experts. The aim of this study is to compare the performance of human readers to the machine and against the readings from a Core Laboratory. This is a post-hoc, cross-sectional analysis of the IBIS-4 study. Forty frames were randomly selected and analyzed by 10 readers of varying expertise two separate times, 1 week apart. Their measurements of lumen, vessel, plaque areas, and plaque burden were performed in an offline software. Among humans, the intra-observer variability was not statistically significant. For the total 80 frames, inter-observer variability between human readers, the ML algorithm and Core Laboratory for lumen area, vessel area, plaque area and plaque burden were not statistically different. For lumen area, however, relative differences between the human readers and the Core Lab ranged from 0.26 to 12.61%. For vessel area, they ranged from 1.25 to 9.54%. Efficiency between the ML algorithm and the readers differed notably. Humans spent 47 min on average to complete the analyses, while the ML algorithm took on average less than 1 min. The overall lumen, vessel and plaque means analyzed by humans and the proposed ML algorithm are similar to those of the Core Lab. Machines, however, are more time efficient. It is warranted to consider use of the ML algorithm in clinical practice.
Collapse
Affiliation(s)
- Ronald D Bass
- School of Medicine, Georgetown University, Washington, DC, USA
| | - Hector M Garcia-Garcia
- Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA.
- Division of Interventional Cardiology of MedStar Cardiovascular Research Network at MedStar Washington Hospital Center, 110 Irving Street, Suite 4B-1, Washington, DC, 20010, USA.
| | - Jorge Sanz-Sánchez
- Hospital Universitari i Politecnic La Fe, Valencia, Spain
- Centro de Investigación Biomedica en Red (CIBERCV), Madrid, Spain
| | - Paulo G P Ziemer
- National Laboratory for Scientific Computing, Petrópolis, Brazil
| | - Carlos A Bulant
- National Scientific and Technical Research Council, Tandil, Argentina
| | - Kayode K Kuku
- Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Yirga A Kahsay
- Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Solomon Beyene
- Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Gebremedhin Melaku
- Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Tatsuhiko Otsuka
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - JooHee Choi
- School of Medicine, Georgetown University, Washington, DC, USA
| | | | - Emrah Erdogan
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Nieves Gonzalo
- Interventional Cardiology, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Christos V Bourantas
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Pablo J Blanco
- National Laboratory for Scientific Computing, Petrópolis, Brazil
| | - Lorenz Räber
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Dawson LP, Layland J. High-Risk Coronary Plaque Features: A Narrative Review. Cardiol Ther 2022; 11:319-335. [PMID: 35731471 PMCID: PMC9381667 DOI: 10.1007/s40119-022-00271-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/13/2022] [Indexed: 11/30/2022] Open
Abstract
Advances in coronary plaque imaging over the last few decades have led to an increased interest in the identification of novel high-risk plaque features that are associated with cardiovascular events. Existing practices focus on risk stratification and lipid monitoring for primary and secondary prevention of cardiac events, which is limited by a lack of assessment and treatment of vulnerable plaque. In this review, we summarize the multitude of studies that have identified plaque, haemodynamic and patient factors associated with risk of acute coronary syndrome. Future progress in multi-modal imaging strategies and in our understanding of high-risk plaque features could expand treatment options for coronary disease and improve patient outcomes.
Collapse
Affiliation(s)
- Luke P Dawson
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia.,Department of Cardiology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Jamie Layland
- Department of Medicine, Monash University, Clayton campus, Melbourne, VIC, Australia. .,Department of Cardiology, Peninsula Health, 2 Hastings Rd, Frankston, VIC, 3199, Australia.
| |
Collapse
|
20
|
Kong R, Dai C, Zhang Q, Gao L, Chen Z, Song Y, Wu Z, Wang J, Wang S, Zheng H, Ma T. Integrated US-OCT-NIRF Tri-Modality Endoscopic Imaging System for Pancreaticobiliary Duct Imaging. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2022; 69:1970-1979. [PMID: 35377846 DOI: 10.1109/tuffc.2022.3164777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Pancreaticobiliary carcinomas is a highly malignant gastrointestinal tumor. Most pancreaticobiliary cancers arise from epithelial proliferation within the pancreaticobiliary ducts, referred to as pancreatic intraepithelial neoplasias (PanINs). Some PanINs are benign metaplasia, while others progress to invasive duct adenocarcinoma (IDAC). However, there is no standard program to diagnose the progression from PanINs to IDAC. In this study, we present a tri-modality imaging system, which integrates ultrasound (US), optical coherence tomography (OCT), and near-infrared fluorescence (NIRF) for pancreaticobiliary duct imaging. This system can obtain OCT, US, and NIRF images in real-time with a frame rate of 30 frames per second. For the endoscopy probe with an outer diameter of 0.9 mm, the US transducer and fiber ball lens were placed back to back. In vivo experiments were performed on the rectums of Sprague-Dawley rats to demonstrate the imaging performance of US, OCT, and fluorescence angiography. An ex vivo experiment on a human pancreatic duct was performed for a more accurate assessment of the pancreaticobiliary duct. The tomography images of rat rectums and human pancreatic ducts were correlated with hematoxylin and eosin (H&E) histology to check the measurement accuracy. The integrated tri-modality system has great clinical potential in mechanism studies, early diagnosis, and prognosis evaluation of malignant pancreaticobiliary carcinomas.
Collapse
|
21
|
Kim M, Lee KW, Kim K, Gulenko O, Lee C, Keum B, Chun HJ, Choi HS, Kim CU, Yang JM. Intra-instrument channel workable, optical-resolution photoacoustic and ultrasonic mini-probe system for gastrointestinal endoscopy. PHOTOACOUSTICS 2022; 26:100346. [PMID: 35313458 PMCID: PMC8933520 DOI: 10.1016/j.pacs.2022.100346] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/08/2022] [Indexed: 05/04/2023]
Abstract
There has been a long-standing expectation that the optical-resolution embodiment of photoacoustic tomography could have a substantial impact on gastrointestinal endoscopy by enabling microscopic visualization of the vasculature based on the endogenous contrast mechanism. Although multiple studies have demonstrated the in vivo imaging capability of a developed imaging device over the last decade, the implementation of such an endoscopic system that can be applied immediately when necessary via the instrument channel of a video endoscope has been a challenge. In this study, we developed a 3.38-mm diameter catheter-based, integrated optical-resolution photoacoustic and ultrasonic mini-probe system and successfully demonstrated its intra-instrument channel workability for the standard 3.7-mm diameter instrument channel of a clinical video endoscope based on a swine model. Through the instrument channel, we acquired the first in vivo dual-mode photoacoustic and ultrasonic endoscopic images from the esophagogastric junction of a swine. Further, in a rat colorectum in vivo imaging experiment, we visualized hierarchically developed mesh-like capillary networks with a hole size as small as ~50 µm, which suggests the potential level of image details that could be photoacoustically provided in clinical settings in the future.
Collapse
Affiliation(s)
- Minjae Kim
- Center for Photoacoustic Medical Instruments, Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| | - Kang Won Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, South Korea
| | - KiSik Kim
- Center for Photoacoustic Medical Instruments, Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| | - Oleksandra Gulenko
- Center for Photoacoustic Medical Instruments, Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| | - Cheol Lee
- Department of Physics, UNIST, Ulsan 44919, South Korea
| | - Bora Keum
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, South Korea
| | - Hoon Jai Chun
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, South Korea
| | - Hyuk Soon Choi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, South Korea
| | - Chae Un Kim
- Department of Physics, UNIST, Ulsan 44919, South Korea
| | - Joon-Mo Yang
- Center for Photoacoustic Medical Instruments, Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| |
Collapse
|
22
|
Lim J. Circuits on miniaturized ultrasound imaging system-on-a-chip: a review. Biomed Eng Lett 2022; 12:219-228. [PMID: 35892032 PMCID: PMC9308847 DOI: 10.1007/s13534-022-00228-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/25/2022] Open
Abstract
Trends of medical system move from a traditional in-person visit to virtual healthcare increases demands on point-of-care devices. Because ultrasound (US) is non-invasive, the demands highlight US imaging among other imaging modalities. Thanks to the development of US transducer technology, miniaturized US with application-specific integrated circuits (ASIC) have been researched. For example, applications that require small aperture sizes such as intravascular US (IVUS) and intra-cardiac echocardiography (ICE) require integration of system-on-a-chip (SoC) on the transducer. This paper reviews circuit techniques on the transmitter (TX) and receiver (RX) of the US imaging system. As TX circuits, pulser, T/RX switch, TX beamformer, and power management circuits are discussed. State-of-the-art transducer modeling, pre-amplifier, time-gain compensation, RX beamformer, quadrature sampler, and output driver are introduced as RX circuits.
Collapse
Affiliation(s)
- Jaemyung Lim
- Department of Electrical Engineering in Hanyang University, Seoul, South Korea
| |
Collapse
|
23
|
Olender ML, Niu Y, Marlevi D, Edelman ER, Nezami FR. Impact and Implications of Mixed Plaque Class in Automated Characterization of Complex Atherosclerotic Lesions. Comput Med Imaging Graph 2022; 97:102051. [DOI: 10.1016/j.compmedimag.2022.102051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/19/2021] [Accepted: 02/17/2022] [Indexed: 01/16/2023]
|
24
|
Li J, Shang C, Rong Y, Sun J, Cheng Y, He B, Wang Z, Li M, Ma J, Fu B, Ji X. Review on Laser Technology in Intravascular Imaging and Treatment. Aging Dis 2022; 13:246-266. [PMID: 35111372 PMCID: PMC8782552 DOI: 10.14336/ad.2021.0711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/11/2021] [Indexed: 12/14/2022] Open
Abstract
Blood vessels are one of the most essential organs, which nourish all tissues in our body. Once there are intravascular plaques or vascular occlusion, other organs and circulatory systems will not work properly. Therefore, it is necessary to detect abnormal blood vessels by intravascular imaging technologies for subsequent vascular treatment. The emergence of lasers and fiber optics promotes the development of intravascular imaging and treatment. Laser imaging techniques can obtain deep vascular images owing to light scattering and absorption properties. Moreover, photothermal and photomechanical effects of laser make it possible to treat vascular diseases accurately. In this review, we present the research progress and applications of laser techniques in intravascular imaging and treatment. Firstly, we introduce intravascular optical coherent tomography and intravascular photoacoustic imaging, which can obtain various information of plaques. Multimodal intravascular imaging techniques provide more information about intravascular plaques, which have an essential influence on intravascular imaging. Secondly, two laser techniques including laser angioplasty and endovenous laser ablation are discussed for the treatment of arterial and venous diseases, respectively. Finally, the outlook of laser techniques in blood vessels, as well as the integration of laser imaging and treatment are prospected in the section of discussions.
Collapse
Affiliation(s)
- Jing Li
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Ce Shang
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Yao Rong
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
- Medical Engineering Devices of Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Jingxuan Sun
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
| | - Yuan Cheng
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
| | - Boqu He
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
| | - Zihao Wang
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
| | - Ming Li
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Jianguo Ma
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
| | - Bo Fu
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
- Key Laboratory of Big Data-Based Precision Medicine Ministry of Industry and Information Technology, Interdisciplinary Innovation Institute of Medicine and Engineering, Beihang University, Beijing, China.
| | - Xunming Ji
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Neurosurgery Department of Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
25
|
Pitavastatin: Coronary Atherosclerotic Plaques Changes and Cardiovascular Prevention. High Blood Press Cardiovasc Prev 2022; 29:137-144. [DOI: 10.1007/s40292-021-00496-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/30/2021] [Indexed: 10/19/2022] Open
|
26
|
Dawson LP, Lum M, Nerleker N, Nicholls SJ, Layland J. Coronary Atherosclerotic Plaque Regression: JACC State-of-the-Art Review. J Am Coll Cardiol 2022; 79:66-82. [PMID: 34991791 DOI: 10.1016/j.jacc.2021.10.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/03/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022]
Abstract
Over the last 3 decades there have been substantial improvements in treatments aimed at reducing cardiovascular (CV) events. As these treatments have been developed, there have been parallel improvements in coronary imaging modalities that can assess plaque volumes and composition, using both invasive and noninvasive techniques. Plaque progression can be seen to precede CV events, and therefore, many studies have longitudinally assessed changes in plaque characteristics in response to various treatments, aiming to demonstrate plaque regression and improvements in high-risk features, with the rationale being that this will reduce CV events. In the past, decisions surrounding treatments for atherosclerosis have been informed by population-based risk scores for initiation in primary prevention and low-density lipoprotein cholesterol levels for titration in secondary prevention. If outcome data linking plaque regression to reduced CV events emerge, it may become possible to directly image plaque treatment response to guide management decisions.
Collapse
Affiliation(s)
- Luke P Dawson
- Department of Cardiology, Peninsula Health, Victoria, Australia; Monash University, Melbourne, Victoria, Australia; Department of Cardiology, The Royal Melbourne Hospital, Victoria, Australia; Department of Cardiology, The Alfred Hospital, Victoria, Australia
| | - Mark Lum
- Monash University, Melbourne, Victoria, Australia
| | - Nitesh Nerleker
- Monash University, Melbourne, Victoria, Australia; Department of Cardiology, Monash Health, Clayton, Victoria, Australia; The Baker Institute, Melbourne, Victoria, Australia
| | - Stephen J Nicholls
- Monash University, Melbourne, Victoria, Australia; Department of Cardiology, Monash Health, Clayton, Victoria, Australia
| | - Jamie Layland
- Department of Cardiology, Peninsula Health, Victoria, Australia; Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
27
|
Hou S, Li Y, Zou C, Li Y, Tang H, Liu Z, Chen S, Peng J. A Novel Distal Micromotor-Based Side-Looking Intravascular Ultrasound Transducer. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2022; 69:283-290. [PMID: 34550882 DOI: 10.1109/tuffc.2021.3114414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Cardiovascular disease has become one of the leading causes of death in China, accounting for 45.5% of all deaths in rural areas and 43.16% in urban areas. Hence, its early diagnosis is important. With the development of intravascular imaging technology, the intravascular ultrasound (IVUS) is widely used. The available commercial mechanical rotary side-looking IVUS (SL-IVUS) transducers are driven by external motors that use long flexible shafts to transmit the rotation. However, when the transducer passes through a long-curved blood vessel, it easily causes the nonuniform rotation distortion (NURD) of the image. A catheter which contains a distal motor and sodium chloride (NaCl) solution is presented in this study as an attempt to solve such issues. The NaCl solution is used to connect the transducer and micromotor so that the motor can directly drive the transducer to rotate and acquire the information of the blood vessel. The results showed that the center frequency and -6-dB fraction bandwidth of the single element were 47 MHz and 98%, respectively. The SL-IVUS catheter consists of a distal motor, with speed stability and high resolution, and has the potential to diagnose cardiovascular disease. This novel structure can decrease the dimension at the top of the catheter and reduce the risks of clinical diagnosis.
Collapse
|
28
|
Case BC, Shea C, Torguson R, Zhang C, Yerasi C, Medranda GA, Kuku KO, Garcia-Garcia HM, Mintz GS, Waksman R. Impact of baseline imaging of non-culprit coronary lesions and adverse events: Insight from LRP study. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2021; 39:1-5. [PMID: 34972665 DOI: 10.1016/j.carrev.2021.12.012] [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: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND/PURPOSE Intravascular ultrasound (IVUS) and near-infrared spectroscopy (NIRS) can identify vulnerable coronary atherosclerotic plaques. We aimed to compare the presence or absence of baseline intravascular imaging of non-culprit lesions and their subsequent adverse events. METHODS/MATERIALS We identified patients from the Lipid Rich Plaque (LRP) study who had a non-culprit-lesion adverse event and divided them into 2 cohorts: those with lesions detected with NIRS-IVUS imaging at baseline and those with lesions not imaged at baseline. RESULTS Overall, 73 patients had an adverse event (99 coronary segments) during the 24-month follow-up period. Among them, 41 patients (56.2%) had a non-culprit-lesion adverse event related to a coronary segment imaged at baseline, and 32 patients (43.8%) had a non-culprit-lesion adverse event adjudicated to a segment that was not scanned at baseline. Angiographic core laboratory analysis suggested that unscanned lesions were more often in the right coronary artery (~50%); branches of the left coronary artery, i.e., diagonal or left obtuse marginal arteries (~20%); smaller vessels; or more tortuous vessels; and less often in the left anterior descending or distal locations. There was a weak trend for acute severe events (adjudicated myocardial infarction and acute coronary syndrome) in patients with lesions not scanned at baseline (50.0% versus 36.6%, p = 0.250). CONCLUSIONS In patients with follow-up non-culprit-lesion adverse events, nearly half were not imaged with NIRS-IVUS at baseline. Because events related to non-imaged lesions were at least as severe as events related to imaged lesions, future clinical trials and clinical protocols should be designed to minimize this issue. CLINICAL TRIAL REGISTRATION The Lipid-Rich Plaque Study (LRP), https://clinicaltrials.gov/ct2/show/NCT02033694, NCT02033694.
Collapse
Affiliation(s)
- Brian C Case
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Corey Shea
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Rebecca Torguson
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cheng Zhang
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Charan Yerasi
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Giorgio A Medranda
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Kayode O Kuku
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Hector M Garcia-Garcia
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Gary S Mintz
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Ron Waksman
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA.
| |
Collapse
|
29
|
Kellnberger S, Wissmeyer G, Albaghdadi M, Piao Z, Li W, Mauskapf A, Rauschendorfer P, Tearney GJ, Ntziachristos V, Jaffer FA. Intravascular molecular-structural imaging with a miniaturized integrated near-infrared fluorescence and ultrasound catheter. JOURNAL OF BIOPHOTONICS 2021; 14:e202100048. [PMID: 34164943 PMCID: PMC8492488 DOI: 10.1002/jbio.202100048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/11/2021] [Accepted: 06/10/2021] [Indexed: 05/29/2023]
Abstract
Coronary artery disease (CAD) remains a leading cause of mortality and warrants new imaging approaches to better guide clinical care. We report on a miniaturized, hybrid intravascular catheter and imaging system for comprehensive coronary artery imaging in vivo. Our catheter exhibits a total diameter of 1.0 mm (3.0 French), equivalent to standalone clinical intravascular ultrasound (IVUS) catheters but enables simultaneous near-infrared fluorescence (NIRF) and IVUS molecular-structural imaging. We demonstrate NIRF-IVUS imaging in vitro in coronary stents using NIR fluorophores, and compare NIRF signal strengths for prism and ball lens sensor designs in both low and high scattering media. Next, in vivo intravascular imaging in pig coronary arteries demonstrates simultaneous, co-registered molecular-structural imaging of experimental CAD inflammation on IVUS and distance-corrected NIRF images. The obtained results suggest substantial potential for the NIRF-IVUS catheter to advance standalone IVUS, and enable comprehensive phenotyping of vascular disease to better assess and treat patients with CAD.
Collapse
Affiliation(s)
- Stephan Kellnberger
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Boston, MA 02114
| | - Georg Wissmeyer
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Boston, MA 02114
| | - Mazen Albaghdadi
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Boston, MA 02114
| | - Zhonglie Piao
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114
| | - Wenzhu Li
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Boston, MA 02114
| | - Adam Mauskapf
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Boston, MA 02114
| | - Philipp Rauschendorfer
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Germany
| | - Guillermo J. Tearney
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114
| | - Vasilis Ntziachristos
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Germany
| | - Farouc A. Jaffer
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Boston, MA 02114
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114
| |
Collapse
|
30
|
Zanchin C, Ueki Y, Losdat S, Fahrni G, Daemen J, Ondracek AS, Häner JD, Stortecky S, Otsuka T, Siontis GCM, Rigamonti F, Radu M, Spirk D, Kaiser C, Engstrom T, Lang I, Koskinas KC, Räber L. In vivo relationship between near-infrared spectroscopy-detected lipid-rich plaques and morphological plaque characteristics by optical coherence tomography and intravascular ultrasound: a multimodality intravascular imaging study. Eur Heart J Cardiovasc Imaging 2021; 22:824-834. [PMID: 31990323 DOI: 10.1093/ehjci/jez318] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/09/2020] [Indexed: 12/19/2022] Open
Abstract
AIMS We assessed morphological features of near-infrared spectroscopy (NIRS)-detected lipid-rich plaques (LRPs) by using optical coherence tomography (OCT) and intravascular ultrasound (IVUS). METHODS AND RESULTS IVUS-NIRS and OCT were performed in the two non-infarct-related arteries (non-IRAs) in patients undergoing percutaneous coronary intervention for treatment of an acute coronary syndrome. A lesion was defined as the 4 mm segment with the maximum amount of lipid core burden index (maxLCBI4mm) of each LRP detected by NIRS. We divided the lesions into three groups based on the maxLCBI4mm value: <250, 250-399, and ≥400. OCT analysis and IVUS analysis were performed blinded for NIRS. We measured fibrous cap thickness (FCT) by using a semi-automated method. A total of 104 patients underwent multimodality imaging of 209 non-IRAs. NIRS detected 299 LRPs. Of those, 41% showed a maxLCBI4mm <250, 39% a maxLCBI4mm 251-399, and 19% a maxLCBI4mm ≥400. LRPs with a maxLCBI4mm ≥400, as compared with LRPs with a maxLCBI4mm 250-399 and <250, were more frequently thin-cap fibroatheroma (TCFA) (42.1% vs. 5.1% and 0.8%; P < 0.001) with a smaller minimum FCT (80 μm vs. 110 μm and 120 μm; P < 0.001); a higher IVUS-derived percent atheroma volume (53% vs. 53% and 44%; P < 0.001) and a higher remodelling index (1.08 vs. 1.02 and 1.01; P < 0.001). MaxLCBI4mm correlated with OCT-derived FCT (r = 0.404; P < 0.001) and was the best predictor for TCFA with an optimal cut-off value of 401 (area under the curve = 0.882; P < 0.001). CONCLUSION LRPs with increasing maxLCBI4mm exhibit OCT and IVUS features of presumed plaque vulnerability including TCFA morphology, increased plaque burden, and positive remodelling.
Collapse
Affiliation(s)
- Christian Zanchin
- Cardiology Department, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
| | - Yasushi Ueki
- Cardiology Department, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
| | - Sylvain Losdat
- Department of Social and Preventive Medicine, Clinical Trials Unit, Institute of Social and Preventive Medicine, Bern University Hospital, 3012 Bern, Switzerland
| | - Gregor Fahrni
- Department of Cardiology, University Hospital Basel, 4031 Basel, Switzerland
| | - Joost Daemen
- Department of Cardiology, Erasmus Medical Center, 3015 Rotterdam, the Netherlands
| | - Anna S Ondracek
- Department of Cardiology, Medical University of Vienna, 1090 Vienna, Austria
| | - Jonas D Häner
- Cardiology Department, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
| | - Stefan Stortecky
- Cardiology Department, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
| | - Tatsuhiko Otsuka
- Cardiology Department, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
| | - George C M Siontis
- Cardiology Department, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
| | - Fabio Rigamonti
- Department of Cardiology, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Maria Radu
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - David Spirk
- Department of Pharmacology, Institute of Pharmacology, University of Bern, 3012 Bern, Switzerland
| | - Christoph Kaiser
- Department of Cardiology, University Hospital Basel, 4031 Basel, Switzerland
| | - Thomas Engstrom
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Irene Lang
- Department of Cardiology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Lorenz Räber
- Cardiology Department, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
31
|
Zanchin C, Koskinas KC, Ueki Y, Losdat S, Häner JD, Bär S, Otsuka T, Inderkum A, Jensen MRJ, Lonborg J, Fahrni G, Ondracek AS, Daemen J, van Geuns RJ, Iglesias JF, Matter CM, Spirk D, Juni P, Mach F, Heg D, Engstrom T, Lang I, Windecker S, Räber L. Effects of the PCSK9 antibody alirocumab on coronary atherosclerosis in patients with acute myocardial infarction: a serial, multivessel, intravascular ultrasound, near-infrared spectroscopy and optical coherence tomography imaging study-Rationale and design of the PACMAN-AMI trial. Am Heart J 2021; 238:33-44. [PMID: 33951415 DOI: 10.1016/j.ahj.2021.04.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/26/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND The risk for cardiovascular adverse events after acute myocardial infarction (AMI) remains high despite potent medical treatment including low-density lipoprotein cholesterol (LDL-C) lowering with statins. Proprotein convertase subtilisin/kexin type 9 (PCSK9) antibodies substantially reduce LDL-C when added to statin. Alirocumab, a monoclonal antibody to PCSK9, reduces major adverse cardiovascular events after AMI. The effects of alirocumab on coronary atherosclerosis including plaque burden, plaque composition and fibrous cap thickness in patients presenting with AMI remains unknown. AIMS To determine the effect of LDL-C lowering with alirocumab on top of high-intensity statin therapy on intravascular ultrasound (IVUS)-derived percent atheroma volume (PAV), near-infrared spectroscopy (NIRS)-derived maximum lipid core burden index within 4 mm (maxLCBI4 mm) and optical coherence tomography (OCT)-derived fibrous cap thickness (FCT) in patients with AMI. METHODS In this multicenter, double-blind, placebo-controlled trial, 300 patients with AMI (ST-elevation or non-ST-elevation myocardial infarction) were randomly assigned to receive either biweekly subcutaneous alirocumab (150 mg) or placebo beginning <24 hours after the acute event as add-on therapy to rosuvastatin 20 mg. Patients undergo serial IVUS, NIRS and OCT in the two non-infarct related arteries at baseline (at the time of treatment of the culprit lesion) and at 52 weeks. The primary endpoint, change in IVUS-derived PAV, and the powered secondary endpoints, change in NIRS-derived maxLCBI4 mm, and OCT-derived minimal FCT, will be assessed 52 weeks post randomization. SUMMARY The PACMAN-AMI trial will determine the effect of alirocumab on top of high-intensity statin therapy on high-risk coronary plaque characteristics as assessed by serial, multimodality intracoronary imaging in patients presenting with AMI. CLINICAL TRIAL REGISTRATION NCT03067844.
Collapse
Affiliation(s)
- Christian Zanchin
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Konstantinos C Koskinas
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yasushi Ueki
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sylvain Losdat
- Institute of Social and Preventive Medicine and Clinical Trials Unit, Bern University Hospital, Bern, Switzerland
| | - Jonas D Häner
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sarah Bär
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Tatsuhiko Otsuka
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andrea Inderkum
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Maria Radu Juul Jensen
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jacob Lonborg
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Gregor Fahrni
- Department of Cardiology, University Hospital Basel, Basel, Switzerland
| | - Anna S Ondracek
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Joost Daemen
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Juan F Iglesias
- Department of Cardiology, Geneva University Hospital, Geneva, Switzerland
| | - Christian M Matter
- Department of Cardiology, University Heart Center Zurich, University Hospital Zurich, Zurich, Switzerland
| | - David Spirk
- Department of Pharmacology, Bern University Hospital, Bern, Switzerland and Sanofi, Switzerland
| | - Peter Juni
- Department of Medicine and Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Francois Mach
- Department of Cardiology, Geneva University Hospital, Geneva, Switzerland
| | - Dik Heg
- Institute of Social and Preventive Medicine and Clinical Trials Unit, Bern University Hospital, Bern, Switzerland
| | - Thomas Engstrom
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Irene Lang
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Stephan Windecker
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Lorenz Räber
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
32
|
Cismaru G, Serban T, Tirpe A. Ultrasound Methods in the Evaluation of Atherosclerosis: From Pathophysiology to Clinic. Biomedicines 2021; 9:418. [PMID: 33924492 PMCID: PMC8070406 DOI: 10.3390/biomedicines9040418] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/09/2021] [Accepted: 04/10/2021] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is a key pathological process that causes a plethora of pathologies, including coronary artery disease, peripheral artery disease, and ischemic stroke. The silent progression of the atherosclerotic disease prompts for new surveillance tools that can visualize, characterize, and provide a risk evaluation of the atherosclerotic plaque. Conventional ultrasound methods-bright (B)-mode US plus Doppler mode-provide a rapid, cost-efficient way to visualize an established plaque and give a rapid risk stratification of the patient through the Gray-Weale standardization-echolucent plaques with ≥50% stenosis have a significantly greater risk of ipsilateral stroke. Although rather disputed, the measurement of carotid intima-media thickness (C-IMT) may prove useful in identifying subclinical atherosclerosis. In addition, contrast-enhanced ultrasonography (CEUS) allows for a better image resolution and the visualization and quantification of plaque neovascularization, which has been correlated with future cardiovascular events. Newly emerging elastography techniques such as strain elastography and shear-wave elastography add a new dimension to this evaluation-the biomechanics of the arterial wall, which is altered in atherosclerosis. The invasive counterpart, intravascular ultrasound (IVUS), enables an individualized assessment of the anti-atherosclerotic therapies, as well as a direct risk assessment of these lesions through virtual histology IVUS.
Collapse
Affiliation(s)
- Gabriel Cismaru
- Fifth Department of Internal Medicine, Cardiology-Rehabilitation, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Teodora Serban
- Medical Imaging Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania;
| | - Alexandru Tirpe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| |
Collapse
|
33
|
Leng J, Zhang J, Li C, Shu C, Wang B, Lin R, Liang Y, Wang K, Shen L, Lam KH, Xie Z, Gong X, Ge J, Song L. Multi-spectral intravascular photoacoustic/ultrasound/optical coherence tomography tri-modality system with a fully-integrated 0.9-mm full field-of-view catheter for plaque vulnerability imaging. BIOMEDICAL OPTICS EXPRESS 2021; 12:1934-1946. [PMID: 33996208 PMCID: PMC8086469 DOI: 10.1364/boe.420724] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 05/15/2023]
Abstract
Myocardial infarctions are most often caused by the so-called vulnerable plaques, usually featured as non-obstructive lesions with a lipid-rich necrotic core, thin-cap fibroatheroma, and large plaque size. The identification and quantification of these characteristics are the keys to evaluate plaque vulnerability. However, single modality intravascular methods, such as intravascular ultrasound, optical coherence tomography and photoacoustic, can hardly achieve all the comprehensive information to satisfy clinical needs. In this paper, for the first time, we developed a novel multi-spectral intravascular tri-modality (MS-IVTM) imaging system, which can perform 360° continuous rotation and pull-backing with a 0.9-mm miniature catheter and achieve simultaneous acquisition of both morphological characteristics and pathological compositions. Intravascular tri-modality imaging demonstrates the ability of our MS-IVTM system to provide macroscopic and microscopic structural information of the vessel wall, with identity and quantification of lipids with multi-wavelength excitation. This study offers clinicians and researchers a novel imaging tool to facilitate the accurate diagnosis of vulnerable atherosclerotic plaques. It also has the potential of clinical translations to help better identify and evaluate high-risk plaques during coronary interventions.
Collapse
Affiliation(s)
- Ji Leng
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Equal contribution
| | - Jinke Zhang
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Equal contribution
| | - Chenguang Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Equal contribution
| | - Chengyou Shu
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Boquan Wang
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Riqiang Lin
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yanmei Liang
- Institute of Modern Optics, Nankai University, Tianjin Key Laboratory of Micro-scale Optical Information Science and Technology, Tianjin, 300350, China
| | - Keqiang Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Shen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kwok-Ho Lam
- Department of Electrical Engineering, Research Institute for Smart Energy, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Zhihua Xie
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiaojing Gong
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liang Song
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
34
|
Zheng J, Sun B, Lin R, Teng Y, Zhao X, Xue Y. Association between the vertebrobasilar artery geometry and basilar artery plaques determined by high-resolution magnetic resonance imaging. BMC Neurosci 2021; 22:20. [PMID: 33765922 PMCID: PMC7992992 DOI: 10.1186/s12868-021-00624-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/09/2021] [Indexed: 11/29/2022] Open
Abstract
Background Atherosclerotic plaques are often present in regions of arteries with complicated flow patterns. Vascular morphology plays important role in hemodynamics. In this study, we investigated the relationship between the geometry of the vertebrobasilar artery system and presence of basilar artery (BA) plaque. Methods We enrolled 290 patients with posterior circulation ischemic stroke. We distinguished four configurations of the vertebrobasilar artery: Walking, Tuning Fork, Lambda, and No Confluence. Patients were divided into multi-bending (≥ 3 bends) and oligo-bending (< 3 bends) VA groups. The diameter of the vertebral artery (VA) and the number of bends in the intracranial VA segment were assessed using three-dimensional time-of-flight magnetic resonance angiography. High-resolution magnetic resonance imaging was used to evaluate BA plaques. Logistic regression models were used to determine the relationship between the geometry type and BA plaque prevalence. Results After adjusting for sex, age, body mass index ≥ 28, hypertension, and diabetes mellitus, the Walking, Lambda, and No Confluence geometries were associated with the presence of BA plaque (all p < 0.05). Patients with multi-bending VAs in both the Walking (20/28, 71.43% vs. 6/21, 28.57%, p = 0.003) and Lambda group (19/47, 40.43% vs. 21/97, 21.65%, p = 0.018) had more plaques compared to patients with oligo-bending VAs in these groups. In the Lambda group, the difference in diameter of bilateral VAs was larger in patients with BA plaques than that in patients without BA plaques (1.4 mm [IQR: 0.9–1.6 mm] vs. 0.9 mm [IQR: 0.6–1.3 mm], p < 0.001). Conclusions The Walking, Lambda, and No Confluence geometry, ≥ 3 bends in the VAs, and a large diameter difference between bilateral VAs are associated with the presence of BA plaque. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-021-00624-5.
Collapse
Affiliation(s)
- Jinmei Zheng
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Bin Sun
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Ruolan Lin
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Yongqi Teng
- Department of Radiology, Changle District Hospital of Fuzhou, Fuzhou, 350299, Fujian, China
| | - Xihai Zhao
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Yunjing Xue
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
35
|
Close Association of Matrix Metalloproteinase-9 Levels With the Presence of Thin-Cap Fibroatheroma in Acute Coronary Syndrome Patients: Assessment by Optical Coherence Tomography and Intravascular Ultrasonography. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2021; 32:5-10. [PMID: 33485858 DOI: 10.1016/j.carrev.2020.12.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Thin-cap fibroatheroma (TCFA) has been suggested as a precursor lesion of coronary plaque rupture. As elevated plasma matrix metalloproteinase-9 (MMP-9) levels have been documented in patients with acute coronary syndrome (ACS), we sought to determine whether the presence of TCFA is linked to MMP-9 levels in these patients. METHODS We evaluated 51 ACS patients with de novo culprit lesions who were examined via optical coherence tomography and intravascular ultrasound. Blood samples were obtained from the peripheral vein (PV) and the ostium and culprit lesion of the infarct-related coronary artery (CA) in the acute phase of ACS and from the PV in the chronic phase (8 months after ACS). RESULTS The plasma MMP-9 level in the acute phase was significantly higher than that in the chronic phase. Plasma MMP-9 levels at the culprit lesion of the infarct-related CA were significantly higher than, but positively correlated with those in the PV (10.9 (5.9-16.1) ng/mL and 8.9 (5.6-13.0) ng/mL, p < 0.0001, respectively; Spearman ρ = 0.84, p < 0.0001). Significantly higher PV plasma MMP-9 levels were observed in patients with TCFA than in patients without TCFA (12.1 (7.0-13.5) and 5.7 (4.0-8.2) ng/ml, p<0.0001, respectively). Further, plasma MMP-9 levels in the PV were positively correlated with the remodeling index (Spearman ρ = 0.29, p = 0.039) and negatively correlated with fibrous cap thickness (Spearman ρ = -0.42, p = 0.0021). Receiver operating characteristic curve analysis showed that the plasma MMP-9 levels in the PV could predict the presence of TCFA at a cut-off value of 9.9 ng/mL. CONCLUSIONS Plasma MMP-9 levels were closely associated with MMP-9 levels in the CA and were further linked with TCFA in patients with ACS.
Collapse
|
36
|
Zhang J, Lu S, Liao X, Feng Z. Construction of an intravascular ultrasound catheter with a micropiezoelectric motor internally installed. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2021; 92:015005. [PMID: 33514220 DOI: 10.1063/5.0020260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/27/2020] [Indexed: 06/12/2023]
Abstract
Intravascular ultrasound (IVUS) has become a useful tool in the detection of coronary artery disease. However, non-uniform rotation distortion (NURD) reduces the image quality. In order to suppress the influence of NURD, a piezoelectric motor that can meet the requirements of IVUS catheters has been proposed. The motor has a diameter of 1 mm and a length of 10 mm using the new polarization direction proposed in the paper. A 45° mirror is fixed on the top of the motor to reflect the ultrasound transmitted from the transducer. The manufacture and drive of the piezoelectric motor is simple, and the maximum speed of the piezoelectric motor can reach 6450 rpm under the voltage of 20Vp-p. The minimum power required by the rotating motor is only 0.038 W, which can be directly driven by the signal generator without a power amplifier. The motor can operate at a low voltage and still has a high and stable speed. Meanwhile, the speed of the motor is controllable and has a satisfactory stability with a maximum angular error of 8°. The images detected by the cooperation of the motor and the ultrasonic transducer are also shown, which indicates that the motor has the rotational stability that meets the imaging requirements and the potential for application in the IVUS catheter to help improve the image quality of the coronary arteries and prevent and help treat potential diseases.
Collapse
Affiliation(s)
- Junjian Zhang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, HeFei 230026, China
| | - Shaowei Lu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, HeFei 230026, China
| | - Xinxin Liao
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, HeFei 230026, China
| | - Zhihua Feng
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, HeFei 230026, China
| |
Collapse
|
37
|
Malaiapan Y, Leung M, White AJ. The role of intravascular ultrasound in percutaneous coronary intervention of complex coronary lesions. Cardiovasc Diagn Ther 2020; 10:1371-1388. [PMID: 33224763 DOI: 10.21037/cdt-20-189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Intravascular ultrasound (IVUS) is a catheter-based coronary imaging technique. It utilises the emission & subsequent detection of reflected high frequency (30-60 MHz) sound waves to create high resolution, cross-sectional images of the coronary artery. IVUS has been the cornerstone of intracoronary imaging for more than two decades. When compared to the invasive coronary angiogram which studies only the silhouette of the contrast-filled artery lumen, IVUS also crucially images the vessel wall. Because of this capability, IVUS has greatly facilitated understanding of the coronary atherosclerosis process. Such insights from IVUS reveal how commonly and extensively plain angiography underestimates the true extent of coronary plaque, the characteristics of plaques prone to rupture and cause acute coronary syndromes (lipid rich, thin cap atheroma), and a realisation of the widespread occurrence of vessel remodelling in response to atherosclerosis. Similarly, IVUS has historically provided salutary mechanistic insights that have guided many of the incremental advances in the techniques of percutaneous coronary intervention (PCI). Examples include mechanisms of in-stent restenosis, and the importance of high-pressure post-dilatation of stents to ensure adequate stent apposition and thereby reduce the occurrence of stent thrombosis. IVUS also greatly facilitates the choice of correct diameter and length of stent to implant. Overall, a compelling body of evidence indicates that use of intravascular ultrasound in PCI helps to achieve optimal technical results and to mitigate the risk of adverse cardiac events. In this review, the role of intravascular ultrasound as an adjunct to PCI in complex coronary lesions is explored. The complex coronary situations discussed are the left main stem, ostial stenoses, bifurcation stenoses, thrombotic lesions, the chronically occluded coronary artery, and calcified coronary artery disease. By thorough review of the available evidence, we establish that the advantages of IVUS guidance are particularly evident in each of these complex CAD subsets. In particular, some consider the use of IVUS to be almost mandatory in left main PCI. A comparison with other intracoronary imaging techniques is also explored.
Collapse
Affiliation(s)
- Yuvaraj Malaiapan
- Monash Heart, Monash Medical Centre, Clayton, VIC, Australia.,Department of Medicine, Monash University, Clayton, VIC, Australia
| | - Michael Leung
- Monash Heart, Monash Medical Centre, Clayton, VIC, Australia.,Department of Medicine, Monash University, Clayton, VIC, Australia
| | - Anthony J White
- Monash Heart, Monash Medical Centre, Clayton, VIC, Australia.,Department of Medicine, Monash University, Clayton, VIC, Australia
| |
Collapse
|
38
|
Sung JH, Jeong EY, Jeong JS. Intravascular Ultrasound Transducer by Using Polarization Inversion Technique for Tissue Harmonic Imaging: Modeling and Experiments. IEEE Trans Biomed Eng 2020; 67:3380-3391. [PMID: 32286955 DOI: 10.1109/tbme.2020.2986284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Intravascular ultrasound (IVUS) tissue harmonic imaging (THI) is a useful vessel imaging technique that can provide deep penetration depth as well as high spatial and contrast resolution. Typically, a high-frequency IVUS transducer for THI requires a broad bandwidth or dual-frequency bandwidth. However, it is very difficult to make an IVUS transducer with a frequency bandwidth covering from the fundamental frequency to the second harmonic or a dual-peak at the desired frequency. To solve this problem, in this study, we applied the polarization inversion technique (PIT) to the IVUS transducer for THI. The PIT makes it relatively easy to design IVUS transducers with suitable frequency characteristics for THI depending on the inversion ratio of the piezoelectric layer and specifications of the passive materials. In this study, two types of IVUS transducers based on the PIT were developed for THI. One is a front-side inversion layer (FSIL) transducer with a broad bandwidth, and the other is a back-side inversion layer (BSIL) transducer with a dual-frequency bandwidth. These transducers were designed using finite element analysis (FEA)-based simulation, and the prototype transducers were fabricated. Subsequently, the performance was evaluated by not only electrical impedance and pulse-echo response tests but also B-mode imaging tests with a 25 μm tungsten wire and tissue-mimicking gelatin phantoms. The FEA simulation and experimental results show that the proposed scheme can successfully implement the tissue harmonic IVUS image, and thus it can be one of the promising techniques for developing IVUS transducers for THI.
Collapse
|
39
|
Olender ML, Athanasiou LS, Michalis LK, Fotiadis DI, Edelman ER. A Domain Enriched Deep Learning Approach to Classify Atherosclerosis using Intravascular Ultrasound Imaging. IEEE JOURNAL OF SELECTED TOPICS IN SIGNAL PROCESSING 2020; 14:1210-1220. [PMID: 33520048 PMCID: PMC7845913 DOI: 10.1109/jstsp.2020.3002385] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Intravascular ultrasound (IVUS) imaging is widely used for diagnostic imaging in interventional cardiology. The detection and quantification of atherosclerosis from acquired images is typically performed manually by medical experts or by virtual histology IVUS (VH-IVUS) software. VH-IVUS analyzes backscattered radio frequency (RF) signals to provide a color-coded tissue map, and is the method of choice for assessing atherosclerotic plaque in situ. However, a significant amount of tissue cannot be analyzed in reasonable time because the method can be applied just once per cardiac cycle. Furthermore, only hardware and software compatible with RF signal acquisition and processing may be used. We present an image-based tissue characterization method that can be applied to entire acquisition sequences post hoc for the assessment of diseased vessels. The pixel-based method utilizes domain knowledge of arterial pathology and physiology, and leverages technological advances of convolutional neural networks to segment diseased vessel walls into the same tissue classes as virtual histology using only grayscale IVUS images. The method was trained and tested on patches extracted from VH-IVUS images acquired from several patients, and achieved overall accuracy of 93.5% for all segmented tissue. Imposing physically-relevant spatial constraints driven by domain knowledge was key to achieving such strong performance. This enriched approach offers capabilities akin to VH-IVUS without the constraints of RF signals or limited once-per-cycle analysis, offering superior potential information acquisition speed, reduced hardware and software requirements, and more widespread applicability. Such an approach may well yield promise for future clinical and research applications.
Collapse
Affiliation(s)
- Max L Olender
- Department of Mechanical Engineering and the Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Lambros S Athanasiou
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139 USA; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Lampros K Michalis
- Faculty of Medicine, School of Health Sciences, University of Ioannina and the 2nd Department of Cardiology, University Hospital of Ioannina, Ioannina, 45500 Greece
| | - Dimitris I Fotiadis
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, Ioannina, 45110 Greece; Department of Biomedical Research, Institute of Molecular Biology and Biotechnology - FORTH, Ioannina, 45110 Greece
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139 USA; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
40
|
Wang J, Zheng Z, Chan J, Yeow JTW. Capacitive micromachined ultrasound transducers for intravascular ultrasound imaging. MICROSYSTEMS & NANOENGINEERING 2020; 6:73. [PMID: 34567683 PMCID: PMC8433336 DOI: 10.1038/s41378-020-0181-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/28/2020] [Accepted: 05/23/2020] [Indexed: 05/27/2023]
Abstract
Intravascular ultrasound (IVUS) is a burgeoning imaging technology that provides vital information for the diagnosis of coronary arterial diseases. A significant constituent that enables the IVUS system to attain high-resolution images is the ultrasound transducer, which acts as both a transmitter that sends acoustic waves and a detector that receives the returning signals. Being the most mature form of ultrasound transducer available in the market, piezoelectric transducers have dominated the field of biomedical imaging. However, there are some drawbacks associated with using the traditional piezoelectric ultrasound transducers such as difficulties in the fabrication of high-density arrays, which would aid in the acceleration of the imaging speed and alleviate motion artifact. The advent of microelectromechanical system (MEMS) technology has brought about the development of micromachined ultrasound transducers that would help to address this issue. Apart from the advantage of being able to be fabricated into arrays with lesser complications, the image quality of IVUS can be further enhanced with the easy integration of micromachined ultrasound transducers with complementary metal-oxide-semiconductor (CMOS). This would aid in the mitigation of parasitic capacitance, thereby improving the signal-to-noise. Currently, there are two commonly investigated micromachined ultrasound transducers, piezoelectric micromachined ultrasound transducers (PMUTs) and capacitive micromachined ultrasound transducers (CMUTs). Currently, PMUTs face a significant challenge where the fabricated PMUTs do not function as per their design. Thus, CMUTs with different array configurations have been developed for IVUS. In this paper, the different ultrasound transducers, including conventional-piezoelectric transducers, PMUTs and CMUTs, are reviewed, and a summary of the recent progress of CMUTs for IVUS is presented.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of Systems Design Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Zhou Zheng
- Department of Systems Design Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Jasmine Chan
- Department of Systems Design Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - John T. W. Yeow
- Department of Systems Design Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| |
Collapse
|
41
|
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: 1.8] [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
|
42
|
Lim J, Tekes C, Arkan EF, Rezvanitabar A, Degertekin FL, Ghovanloo M. Highly Integrated Guidewire Ultrasound Imaging System-on-a-Chip. IEEE JOURNAL OF SOLID-STATE CIRCUITS 2020; 55:1310-1323. [PMID: 32341598 PMCID: PMC7184934 DOI: 10.1109/jssc.2020.2967551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this article, we present a highly integrated guidewire ultrasound (US) imaging system-on-a-chip (GUISoC) for vascular imaging. The SoC consists of a 16-channel US transmitter (Tx) and receiver (Rx) electronics, on-chip power management IC (PMIC), and quadrature sampler. Using a synthetic aperture imaging algorithm, a Tx/Rx pair, connected to capacitive micromachined ultrasound transducers (CMUTs), can be activated at any time. The Tx generates acoustic waves by driving the CMUT, while the Rx picks up the echo signal and amplify it to be delivered through an interconnect that is driven by a buffer. On-chip logic controls the pulsers that generate the high-voltage (HV)-pulse for Tx. An on-chip PMIC provides 1.8-, 5-, 39-, and 44-V supplies and a clock signal from the two interconnects besides GND. A quadrature sampler down-converts the Rx echo signal to baseband, reducing its bandwidth requirement for the output interconnect. The system design, including transimpedance amplifier (TIA) optimization, based on the equivalent circuit of a specific CMUT is presented. The SoC was fabricated by a 0.18-μm HV CMOS process, occupying 1.5-mm2 active area and consuming 25.2 and 44 mW from 1.8 to 44 V supplies, respectively. The US Tx and Rx show bandwidths of 32-42 and 32.7-37.5 MHz, respectively. The input-referred noise of the system was measured as 9.66 nA in band with 2-m-long 52 American Wire Gauge (AWG) wire interconnects. The functionality of the GUISoC was verified in vitro by imaging wire targets.
Collapse
Affiliation(s)
- Jaemyung Lim
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Coskun Tekes
- School of Computer Engineering, Kennesaw State University, Marietta, GA 30060 USA
| | - Evren F Arkan
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Ahmad Rezvanitabar
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - F Levent Degertekin
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | | |
Collapse
|
43
|
Ma X, Cao W. Single-Crystal High-Frequency Intravascular Ultrasound Transducer With 40- μ m Axial Resolution. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2020; 67:810-816. [PMID: 31794395 DOI: 10.1109/tuffc.2019.2956603] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Intravascular ultrasound (IVUS) is one of the most useful tools available today to assist intravascular stenting procedures. Having higher resolution is very important for helping doctors to evaluate the nature of atherosclerotic plaques. The current commercial IVUS systems have a spatial resolution of 70- [Formula: see text] in the axial direction and 200- [Formula: see text] in the lateral direction, which are insufficient for accurate diagnosis. We report here a three-matching-layer IVUS transducer design using a 0.72Pb(Mg1/3Nb2/3O3 - 0.28PbTiO3 single crystal, which can improve the axial resolution to [Formula: see text] without sacrificing the penetration depth. Wire phantom imaging and in vitro porcine coronary artery imaging show noticeably better axial resolution and similar penetration depth compared with a commercial IVUS transducer.
Collapse
|
44
|
Ishikawa M, Muramatsu T, Nanasato M, Nagasaka R, Takatsu H, Yoshiki Y, Hashimoto Y, Ohota M, Okumura M, Naruse H, Ishii J, Ito K, Takahashi H, Kamiya H, Yoshida Y, Ozaki Y. Associations of coronary plaque characteristics by integrated backscatter intravascular ultrasound with detectability of vessel external elastic lamina using optical frequency domain imaging in human coronary arteries: A sub-analysis of the MISTIC-1 trial. Catheter Cardiovasc Interv 2019; 94:947-955. [PMID: 31025511 DOI: 10.1002/ccd.28218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/23/2019] [Indexed: 11/07/2022]
Abstract
OBJECTIVES We sought to examine associations between plaque characteristics by intravascular ultrasound (IVUS) and detectability of external elastic lamina (EEL) by optical frequency domain imaging (OFDI) in human coronary arteries. BACKGROUND It is often challenging to detect EEL which represents vessel size by light-based imaging modalities due to light intensity attenuation through atherosclerotic plaque. METHODS IVUS and OFDI prior to stent implantation were sequentially investigated per protocol. We identified corresponding cross-sections by minimum lumen area (MLA) or just distally to side branches as anatomical landmarks. Plaque characterization was determined by integrated backscatter IVUS analysis. We categorized detectable EEL arc by OFDI into four groups: 0≤ and <1 quadrant (group 1), 1≤ and <2 quadrants (group 2), 2≤ and <3 quadrants (group 3), or 3≤ and <4 quadrants (group 4). RESULTS We prospectively studied 103 vessels in 93 patients with stable coronary artery disease. Corresponding 711 cross-sections were analyzed. Cross-sections with detectable EEL arc <2 quadrants (group 1 or 2) were observed in 86.1% of MLA sites but only in 29.3% of non-MLA sites (p < .05). Percentage plaque area (%PA) appeared to be the strongest predictor to detect EEL arc <2 quadrants with the cut-off of 60.3% (AUC 0.90; sensitivity 79.8%, specificity 85.5%). Lipid pool and calcification remained statistically significant in predicting detectable EEL arc <2 quadrants after adjustment with %PA. CONCLUSIONS Presence of large plaque burden, lipid pool, and calcification significantly predicts the detectability of EEL by OFDI assessment. Locations with detectable EEL arc <2 quadrants should thus be avoided for optimal stent landing zone.
Collapse
Affiliation(s)
- Masato Ishikawa
- Department of Cardiology, Cardiovascular Center, Fujita Health University Hospital, Toyoake, Japan
| | - Takashi Muramatsu
- Department of Cardiology, Cardiovascular Center, Fujita Health University Hospital, Toyoake, Japan
| | - Mamoru Nanasato
- Department of Cardiology, Cardiovascular Center, Japanese Red Cross Nagoya Daini Hospital, Nagoya, Japan
| | - Ryo Nagasaka
- Department of Cardiology, Cardiovascular Center, Fujita Health University Hospital, Toyoake, Japan
| | - Hidemaro Takatsu
- Department of Cardiology, Cardiovascular Center, Fujita Health University Hospital, Toyoake, Japan
| | - Yu Yoshiki
- Department of Cardiology, Cardiovascular Center, Fujita Health University Hospital, Toyoake, Japan
| | - Yosuke Hashimoto
- Department of Cardiology, Cardiovascular Center, Fujita Health University Hospital, Toyoake, Japan
| | - Masaya Ohota
- Department of Cardiology, Cardiovascular Center, Fujita Health University Hospital, Toyoake, Japan
| | - Masanori Okumura
- Department of Cardiology, Cardiovascular Center, Fujita Health University Hospital, Toyoake, Japan
| | - Hiroyuki Naruse
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Junichi Ishii
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Katsuyoshi Ito
- Department of Radiology, Fujita Health University Hospital, Toyoake, Japan
| | - Hiroshi Takahashi
- Division of Medical Statistics, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hiroki Kamiya
- Department of Cardiovascular Medicine, Gifu Heart Center, Gifu, Japan
| | - Yukihiko Yoshida
- Department of Cardiology, Cardiovascular Center, Japanese Red Cross Nagoya Daini Hospital, Nagoya, Japan
| | - Yukio Ozaki
- Department of Cardiology, Cardiovascular Center, Fujita Health University Hospital, Toyoake, Japan
| |
Collapse
|
45
|
Zhao T, Desjardins AE, Ourselin S, Vercauteren T, Xia W. Minimally invasive photoacoustic imaging: Current status and future perspectives. PHOTOACOUSTICS 2019; 16:100146. [PMID: 31871889 PMCID: PMC6909166 DOI: 10.1016/j.pacs.2019.100146] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/26/2019] [Accepted: 09/30/2019] [Indexed: 05/09/2023]
Abstract
Photoacoustic imaging (PAI) is an emerging biomedical imaging modality that is based on optical absorption contrast, capable of revealing distinct spectroscopic signatures of tissue at high spatial resolution and large imaging depths. However, clinical applications of conventional non-invasive PAI systems have been restricted to examinations of tissues at depths less than a few cm due to strong light attenuation. Minimally invasive photoacoustic imaging (miPAI) has greatly extended the landscape of PAI by delivering excitation light within tissue through miniature fibre-optic probes. In the past decade, various miPAI systems have been developed with demonstrated applicability in several clinical fields. In this article, we present an overview of the current status of miPAI and our thoughts on future perspectives.
Collapse
Affiliation(s)
- Tianrui Zhao
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing St Thomas’ Hospital London, London SE1 7EH, United Kingdom
| | - Adrien E. Desjardins
- Department of Medical Physics and Biomedical Engineering, University College London, Gower Street, London WC1E 6BT, United Kingdom
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences, University College London, Charles Bell House, 67-73 Riding House Street, London W1W 7EJ, United Kingdom
| | - Sebastien Ourselin
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing St Thomas’ Hospital London, London SE1 7EH, United Kingdom
- Department of Medical Physics and Biomedical Engineering, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Tom Vercauteren
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing St Thomas’ Hospital London, London SE1 7EH, United Kingdom
- Department of Medical Physics and Biomedical Engineering, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Wenfeng Xia
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing St Thomas’ Hospital London, London SE1 7EH, United Kingdom
- Department of Medical Physics and Biomedical Engineering, University College London, Gower Street, London WC1E 6BT, United Kingdom
| |
Collapse
|
46
|
Prognostic Value of Intravascular Ultrasound in Patients With Coronary Artery Disease. J Am Coll Cardiol 2019; 72:2003-2011. [PMID: 30336823 DOI: 10.1016/j.jacc.2018.08.2140] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND It has been shown that intravascular ultrasound (IVUS) and radiofrequency (RF-)IVUS can detect high-risk coronary plaque characteristics. OBJECTIVES The authors studied the long-term prognostic value of (RF-)IVUS-derived plaque characteristics in patients with coronary artery disease (CAD) undergoing coronary angiography. METHODS From 2008 to 2011, (RF-)IVUS was performed in 1 nonstenotic segment of a nonculprit coronary artery in 581 patients undergoing coronary angiography for acute coronary syndrome (ACS) or stable angina. The pre-defined primary endpoint was major adverse cardiovascular events (MACE), defined as the composite of all-cause death, nonfatal ACS, or unplanned revascularization. Hazard ratios (HRs) were adjusted for age, sex, and clinical risk factors. RESULTS During a median follow-up of 4.7 years, 152 patients (26.2%) had MACE. The presence of a lesion with a minimal luminal area ≤4.0 mm2 was independently associated with MACE (HR: 1.49; 95% CI: 1.07 to 2.08; p = 0.020), whereas the presence of a thin-cap fibroatheroma lesion or a lesion with a plaque burden ≥70% on its own were not. Results were comparable when the composite endpoint included cardiac death instead of all-cause death. The presence of a lesion with a plaque burden of ≥70% was independently associated with the composite endpoint of cardiac death, nonfatal ACS, or unplanned revascularization after exclusion of culprit lesion-related events (HR: 1.66; 95% CI: 1.06 to 2.58; p = 0.026). Likewise, each 10-U increase in segmental plaque burden was independently associated with a 26% increase in risk of this composite endpoint (HR: 1.26 per 10-U increase; 95% CI: 1.03 to 1.52; p = 0.022). CONCLUSIONS IVUS-derived small luminal area and large plaque burden, and not RF-IVUS-derived compositional plaque features on their own, predict adverse cardiovascular outcome during long-term follow-up in patients with CAD. (The European Collaborative Project on Inflammation and Vascular Wall Remodeling in Atherosclerosis-Intravascular Ultrasound Study [AtheroRemoIVUS]; NCT01789411).
Collapse
|
47
|
de Boer S, Baran Y, Garcia-Garcia HM, Eskin I, Lenzen MJ, Kleber ME, Regar E, de Jaegere PJ, Ligthart JM, van Geuns RJ, Lehtimäki T, Laaksonen R, Boersma E, Marz W, Halperin E, Serruys PW, Koenig W. The European Collaborative Project on Inflammation and Vascular Wall Remodeling in Atherosclerosis - Intravascular Ultrasound (ATHEROREMO-IVUS) study. EUROINTERVENTION 2019; 14:194-203. [PMID: 28943493 DOI: 10.4244/eij-d-17-00180] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
AIMS The European Collaborative Project on Inflammation and Vascular Wall Remodeling in Atherosclerosis - Intravascular Ultrasound (ATHEROREMO-IVUS) study was designed as an exploratory clinical study in order to investigate the associations between genetic variation, coronary atherosclerosis phenotypes, and plaque vulnerability as determined by IVUS. METHODS AND RESULTS The ATHEROREMO-IVUS study was a prospective, observational study of 581 patients with stable angina pectoris or acute coronary syndrome (ACS) who were referred for coronary angiography to the Thoraxcenter, Rotterdam, enriched with 265 IBIS-2 participants (total population, n=846). Prior to catheterisation, blood samples were drawn for genetic analyses. During the catheterisation procedure, IVUS was performed in a non-culprit coronary artery. The primary endpoint was the presence of vulnerable plaque as determined by IVUS virtual histology (VH). In addition, we performed a genome-wide association study of plaque morphology. We observed strong signals associated with plaque morphology in several chromosomal regions: twelve SNPs (rs17300022, rs6904106, rs17177818, rs2248165, rs2477539, rs16865681, rs2396058, rs4753663, rs4082252, rs6932, rs12862206, rs6780676) in or near eight different genes (GNA12, NMBR, SFMBT2, CUL3, SESN3, SLC22A25, EFBN2, SEC62) were most significant. CONCLUSIONS In conclusion, we found twelve SNPs in or in the proximity of eight genes, which were possibly associated with markers of vulnerable plaque. ClinicalTrials.gov Identifier: NCT01789411.
Collapse
|
48
|
Choi SSS, Lashkari B, Mandelis A, Son J, Alves-Kotzev N, Foster SF, Harduar M, Courtney B. Frequency-domain differential photoacoustic radar: theory and validation for ultrasensitive atherosclerotic plaque imaging. JOURNAL OF BIOMEDICAL OPTICS 2019; 24:1-12. [PMID: 31197987 PMCID: PMC6977017 DOI: 10.1117/1.jbo.24.6.066003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/21/2019] [Indexed: 05/10/2023]
Abstract
Lipid composition of atherosclerotic plaques is considered to be highly related to plaque vulnerability. Therefore, a specific diagnostic or imaging modality that can sensitively evaluate plaques' necrotic core is desirable in atherosclerosis imaging. In this regard, intravascular photoacoustic (IVPA) imaging is an emerging plaque detection technique that provides lipid-specific chemical information from an arterial wall with great optical contrast and long acoustic penetration depth. While, in the near-infrared window, a 1210-nm optical source is usually chosen for IVPA applications since lipids exhibit a strong absorption peak at that wavelength, the sensitivity problem arises in the conventional single-ended systems as other arterial tissues also show some degree of absorption near that spectral region, thereby generating undesirably interfering photoacoustic (PA) signals. A theory of the high-frequency frequency-domain differential photoacoustic radar (DPAR) modality is introduced as a unique detection technique for accurate and molecularly specific evaluation of vulnerable plaques. By assuming two low-power continuous-wave optical sources at ∼1210 and ∼970 nm in a differential manner, DPAR theory and the corresponding simulation/experiment studies suggest an imaging modality that is only sensitive and specific to the spectroscopically defined imaging target, cholesterol.
Collapse
Affiliation(s)
- Sung Soo Sean Choi
- University of Toronto, Center for Advanced Diffusion-Wave and Photoacoustic Technologies, Department of Mechanical and Industrial Engineering, Toronto, Ontario, Canada
| | - Bahman Lashkari
- University of Toronto, Center for Advanced Diffusion-Wave and Photoacoustic Technologies, Department of Mechanical and Industrial Engineering, Toronto, Ontario, Canada
| | - Andreas Mandelis
- University of Toronto, Center for Advanced Diffusion-Wave and Photoacoustic Technologies, Department of Mechanical and Industrial Engineering, Toronto, Ontario, Canada
| | - Jungik Son
- Sunnybrook Research Institute, Physical Sciences Department, Toronto, Ontario, Canada
| | - Natasha Alves-Kotzev
- Sunnybrook Research Institute, Physical Sciences Department, Toronto, Ontario, Canada
| | - Stuart F. Foster
- Sunnybrook Research Institute, Physical Sciences Department, Toronto, Ontario, Canada
| | | | - Brian Courtney
- Sunnybrook Research Institute, Physical Sciences Department, Toronto, Ontario, Canada
- Conavi Medical, Toronto, Ontario, Canada
| |
Collapse
|
49
|
Jamthikar A, Gupta D, Khanna NN, Araki T, Saba L, Nicolaides A, Sharma A, Omerzu T, Suri HS, Gupta A, Mavrogeni S, Turk M, Laird JR, Protogerou A, Sfikakis PP, Kitas GD, Viswanathan V, Pareek G, Miner M, Suri JS. A Special Report on Changing Trends in Preventive Stroke/Cardiovascular Risk Assessment Via B-Mode Ultrasonography. Curr Atheroscler Rep 2019; 21:25. [PMID: 31041615 DOI: 10.1007/s11883-019-0788-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Cardiovascular disease (CVD) and stroke risk assessment have been largely based on the success of traditional statistically derived risk calculators such as Pooled Cohort Risk Score or Framingham Risk Score. However, over the last decade, automated computational paradigms such as machine learning (ML) and deep learning (DL) techniques have penetrated into a variety of medical domains including CVD/stroke risk assessment. This review is mainly focused on the changing trends in CVD/stroke risk assessment and its stratification from statistical-based models to ML-based paradigms using non-invasive carotid ultrasonography. RECENT FINDINGS In this review, ML-based strategies are categorized into two types: non-image (or conventional ML-based) and image-based (or integrated ML-based). The success of conventional (non-image-based) ML-based algorithms lies in the different data-driven patterns or features which are used to train the ML systems. Typically these features are the patients' demographics, serum biomarkers, and multiple clinical parameters. The integrated (image-based) ML-based algorithms integrate the features derived from the ultrasound scans of the arterial walls (such as morphological measurements) with conventional risk factors in ML frameworks. Even though the review covers ML-based system designs for carotid and coronary ultrasonography, the main focus of the review is on CVD/stroke risk scores based on carotid ultrasound. There are two key conclusions from this review: (i) fusion of image-based features with conventional cardiovascular risk factors can lead to more accurate CVD/stroke risk stratification; (ii) the ability to handle multiple sources of information in big data framework using artificial intelligence-based paradigms (such as ML and DL) is likely to be the future in preventive CVD/stroke risk assessment.
Collapse
Affiliation(s)
- Ankush Jamthikar
- Department of ECE, Visvesvaraya National Institute of Technology, Nagpur, Maharashtra, India
| | - Deep Gupta
- Department of ECE, Visvesvaraya National Institute of Technology, Nagpur, Maharashtra, India
| | - Narendra N Khanna
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi, India
| | - Tadashi Araki
- Division of Cardiovascular Medicine, Toho University, Tokyo, Japan
| | - Luca Saba
- Department of Radiology, University of Cagliari, Cagliari, Italy
| | - Andrew Nicolaides
- Vascular Screening and Diagnostic Centre, University of Cyprus, Nicosia, Cyprus
| | - Aditya Sharma
- Cardiovascular Medicine, University of Virginia, Charlottesville, VA, USA
| | - Tomaz Omerzu
- Department of Neurology, University Medical Centre Maribor, Maribor, Slovenia
| | | | - Ajay Gupta
- Department of Radiology, Cornell Medical Center, New York, NY, USA
| | - Sophie Mavrogeni
- Cardiology Clinic, Onassis Cardiac Surgery Center, Athens, Greece
| | - Monika Turk
- Department of Neurology, University Medical Centre Maribor, Maribor, Slovenia
| | - John R Laird
- Heart and Vascular Institute, Adventist Health St. Helena, St. Helena, CA, USA
| | - Athanasios Protogerou
- Department of Cardiovascular Prevention & Research Unit Clinic & Laboratory of Pathophysiology
- , National and Kapodistrian University of Athens, Athens, Greece
| | - Petros P Sfikakis
- Rheumatology Unit, National Kapodistrian University of Athens, Athens, Greece
| | - George D Kitas
- R&D Academic Affairs, Dudley Group NHS Foundation Trust, Dudley, UK
| | - Vijay Viswanathan
- MV Hospital for Diabetes and Professor M Viswanathan Diabetes Research Centre, Chennai, India
| | - Gyan Pareek
- Minimally Invasive Urology Institute, Brown University, Providence, RI, USA
| | - Martin Miner
- Men's Health Center, Miriam Hospital Providence, Providence, RI, USA
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, 95661, USA.
| |
Collapse
|
50
|
Zaman RT, Yousefi S, Chibana H, Ikeno F, Long SR, Gambhir SS, Chin FT, McConnell MV, Xing L, Yeung A. In Vivo Translation of the CIRPI System: Revealing Molecular Pathology of Rabbit Aortic Atherosclerotic Plaques. J Nucl Med 2019; 60:1308-1316. [PMID: 30737298 DOI: 10.2967/jnumed.118.222471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/13/2018] [Indexed: 01/13/2023] Open
Abstract
Thin-cap fibroatheroma (TCFA) are the unstable lesions in coronary artery disease that are prone to rupture, resulting in substantial morbidity and mortality worldwide. However, their small size and complex morphologic and biologic features make early detection and risk assessment difficult. We tested our newly developed catheter-based Circumferential-Intravascular-Radioluminescence-Photoacoustic-Imaging (CIRPI) system in vivo to enable detection and characterization of vulnerable plaque structure and biology in rabbit abdominal aorta. Methods: The CIRPI system includes a novel optical probe combining circumferential radioluminescence imaging and photoacoustic tomography (PAT). The probe's CaF2:Eu-based scintillating imaging window captures radioluminescence images (360° view) of plaques by detecting β-particles during 18F-FDG decay. A tunable laser-based PAT characterizes tissue constituents of plaque at 7 different wavelengths-540 and 560 nm (calcification), 920 nm (cholesteryl ester), 1040 nm (phospholipids), 1180 nm (elastin/collagen), 1210 nm (cholesterol), and 1235 nm (triglyceride). A single B-scan is concatenated from 330 A-lines captured during a 360° rotation. The abdominal aorta was imaged in vivo in both atherosclerotic rabbits (Watanabe Heritable Hyper Lipidemic [WHHL], 13-mo-old male, n = 5) and controls (New Zealand White, n = 2). Rabbits were fasted for 6 h before 5.55 × 107 Bq (1.5 mCi) of 18F-FDG were injected 1 h before the imaging procedure. Rabbits were anesthetized, and the right or left common carotid artery was surgically exposed. An 8 French catheter sheath was inserted into the common carotid artery, and a 0.035-cm (0.014-in) guidewire was advanced to the iliac artery, guided by x-ray fluoroscopy. A bare metal stent was implanted in the dorsal abdominal aorta as a landmark, followed by the 7 French imaging catheters that were advanced up to the proximal stent edge. Our CIRPI and clinical optical coherence tomography (OCT) were performed using pullback and nonocclusive flushing techniques. After imaging with the CIRPI system, the descending aorta was flushed with contrast agent, and OCT images were obtained with a pullback speed of 20 mm/s, providing images at 100 frames/s. Results were verified with histochemical analysis. Results: Our CIRPI system successfully detected the locations and characterized both stable and vulnerable aortic plaques in vivo among all WHHL rabbits. Calcification was detected from the stable plaque (540 and 560 nm), whereas TCFA exhibited phospholipids/cholesterol (1040 nm, 1210 nm). These findings were further verified with the clinical OCT system showing an area of low attenuation filled with lipids within TCFA. PAT images illustrated broken elastic fiber/collagen that could be verified with the histochemical analysis. All WHHL rabbits exhibited sparse to severe macrophages. Only 4 rabbits showed both moderate-to-severe level of calcifications and cholesterol clefts. However, all rabbits exhibited broken elastic fibers and collagen deposition. Control rabbits showed normal wall thickness with no presence of plaque tissue compositions. These findings were verified with OCT and histochemical analysis. Conclusion: Our novel multimodality hybrid system has been successfully translated to in vivo evaluation of atherosclerotic plaque structure and biology in a preclinical rabbit model. This system proposed a paradigm shift that unites molecular and pathologic imaging technologies. Therefore, the system may enhance the clinical evaluation of TCFA, as well as expand our understanding of coronary artery disease.
Collapse
Affiliation(s)
- Raiyan T Zaman
- Department of Radiology, Harvard Medical School, Boston, Massachusetts .,Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, Massachusetts.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Siavash Yousefi
- Division of Medical Physics, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Hidetoshi Chibana
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Fumiaki Ikeno
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Steven R Long
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Sanjiv S Gambhir
- Department of Radiology, Stanford University School of Medicine, Stanford, California.,Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, California.,Department of Bioengineering, Stanford University Schools of Medicine and of Engineering, Stanford, California; and
| | - Frederick T Chin
- Department of Radiology, Stanford University School of Medicine, Stanford, California.,Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, California
| | - Michael V McConnell
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California.,Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, California.,Verily Life Sciences, San Francisco, California
| | - Lei Xing
- Division of Medical Physics, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.,Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, California
| | - Alan Yeung
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|