1
|
Guo L, Wang J, Li N, Cui J, Su Y. Peptides for diagnosis and treatment of ovarian cancer. Front Oncol 2023; 13:1135523. [PMID: 37213272 PMCID: PMC10196167 DOI: 10.3389/fonc.2023.1135523] [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: 01/01/2023] [Accepted: 04/24/2023] [Indexed: 05/23/2023] Open
Abstract
Ovarian cancer is the most deadly gynecologic malignancy, and its incidence is gradually increasing. Despite improvements after treatment, the results are unsatisfactory and survival rates are relatively low. Therefore, early diagnosis and effective treatment remain two major challenges. Peptides have received significant attention in the search for new diagnostic and therapeutic approaches. Radiolabeled peptides specifically bind to cancer cell surface receptors for diagnostic purposes, while differential peptides in bodily fluids can also be used as new diagnostic markers. In terms of treatment, peptides can exert cytotoxic effects directly or act as ligands for targeted drug delivery. Peptide-based vaccines are an effective approach for tumor immunotherapy and have achieved clinical benefit. In addition, several advantages of peptides, such as specific targeting, low immunogenicity, ease of synthesis and high biosafety, make peptides attractive alternative tools for the diagnosis and treatment of cancer, particularly ovarian cancer. In this review, we focus on the recent research progress regarding peptides in the diagnosis and treatment of ovarian cancer, and their potential applications in the clinical setting.
Collapse
|
2
|
Geiger MA, Flumignan RLG, Sobreira ML, Avelar WM, Fingerhut C, Stein S, Guillaumon AT. Carotid Plaque Composition and the Importance of Non-Invasive in Imaging Stroke Prevention. Front Cardiovasc Med 2022; 9:885483. [PMID: 35651908 PMCID: PMC9149096 DOI: 10.3389/fcvm.2022.885483] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/27/2022] [Indexed: 12/24/2022] Open
Abstract
Luminal stenosis has been the standard feature for the current management strategies in patients with atherosclerotic carotid disease. Histological and imaging studies show considerable differences between plaques with identical degrees of stenosis. They indicate that specific plaque characteristics like Intraplaque hemorrhage, Lipid Rich Necrotic Core, Plaque Inflammation, Thickness and Ulceration are responsible for the increased risk of ischemic events. Intraplaque hemorrhage is defined by the accumulation of blood components within the plaque, Lipid Rich Necrotic Core is composed of macrophages loaded with lipid, Plaque Inflammation is defined as the process of atherosclerosis itself and Plaque thickness and Ulceration are defined as morphological features. Advances in imaging methods like Magnetic Resonance Imaging, Ultrasound, Computed Tomography and Positron Emission Tomography have enabled a more detailed characterization of the plaque, and its vulnerability is linked to these characteristics, changing the management of these patients based only on the degree of plaque stenosis. Studies like Rotterdam, ARIC, PARISK, CAPIAS and BIOVASC were essential to evaluate and prove the relevance of these characteristics with cerebrovascular symptoms. A better approach for the prevention of stroke is needed. This review summarizes the more frequent carotid plaque features and the available validation from recent studies with the latest evidence.
Collapse
Affiliation(s)
- Martin Andreas Geiger
- Division of Vascular Surgery, Department of Surgery, Universidade Estadual de Campinas—UNICAMP, São Paulo, Brazil
| | - Ronald Luiz Gomes Flumignan
- Division of Vascular and Endovascular Surgery, Department of Surgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marcone Lima Sobreira
- Division of Vascular and Endovascular Surgery, Department of Surgery and Orthopedics, Botucatu Medical School, Universidade Estadual Paulista (UNESP), São Paulo, Brazil
| | - Wagner Mauad Avelar
- Department of Neurology, Universidade Estadual de Campinas—UNICAMP, São Paulo, Brazil
| | - Carla Fingerhut
- Division of Radiology, Department of Anesthesiology and Radiology, Universidade Estadual de Campinas—UNICAMP, São Paulo, Brazil
| | - Sokrates Stein
- Division of Vascular Surgery, Department of Surgery, Universidade Estadual de Campinas—UNICAMP, São Paulo, Brazil
| | - Ana Terezinha Guillaumon
- Division of Vascular Surgery, Department of Surgery, Universidade Estadual de Campinas—UNICAMP, São Paulo, Brazil
| |
Collapse
|
3
|
Yang Y, Li Z, Liu Q, Guo Y, Mei Y, Lyu J, Zhao M, Feng Y, Xie G. Carotid arterial wall MRI of apolipoprotein e-deficient mouse at 7 T using DANTE-prepared variable-flip-angle rapid acquisition with relaxation enhancement. Magn Reson Imaging 2021; 86:1-9. [PMID: 34688846 DOI: 10.1016/j.mri.2021.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/20/2021] [Accepted: 10/17/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE To optimize a sequence combining the delay alternating with nutation for tailored excitation (DANTE) preparative module with the variable-flip-angle rapid acquisition with relaxation enhancement (VF-RARE) sequence (DANTE-VF-RARE) and to investigate its feasibility for vessel wall imaging in Apolipoprotein E-Deficient (ApoE-/-) mouse at 7 Tesla (T). MATERIALS AND METHODS Specific T1/T2 values were used for producing a sharper vessel wall in the variable-flip-angle optimization scheme. The DANTE RF pulse flip angle and pulse train length were optimized for maximizing the wall-lumen contrast. ApoE-/- (fed high fat diet for 20/40/ 60 weeks, n = 9/4/4) and wild-type mice (controls, n = 3) were imaged at 7 T using VF-RARE, DANTE-VF-RARE, time-of-flight (TOF) angiography, and multi-slice T1-weighted 2D RARE coupled with inflow outflow saturation bands (IOSB-RARE). Wall-lumen contrast-to-noise-ratio efficiency (CNReff), lumen area (LA), and wall area (WA) were compared between DANTE-VF-RARE and 2D IOSB-RARE sequences. Additionally, linear regression analysis was conducted between MR measurements and histomorphometric planimetry results. RESULTS Residual blood signal was observed in the four out of eighteen carotids on VF-RARE images, whereas it was significantly suppressed on DANTE-VF-RARE images. Compared with IOSB-RARE, DANTE-VF-RARE offered significantly improved CNReff (P < 0.001). The LA and WA were both comparable (P = 0.085 and 0.112, respectively) and showed excellent agreement between DANTE-VF-RARE and IOSB-RARE (ICC = 0.96 and 0.95, respectively). The luminal stenosis identified by DANTE-VF-RARE was in consistent with the results of TOF. Strong correlations were found between MR measurements and histopathological analysis for both WA (DANTE-VF-RARE: r = 0.92, slope = 0.94, P < 0.001; IOSB-RARE: r = 0.93, slope = 0.94, P < 0.001) and LA (DANTE-VF-RARE: r = 0.82, slope = 0.54, P < 0.001; IOSB-RARE: r = 0.78, slope = 0.50, P < 0.001). CONCLUSION DANTE-VF-RARE achieves effective blood signal suppression and is a feasible approach for the 3D carotid arterial wall imaging of ApoE-/- mouse at 7 T.
Collapse
Affiliation(s)
- Yuanbo Yang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhonghao Li
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiang Liu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China
| | - Yihao Guo
- MR Collaboration, Siemens Healthcare Ltd., Guangzhou, China
| | - Yingjie Mei
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China; Philips Healthcare, Guangzhou, China
| | - Jian Lyu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China
| | - Ming Zhao
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China.
| | - Guoxi Xie
- Department of Biomedical Engineering, The Sixth Affiliated Hospital, School of Basic Sciences, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Jin B, Lin H, Yuan J, Dong G, Huang K, Wu W, Chen X, Zhang L, Wang J, Liang X, Dai Y, Xu X, Zhou X, Zhu M, Li G, Cutfield WS, Hofman PL, Derraik JGB, Fu J. Abdominal Adiposity and Total Body Fat as Predictors of Cardiometabolic Health in Children and Adolescents With Obesity. Front Endocrinol (Lausanne) 2020; 11:579. [PMID: 33013688 PMCID: PMC7498567 DOI: 10.3389/fendo.2020.00579] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/16/2020] [Indexed: 12/20/2022] Open
Abstract
Objective: We aimed to assess the role of adipose tissue distribution in cardiometabolic risk (in particular insulin sensitivity) in a population of children and adolescents with obesity. Methods: In this cross-sectional study, participants were 479 children and adolescents with obesity (322 boys and 157 girls) aged 3 to 18 years attending the Children's Hospital at Zhejiang University School of Medicine (Hangzhou, China). Clinical assessments included anthropometry, body composition (DXA scans), carotid artery ultrasounds, and OGTT. Insulin sensitivity was assessed using the Matsuda index. Participants were stratified into groups by sex and pubertal stage. Key predictors were DXA-derived android-to-gynoid-fat ratio (A/G) and total body fat percentage (TBF%). Results: Irrespective of sex and pubertal stage, there was a strong association between increasing A/G (i.e., greater abdominal adiposity) and lower insulin sensitivity. In multivariable models, every 0.1 increase in A/G was associated with a reduction in insulin sensitivity in prepubertal boys [-29% (95% CI -36%, -20%); p < 0.0001], pubertal boys [-13% (95% CI -21%, -6%); p = 0.001], and pubertal girls [-16% (95% CI -24%, -6%); p = 0.002]. In contrast, TBF% was not associated with insulin sensitivity when A/G was adjusted for, irrespective of pubertal stage or sex. In addition, every 0.1 increase in A/G was associated with increased likelihood of dyslipidemia in prepubertal boys [adjusted odds ratio (aOR) 1.62 (95% CI 1.05, 2.49)], impaired glucose tolerance in pubertal boys [aOR 1.64 (95% CI 1.07, 2.51)] and pubertal girls [aOR 1.81 (95% CI 1.10, 2.98)], and odds of NAFLD in both prepubertal [aOR 2.57 (95% CI 1.56, 4.21)] and pubertal [aOR 1.69 (95% CI 1.18, 2.40)] boys. In contrast, higher TBF% was only associated with higher fasting insulin and ALT in pubertal boys, being also predictive of NAFLD in this group [aOR 1.15 per percentage point (95% CI 1.06, 1.26)], but was not associated with the likelihood of other cardiometabolic outcomes assessed in any group. Conclusions: A/G is a much stronger independent predictor of cardiometabolic risk factors in children and adolescents with obesity in China, particularly glucose metabolism.
Collapse
Affiliation(s)
- Binghan Jin
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Hu Lin
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jinna Yuan
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Guanping Dong
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ke Huang
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Wei Wu
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xuefeng Chen
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Li Zhang
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jinling Wang
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xinyi Liang
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yangli Dai
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaoqin Xu
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xuelian Zhou
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Mingqiang Zhu
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Guohua Li
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Wayne S. Cutfield
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start – National Science Challenge, University of Auckland, Auckland, New Zealand
| | - Paul L. Hofman
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - José G. B. Derraik
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start – National Science Challenge, University of Auckland, Auckland, New Zealand
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
- *Correspondence: José G. B. Derraik
| | - Junfen Fu
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Junfen Fu ;
| |
Collapse
|
5
|
Noninvasive Imaging Biomarkers of Vulnerable Coronary Plaques – a Clinical Update. JOURNAL OF INTERDISCIPLINARY MEDICINE 2019. [DOI: 10.2478/jim-2019-0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Atherosclerosis is a slow, progressive disease, its most common manifestation and most severe consequence being coronary artery disease, one of the main causes of mortality and morbidity worldwide. The vast majority of cardiovascular deaths are caused by complications of atherosclerosis, most often being represented by the rupture of an unstable coronary plaque, regularly triggered by inflammation. A vulnerable plaque is characterized by a large, lipid-rich necrotic core, a thin fibrous cap with macrophage infiltration, and the presence of multiple specific biomarkers such as positive remodeling, irregular calcifications, and low attenuation visible with coronary computed tomography angiography (CCTA). Identifying biomarkers that could predict the risk of plaque rupture with high accuracy would be a significant advance in predicting acute cardiac events in asymptomatic patients, furthermore guiding treatment of patients with this disease. The main indication of noninvasive imaging is to identify patients at risk based on the presence or absence of symptoms that can be related to myocardial ischemia. The diagnostic objective is to confirm or to exclude the presence of coronary plaques. Coronary imaging in asymptomatic individuals is used to estimate the risk of future cardiac events through the identification of non-obstructive high-risk plaques. The possibility to monitor the evolution of vulnerable plaques via noninvasive imaging techniques, prior to the occurrence of an acute clinical event, is the main goal in plaque imaging. This manuscript will be focusing on recent advances of noninvasive imaging of vulnerable coronary plaques.
Collapse
|
6
|
Tardif JC, Rhéaume E, Rhainds D, Dubé MP. Lipoprotein (a), arterial inflammation, and PCSK9 inhibition. Eur Heart J 2019; 40:2782-2784. [DOI: 10.1093/eurheartj/ehz087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Jean-Claude Tardif
- Research Centre, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Eric Rhéaume
- Research Centre, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - David Rhainds
- Research Centre, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Marie-Pierre Dubé
- Research Centre, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| |
Collapse
|
7
|
Li L, Li X, Jia Y, Fan J, Wang H, Fan C, Wu L, Si X, Hao X, Wu P, Yan M, Wang R, Hu G, Liu J, Wu Z, Hacker M, Li S. Sodium-fluoride PET-CT for the non-invasive evaluation of coronary plaques in symptomatic patients with coronary artery disease: a cross-correlation study with intravascular ultrasound. Eur J Nucl Med Mol Imaging 2018; 45:2181-2189. [PMID: 30171271 PMCID: PMC6182395 DOI: 10.1007/s00259-018-4122-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the 18F-sodium fluoride (18F-NaF) coronary uptake compared to coronary intravascular ultrasound (IVUS) in patients with symptomatic coronary artery disease. BACKGROUND 18F-NaF PET enables the assessment of vascular osteogenesis by interaction with surface hydroxyapatite, while IVUS enables both identification and quantification of intra-plaque components. METHODS Forty-four patients with symptomatic coronary artery disease were included in this prospective controlled trial, 32 of them (30 patients with unstable angina and 2 patients with stable angina), representing the final study cohort, got additional IVUS. All patients underwent cardiac 18F-NaF PET/CT and IVUS within 2 days. 18F-NaF maximum tissue-to-blood ratios (TBRmax) were calculated for 69 coronary plaques and correlated with IVUS plaque classification. RESULTS Significantly increased 18F-NaF uptake ratios were observed in fibrocalcific lesions (meanTBRmax = 1.42 ± 0.28), thin-cap atheroma with spotty calcifications (meanTBRmax = 1.32 ± 0.23), and thick-cap mixed atheroma (meanTBRmax = 1.28 ± 0.38), while fibrotic plaques showed no increased uptake (meanTBRmax = 0.96 ± 0.18). The 18F-NaF uptake ratio was consistently higher in atherosclerotic lesions with severe calcification (meanTBRmax = 1.34 ± 0.22). The regional 18F-NaF uptake was most likely localized in the border region of intensive calcification. Coronary lesions with positive 18F-NaF uptake showed some increased high-risk anatomical features on IVUS in comparison to 18F-NaF negative plaques. It included a significant severe plaque burden (70.1 ± 13.8 vs. 61.0 ± 13.8, p = 0.01) and positive remodeling index (1.03 ± 0.08 vs. 0.99 ± 0.07, p = 0.05), as well as a higher percentage of necrotic tissue (37.6 ± 13.3 vs. 29.3 ± 15.7, p = 0.02) in positive 18F-NaF lesions. CONCLUSIONS 18F-NaF coronary uptake may provide a molecular insight for the characterization of coronary atherosclerotic lesions. Specific regional uptake is needed to be determined by histology.
Collapse
Affiliation(s)
- Li Li
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Molecular Imaging Precision Medical Collaborative Innovation Center, Taiyuan, 030001, Shanxi Province, China
| | - Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| | - Yongping Jia
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiamao Fan
- Department of Cardiology, The Fourth People's Hospital of Linfen, Linfen, Shanxi, China
| | - Huifeng Wang
- Department of Cardiology, Taigang General Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chunyu Fan
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lei Wu
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xincheng Si
- Department of Cardiology, The Fourth People's Hospital of Linfen, Linfen, Shanxi, China
| | - Xinzhong Hao
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Molecular Imaging Precision Medical Collaborative Innovation Center, Taiyuan, 030001, Shanxi Province, China
| | - Ping Wu
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Molecular Imaging Precision Medical Collaborative Innovation Center, Taiyuan, 030001, Shanxi Province, China
| | - Min Yan
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Molecular Imaging Precision Medical Collaborative Innovation Center, Taiyuan, 030001, Shanxi Province, China
| | - Ruonan Wang
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Molecular Imaging Precision Medical Collaborative Innovation Center, Taiyuan, 030001, Shanxi Province, China
| | - Guang Hu
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Molecular Imaging Precision Medical Collaborative Innovation Center, Taiyuan, 030001, Shanxi Province, China
| | - Jianzhong Liu
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Molecular Imaging Precision Medical Collaborative Innovation Center, Taiyuan, 030001, Shanxi Province, China
| | - Zhifang Wu
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Molecular Imaging Precision Medical Collaborative Innovation Center, Taiyuan, 030001, Shanxi Province, China
| | - Marcus Hacker
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Molecular Imaging Precision Medical Collaborative Innovation Center, Taiyuan, 030001, Shanxi Province, China.,Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Sijin Li
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Molecular Imaging Precision Medical Collaborative Innovation Center, Taiyuan, 030001, Shanxi Province, China.
| |
Collapse
|
8
|
Present therapeutic role of cholesteryl ester transfer protein inhibitors. Pharmacol Res 2017; 128:29-41. [PMID: 29287689 DOI: 10.1016/j.phrs.2017.12.028] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/24/2017] [Accepted: 12/24/2017] [Indexed: 12/16/2022]
Abstract
Therapeutic interventions aimed at increasing high-density lipoprotein (HDL) levels in order to reduce the residual cardiovascular (CV) risk of optimally drug treated patients have not provided convincing results, so far. Transfer of cholesterol from extrahepatic tissues to the liver appears to be the major atheroprotective function of HDL, and an elevation of HDL levels could represent an effective strategy. Inhibition of the cholesteryl ester transfer protein (CETP), raising HDL-cholesterol (HDL-C) and apolipoprotein A-I (apoA-I) levels, reduces low-density lipoprotein-cholesterol (LDL-C) and apoB levels, thus offering a promising approach. Despite the beneficial influence on cholesterol metabolism, off-target effects and lack of reduction in CV events and mortality (with torcetrapib, dalcetrapib and evacetrapib) highlighted the complex mechanism of CETP inhibition. After the failure of the above mentioned inhibitors in phase III clinical development, possibly due to the short duration of the trials masking benefit, the secondary prevention REVEAL trial has recently shown that the inhibitor anacetrapib significantly raised HDL-C (+104%), reduced LDL-C (-18%), with a protective effect on major coronary events (RR, 0.91; 95%CI, 0.85-0.97; p = 0.004). Whether LDL-C lowering fully accounts for the CV benefit or if HDL-C-rise is a crucial factor still needs to be determined, although the reduction of non-HDL (-18%) and Lp(a) (-25%), should be also taken into account. In spite of the positive results of the REVEAL Study, Merck decided not to proceed in asking regulatory approval for anacetrapib. Dalcetrapib (Dal-GenE study) and CKD-519 remain the two molecules within this area still in clinical development.
Collapse
|
9
|
Bertrand MJ, Tardif JC. Inflammation and beyond: new directions and emerging drugs for treating atherosclerosis. Expert Opin Emerg Drugs 2016; 22:1-26. [PMID: 27927063 DOI: 10.1080/14728214.2017.1269743] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Cardiovascular (CV) atherosclerotic disease remains the leading cause of morbidity and mortality worldwide, despite the advances in contemporary therapies. Inflammation is an important process in atherosclerosis, leading to plaque rupture and acute coronary syndrome. Although statin therapy has substantially reduced CV events in primary and secondary prevention, many treated patients will have recurrent adverse CV events despite the standard of care. Thus, drug development aiming to target inflammatory pathways seems a promising avenue for novel therapies in atherosclerosis. Areas covered: Statins have been extensively studied and are the most effective lipid-lowering drugs available for CV prevention. Novel anti-inflammatory drugs are being tested in Phase II and III trials, targeting pathways like interleukin-1, leukotrienes, TNF-α, P-selectin, CCL2-CCR2 and MAP Kinase. Expert opinion: Novel anti-inflammatory therapies seem promising additions to address the residual CV risk present despite the current standard of care, but large clinical trials have not yet shown beneficial effects on clinical events. PCSK9 inhibitors have been shown to substantially reduce LDL-C, however their long-term safety and effects on CV risk are currently being investigated. Pharmacogenomics holds great potential in future lipid trials, enabling the identification of patients who will respond with greater benefits and smaller risk to therapies and to decrease failure rates in drug development, as genotype-dependent effects of the CETP inhibitor dalcetrapib were shown in the dal-OUTCOMES and dal-PLAQUE-2 trials.
Collapse
Affiliation(s)
- Marie-Jeanne Bertrand
- a Montreal Heart Institute, Department of medicine , Université de Montréal , Montreal , Canada
| | - Jean-Claude Tardif
- a Montreal Heart Institute, Department of medicine , Université de Montréal , Montreal , Canada
| |
Collapse
|
10
|
Validating Intravascular Imaging with Serial Optical Coherence Tomography and Confocal Fluorescence Microscopy. Int J Mol Sci 2016; 17:ijms17122110. [PMID: 27983695 PMCID: PMC5187910 DOI: 10.3390/ijms17122110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/25/2016] [Accepted: 12/09/2016] [Indexed: 01/20/2023] Open
Abstract
Atherosclerotic cardiovascular diseases are characterized by the formation of a plaque in the arterial wall. Intravascular ultrasound (IVUS) provides high-resolution images allowing delineation of atherosclerotic plaques. When combined with near infrared fluorescence (NIRF), the plaque can also be studied at a molecular level with a large variety of biomarkers. In this work, we present a system enabling automated volumetric histology imaging of excised aortas that can spatially correlate results with combined IVUS/NIRF imaging of lipid-rich atheroma in cholesterol-fed rabbits. Pullbacks in the rabbit aortas were performed with a dual modality IVUS/NIRF catheter developed by our group. Ex vivo three-dimensional (3D) histology was performed combining optical coherence tomography (OCT) and confocal fluorescence microscopy, providing high-resolution anatomical and molecular information, respectively, to validate in vivo findings. The microscope was combined with a serial slicer allowing for the imaging of the whole vessel automatically. Colocalization of in vivo and ex vivo results is demonstrated. Slices can then be recovered to be tested in conventional histology.
Collapse
|
11
|
Puri R, Nissen SE, Shao M, Elshazly MB, Kataoka Y, Kapadia SR, Tuzcu EM, Nicholls SJ. Non-HDL Cholesterol and Triglycerides. Arterioscler Thromb Vasc Biol 2016; 36:2220-2228. [DOI: 10.1161/atvbaha.116.307601] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 08/01/2016] [Indexed: 01/21/2023]
Abstract
Objectives—
Non–high-density lipoprotein cholesterol (non-HDLC) levels reflect the full burden of cholesterol transported in atherogenic lipoproteins. Genetic studies suggest a causal association between elevated triglycerides (TGs)-rich lipoproteins and atherosclerosis. We evaluated associations between achieved non-HDLC and TG levels on changes in coronary atheroma volume.
Approach and Results—
Data were analyzed from 9 clinical trials involving 4957 patients with coronary disease undergoing serial intravascular ultrasonography to assess changes in percent atheroma volume (ΔPAV) and were evaluated against on-treatment non-HDLC and TG levels. The effects of lower (<100 mg/dL) versus higher (≥100 mg/dL) achieved non-HDLC levels and lower (<200 mg/dL) versus higher (≥200 mg/dL) achieved TG levels were evaluated in populations with variable on-treatment low-density lipoprotein cholesterol (LDLC) </≥70 mg/dL and C-reactive protein </≥2 mg/L and in patients with or without diabetes mellitus. On-treatment non-HDLC levels linearly associated with ΔPAV. Overt PAV progression (ΔPAV>0) was associated with achieved TG levels >200 mg/dL, respectively. Lower on-treatment non-HDLC and TG levels associated with significant PAV regression compared with higher non-HDLC and TG levels across all levels of LDLC and C-reactive protein and irrespective of diabetic status (
P
<0.001 across all comparisons). ΔPAV were more strongly influenced by changes in non-HDLC (β=0.62;
P
<0.001) compared with changes in LDLC (β=0.51;
P
<0.001). Kaplan–Meier sensitivity analyses demonstrated significantly greater major adverse cardiovascular event rates in those with higher versus lower non-HDLC and TG levels, with an earlier separation of the non-HDLC compared with the LDLC curve.
Conclusions—
Achieved non-HDLC levels seem more closely associated with coronary atheroma progression than LDLC. Plaque progression associates with achieved TGs, but only above levels of 200 mg/dL. These observations support a more prominent role for non-HDLC (and possibly TG) lowering in combating residual cardiovascular risk.
Collapse
Affiliation(s)
- Rishi Puri
- From the Cleveland Clinic Coordinating Center for Clinical Research (C5R) (R.P., S.E.N., M.S.), and Department of Cardiovascular Medicine (R.P., S.E.N., M.B.E., S.R.K., E.M.T.), Cleveland Clinic, OH; Department of Medicine, University of Adelaide, Australia (R.P., S.J.N.); and South Australian Health and Medical Research Institute, Adelaide, Australia (Y.K., S.J.N.)
| | - Steven E. Nissen
- From the Cleveland Clinic Coordinating Center for Clinical Research (C5R) (R.P., S.E.N., M.S.), and Department of Cardiovascular Medicine (R.P., S.E.N., M.B.E., S.R.K., E.M.T.), Cleveland Clinic, OH; Department of Medicine, University of Adelaide, Australia (R.P., S.J.N.); and South Australian Health and Medical Research Institute, Adelaide, Australia (Y.K., S.J.N.)
| | - Mingyuan Shao
- From the Cleveland Clinic Coordinating Center for Clinical Research (C5R) (R.P., S.E.N., M.S.), and Department of Cardiovascular Medicine (R.P., S.E.N., M.B.E., S.R.K., E.M.T.), Cleveland Clinic, OH; Department of Medicine, University of Adelaide, Australia (R.P., S.J.N.); and South Australian Health and Medical Research Institute, Adelaide, Australia (Y.K., S.J.N.)
| | - Mohamed B. Elshazly
- From the Cleveland Clinic Coordinating Center for Clinical Research (C5R) (R.P., S.E.N., M.S.), and Department of Cardiovascular Medicine (R.P., S.E.N., M.B.E., S.R.K., E.M.T.), Cleveland Clinic, OH; Department of Medicine, University of Adelaide, Australia (R.P., S.J.N.); and South Australian Health and Medical Research Institute, Adelaide, Australia (Y.K., S.J.N.)
| | - Yu Kataoka
- From the Cleveland Clinic Coordinating Center for Clinical Research (C5R) (R.P., S.E.N., M.S.), and Department of Cardiovascular Medicine (R.P., S.E.N., M.B.E., S.R.K., E.M.T.), Cleveland Clinic, OH; Department of Medicine, University of Adelaide, Australia (R.P., S.J.N.); and South Australian Health and Medical Research Institute, Adelaide, Australia (Y.K., S.J.N.)
| | - Samir R. Kapadia
- From the Cleveland Clinic Coordinating Center for Clinical Research (C5R) (R.P., S.E.N., M.S.), and Department of Cardiovascular Medicine (R.P., S.E.N., M.B.E., S.R.K., E.M.T.), Cleveland Clinic, OH; Department of Medicine, University of Adelaide, Australia (R.P., S.J.N.); and South Australian Health and Medical Research Institute, Adelaide, Australia (Y.K., S.J.N.)
| | - E. Murat Tuzcu
- From the Cleveland Clinic Coordinating Center for Clinical Research (C5R) (R.P., S.E.N., M.S.), and Department of Cardiovascular Medicine (R.P., S.E.N., M.B.E., S.R.K., E.M.T.), Cleveland Clinic, OH; Department of Medicine, University of Adelaide, Australia (R.P., S.J.N.); and South Australian Health and Medical Research Institute, Adelaide, Australia (Y.K., S.J.N.)
| | - Stephen J. Nicholls
- From the Cleveland Clinic Coordinating Center for Clinical Research (C5R) (R.P., S.E.N., M.S.), and Department of Cardiovascular Medicine (R.P., S.E.N., M.B.E., S.R.K., E.M.T.), Cleveland Clinic, OH; Department of Medicine, University of Adelaide, Australia (R.P., S.J.N.); and South Australian Health and Medical Research Institute, Adelaide, Australia (Y.K., S.J.N.)
| |
Collapse
|
12
|
Watts GF, Pang J, Chan DC, Brunt JN, Lewis B. Angiographic progression of coronary atherosclerosis in patients with familial hypercholesterolaemia treated with non-statin therapy: Impact of a fat-modified diet and a resin. Atherosclerosis 2016; 252:82-87. [DOI: 10.1016/j.atherosclerosis.2016.07.923] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/29/2016] [Accepted: 07/27/2016] [Indexed: 11/30/2022]
|
13
|
Kazuma SM, Sultan D, Zhao Y, Detering L, You M, Luehmann HP, Abdalla DSP, Liu Y. Recent Advances of Radionuclide-Based Molecular Imaging of Atherosclerosis. Curr Pharm Des 2016; 21:5267-76. [PMID: 26369676 DOI: 10.2174/1381612821666150915104529] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is a systemic disease characterized by the development of multifocal plaque lesions within vessel walls and extending into the vascular lumen. The disease takes decades to develop symptomatic lesions, affording opportunities for accurate detection of plaque progression, analysis of risk factors responsible for clinical events, and planning personalized treatment. Of the available molecular imaging modalities, radionuclidebased imaging strategies have been favored due to their sensitivity, quantitative detection and pathways for translational research. This review summarizes recent advances of radiolabeled small molecules, peptides, antibodies and nanoparticles for atherosclerotic plaque imaging during disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, 63110, United States.
| |
Collapse
|
14
|
Ellis KL, Hooper AJ, Burnett JR, Watts GF. Progress in the care of common inherited atherogenic disorders of apolipoprotein B metabolism. Nat Rev Endocrinol 2016; 12:467-84. [PMID: 27199287 DOI: 10.1038/nrendo.2016.69] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Familial hypercholesterolaemia, familial combined hyperlipidaemia (FCH) and elevated lipoprotein(a) are common, inherited disorders of apolipoprotein B metabolism that markedly accelerate the onset of atherosclerotic cardiovascular disease (ASCVD). These disorders are frequently encountered in clinical lipidology and need to be accurately identified and treated in both index patients and their family members, to prevent the development of premature ASCVD. The optimal screening strategies depend on the patterns of heritability for each condition. Established therapies are widely used along with lifestyle interventions to regulate levels of circulating lipoproteins. New therapeutic strategies are becoming available, and could supplement traditional approaches in the most severe cases, but their long-term cost-effectiveness and safety have yet to be confirmed. We review contemporary developments in the understanding, detection and care of these highly atherogenic disorders of apolipoprotein B metabolism.
Collapse
Affiliation(s)
- Katrina L Ellis
- School of Medicine and Pharmacology, The University of Western Australia, PO Box X2213, Perth, Western Australia 6847, Australia
- Centre for Genetic Origins of Health and Disease, The University of Western Australia and Curtin University, 35 Stirling Highway, Crawley, Perth, Western Australia 6009, Australia
| | - Amanda J Hooper
- School of Medicine and Pharmacology, The University of Western Australia, PO Box X2213, Perth, Western Australia 6847, Australia
- PathWest Laboratory Medicine WA, Royal Perth Hospital and Fiona Stanley Hospital Network, Perth, Western Australia, Australia
- School of Pathology and Laboratory Medicine, The University of Western Australia, 35 Stirling Highway, Crawley, Perth, Western Australia 6009, Australia
| | - John R Burnett
- School of Medicine and Pharmacology, The University of Western Australia, PO Box X2213, Perth, Western Australia 6847, Australia
- PathWest Laboratory Medicine WA, Royal Perth Hospital and Fiona Stanley Hospital Network, Perth, Western Australia, Australia
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Wellington Street Perth, Western Australia, Australia
| | - Gerald F Watts
- School of Medicine and Pharmacology, The University of Western Australia, PO Box X2213, Perth, Western Australia 6847, Australia
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Wellington Street Perth, Western Australia, Australia
| |
Collapse
|
15
|
Najam O, Ray KK. Where to now in cardiovascular disease prevention. Atherosclerosis 2016; 251:483-489. [DOI: 10.1016/j.atherosclerosis.2016.06.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 05/16/2016] [Accepted: 06/17/2016] [Indexed: 01/24/2023]
|
16
|
|
17
|
Jung C, Dučić T, Reimer R, Koziolek E, Kording F, Heine M, Adam G, Ittrich H, Kaul MG. Gadospin F-enhanced magnetic resonance imaging for diagnosis and monitoring of atherosclerosis: validation with transmission electron microscopy and x-ray fluorescence imaging in the apolipoprotein e-deficient mouse. Mol Imaging 2015; 13. [PMID: 25342533 DOI: 10.2310/7290.2014.00039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The aim of this study was to investigate the feasibility of noninvasive monitoring of plaque burden in apolipoprotein E-deficient (ApoE-/-) mice by Gadospin F (GDF)-enhanced magnetic resonance imaging (MRI). Gadolinium uptake in plaques was controlled using transmission electron microscopy (TEM) and x-ray fluorescence (XRF) microscopy. To monitor the progression of atherosclerosis, ApoE-/- (n = 5) and wild-type (n = 2) mice were fed a Western diet and imaged at 5, 10, 15, and 20 weeks. Contrast-enhanced MRI was performed at 7 T Clinscan (Bruker, Ettlingen, Germany) before and 2 hours after intravenous injection of GDF (100 μmol/kg) to determine the blood clearance. Plaque size and contrast to noise ratio (CNR) were calculated for each time point using region of interest measurements to evaluate plaque progression. Following MRI, aortas were excised and GDF uptake was cross-validated by TEM and XRF microscopy. The best signal enhancement in aortic plaque was achieved 2 hours after application of GDF. No signal differences between pre- and postcontrast MRI were detectable in wild-type mice. We observed a gradual and considerable increase in plaque CNR and size for the different disease stages. TEM and XRF microscopy confirmed the localization of GDF within the plaque. GDF-enhanced MRI allows noninvasive and reliable estimation of plaque burden and monitoring of atherosclerotic progression in vivo.
Collapse
|
18
|
Abran M, Stähli BE, Merlet N, Mihalache-Avram T, Mecteau M, Rhéaume E, Busseuil D, Tardif JC, Lesage F. Validating a bimodal intravascular ultrasound (IVUS) and near-infrared fluorescence (NIRF) catheter for atherosclerotic plaque detection in rabbits. BIOMEDICAL OPTICS EXPRESS 2015; 6:3989-99. [PMID: 26504648 PMCID: PMC4605057 DOI: 10.1364/boe.6.003989] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 07/30/2015] [Accepted: 08/08/2015] [Indexed: 05/03/2023]
Abstract
Coronary artery disease is characterized by atherosclerotic plaque formation. Despite impressive advances in intravascular imaging modalities, in vivo molecular plaque characterization remains challenging, and different multimodality imaging systems have been proposed. We validated an engineered bimodal intravascular ultrasound imaging (IVUS) / near-infrared fluorescence (NIRF) imaging catheter in vivo using a balloon injury atherosclerosis rabbit model. Rabbit aortas and right iliac arteries were scanned in vivo after indocyanine green (ICG) injection, and compared to corresponding ex vivo fluorescence and white light images. Areas of ICG accumulation were colocalized with macroscopic atherosclerotic plaque formation. In vivo imaging was performed with the bimodal catheter integrating ICG-induced fluorescence signals into cross-sectional IVUS imaging. In vivo ICG accumulation corresponded to ex vivo fluorescence signal intensity and IVUS identified plaques.
Collapse
Affiliation(s)
- Maxime Abran
- Département de Génie Électrique and Institut de Génie Biomédical, École Polytechnique de Montréal, 2900 Édouard-Montpetit, Montreal, Qc, H3T 1J4, Canada
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montreal, Qc, H1T 1C8, Canada
| | - Barbara E. Stähli
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montreal, Qc, H1T 1C8, Canada
| | - Nolwenn Merlet
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montreal, Qc, H1T 1C8, Canada
| | | | - Mélanie Mecteau
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montreal, Qc, H1T 1C8, Canada
| | - Eric Rhéaume
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montreal, Qc, H1T 1C8, Canada
- Département de médecine, Université de Montréal, 2900 Édouard-Montpetit, Montreal, Qc, H3T 1J4, Canada
| | - David Busseuil
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montreal, Qc, H1T 1C8, Canada
| | - Jean-Claude Tardif
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montreal, Qc, H1T 1C8, Canada
- Département de médecine, Université de Montréal, 2900 Édouard-Montpetit, Montreal, Qc, H3T 1J4, Canada
| | - Frédéric Lesage
- Département de Génie Électrique and Institut de Génie Biomédical, École Polytechnique de Montréal, 2900 Édouard-Montpetit, Montreal, Qc, H3T 1J4, Canada
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montreal, Qc, H1T 1C8, Canada
| |
Collapse
|
19
|
Albert BB, Derraik JGB, Brennan CM, Biggs JB, Garg ML, Cameron-Smith D, Hofman PL, Cutfield WS. Supplementation with a blend of krill and salmon oil is associated with increased metabolic risk in overweight men. Am J Clin Nutr 2015; 102:49-57. [PMID: 26016867 DOI: 10.3945/ajcn.114.103028] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 04/30/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Krill is an increasingly popular source of marine n-3 (ω-3) PUFA that is seen as a premium product. However, to our knowledge, the effect of krill-oil supplementation on insulin sensitivity in humans has not been reported. OBJECTIVE We assessed whether supplementation with a blend of krill and salmon (KS) oil [which is rich in eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)] affects insulin sensitivity in overweight men. DESIGN The design was a randomized, double-blind, controlled crossover trial. A total of 47 men with a mean ± SD age of 46.5 ± 5.1 y, who were overweight [body mass index (in kg/m(2)) from 25 to 30] but otherwise healthy, received 5 1-g capsules of KS oil or a control (canola oil) for 8 wk and crossed over to another treatment after an 8-wk washout period. The primary outcome was insulin sensitivity assessed by using the Matsuda method from an oral-glucose-tolerance test. Secondary outcomes included lipid profiles, inflammatory markers, 24-h ambulatory blood pressure, and carotid artery intimamedia thickness. RESULTS Unexpectedly, insulin sensitivity (per the Matsuda index) was 14% lower with the KS oil than with the control oil (P = 0.049). A mediation analysis showed that, after controlling for the likely positive effects of blood EPA and DHA (i.e., the omega-3 index), the reduction in insulin sensitivity after KS-oil supplementation was more marked [27% lower than with the control oil (P = 0.009)]. CONCLUSIONS Supplementation with a blend of KS oil is associated with decreased insulin sensitivity. Thus, krill-oil supplementation in overweight adults could exacerbate risk of diabetes and cardiovascular disease. This trial was prospectively registered at the Australian New Zealand Clinical Trials Registry as ACTRN12611000602921.
Collapse
Affiliation(s)
- Benjamin B Albert
- Liggins Institute, University of Auckland, Auckland, New Zealand; and
| | - José G B Derraik
- Liggins Institute, University of Auckland, Auckland, New Zealand; and
| | | | - Janene B Biggs
- Liggins Institute, University of Auckland, Auckland, New Zealand; and
| | - Manohar L Garg
- Nutraceuticals Research Group, University of Newcastle, Callaghan, Australia
| | | | - Paul L Hofman
- Liggins Institute, University of Auckland, Auckland, New Zealand; and
| | - Wayne S Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand; and
| |
Collapse
|
20
|
Yip VLM, Hawcutt DB, Pirmohamed M. Pharmacogenetic Markers of Drug Efficacy and Toxicity. Clin Pharmacol Ther 2015; 98:61-70. [PMID: 25870137 DOI: 10.1002/cpt.135] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/08/2015] [Indexed: 12/23/2022]
Abstract
The action of a drug is dictated by its pharmacokinetic and pharmacodynamics properties, both of which can vary in different individuals because of environmental and genetic factors. Pharmacogenetics, the study of genetic factors determining drug response, has the potential to improve clinical outcomes through targeting therapies, individualizing dosing, preventing adverse drug reactions, and potentially rescuing previously failed therapies. Although there have been significant advances in pharmacogenetics over the last decade, only a few have been translated into clinical practice. However, with new rapid genotyping technologies, regulatory modernization, novel clinical trial designs, systems approaches, and integration of pharmacogenetic data into decision support systems, there is hope that pharmacogenetics, as an important component of the overall drive towards personalized medicine, will advance more quickly in the future. There will continue to be a need for collaboration between centers all over the world, and multisector working, capitalizing on the current data revolution.
Collapse
Affiliation(s)
- V L M Yip
- Departments of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK.,Royal Liverpool University Hospital, Liverpool, UK
| | - D B Hawcutt
- Women and Child Health Institute of Translational Medicine, University of Liverpool, Liverpool, UK.,Alder Hey Children's Hospital, Liverpool, UK
| | - M Pirmohamed
- Departments of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK.,Royal Liverpool University Hospital, Liverpool, UK
| |
Collapse
|
21
|
Thukkani AK, Jaffer FA. Molecular Imaging. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
22
|
Tardif JC, Rhéaume E, Lemieux Perreault LP, Grégoire JC, Feroz Zada Y, Asselin G, Provost S, Barhdadi A, Rhainds D, L'Allier PL, Ibrahim R, Upmanyu R, Niesor EJ, Benghozi R, Suchankova G, Laghrissi-Thode F, Guertin MC, Olsson AG, Mongrain I, Schwartz GG, Dubé MP. Pharmacogenomic determinants of the cardiovascular effects of dalcetrapib. ACTA ACUST UNITED AC 2015; 8:372-82. [PMID: 25583994 DOI: 10.1161/circgenetics.114.000663] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 12/16/2014] [Indexed: 11/16/2022]
Abstract
BACKGROUND Dalcetrapib did not improve clinical outcomes, despite increasing high-density lipoprotein cholesterol by 30%. These results differ from other evidence supporting high-density lipoprotein as a therapeutic target. Responses to dalcetrapib may vary according to patients' genetic profile. METHODS AND RESULTS We conducted a pharmacogenomic evaluation using a genome-wide approach in the dal-OUTCOMES study (discovery cohort, n=5749) and a targeted genotyping panel in the dal-PLAQUE-2 imaging trial (support cohort, n=386). The primary endpoint for the discovery cohort was a composite of cardiovascular events. The change from baseline in carotid intima-media thickness on ultrasonography at 6 and 12 months was evaluated as supporting evidence. A single-nucleotide polymorphism was found to be associated with cardiovascular events in the dalcetrapib arm, identifying the ADCY9 gene on chromosome 16 (rs1967309; P=2.41×10(-8)), with 8 polymorphisms providing P<10(-6) in this gene. Considering patients with genotype AA at rs1967309, there was a 39% reduction in the composite cardiovascular endpoint with dalcetrapib compared with placebo (hazard ratio, 0.61; 95% confidence interval, 0.41-0.92). In patients with genotype GG, there was a 27% increase in events with dalcetrapib versus placebo. Ten single-nucleotide polymorphism in the ADCY9 gene, the majority in linkage disequilibrium with rs1967309, were associated with the effect of dalcetrapib on intima-media thickness (P<0.05). Marker rs2238448 in ADCY9, in linkage disequilibrium with rs1967309 (r(2)=0.8), was associated with both the effects of dalcetrapib on intima-media thickness in dal-PLAQUE-2 (P=0.009) and events in dal-OUTCOMES (P=8.88×10(-8); hazard ratio, 0.67; 95% confidence interval, 0.58-0.78). CONCLUSIONS The effects of dalcetrapib on atherosclerotic outcomes are determined by correlated polymorphisms in the ADCY9 gene. CLINICAL TRIAL INFORMATION URL: http://www.clinicaltrials.gov. Unique identifiers: NCT00658515 and NCT01059682.
Collapse
Affiliation(s)
- Jean-Claude Tardif
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.).
| | - Eric Rhéaume
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Louis-Philippe Lemieux Perreault
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Jean C Grégoire
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Yassamin Feroz Zada
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Géraldine Asselin
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Sylvie Provost
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Amina Barhdadi
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - David Rhainds
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Philippe L L'Allier
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Reda Ibrahim
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Ruchi Upmanyu
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Eric J Niesor
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Renée Benghozi
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Gabriela Suchankova
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Fouzia Laghrissi-Thode
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Marie-Claude Guertin
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Anders G Olsson
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Ian Mongrain
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Gregory G Schwartz
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Marie-Pierre Dubé
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.).
| |
Collapse
|
23
|
Puri R, Nissen SE, Shao M, Uno K, Kataoka Y, Kapadia SR, Tuzcu EM, Nicholls SJ. Impact of baseline lipoprotein and C-reactive protein levels on coronary atheroma regression following high-intensity statin therapy. Am J Cardiol 2014; 114:1465-72. [PMID: 25282317 DOI: 10.1016/j.amjcard.2014.08.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/12/2014] [Accepted: 08/12/2014] [Indexed: 01/22/2023]
Abstract
Guidelines now recommend high-intensity statin therapy in all patients with proven atherosclerotic disease. Yet the impact of baseline lipoprotein and C-reactive protein (CRP) levels on measures of disease regression to this therapy are unknown. The aim of this study was to test the hypothesis that high-intensity statin therapy causes equivalent degrees of coronary atheroma regression irrespective of baseline lipoprotein and CRP levels. In 8 prospective randomized trials using serial coronary intravascular ultrasound, 1,881 patients who maintained or switched to 18- to 24 months of high-intensity statin therapy (rosuvastatin 40 mg or atorvastatin 80 mg) were stratified according to baseline lipoprotein and CRP levels. Changes in coronary percentage atheroma volume (PAV) and total atheroma volume (TAV) were evaluated. High-intensity statin therapy produced significant reductions from baseline in low-density lipoprotein cholesterol by 38.4%, non-high-density lipoprotein (HDL) cholesterol by 33.6%, triglycerides by 13.1%, and CRP by 33.3%, while increasing HDL cholesterol by 11.7% (p <0.001 for all). This was associated with regression of PAV by 0.7% and of TAV by 8.2 mm(3) (p <0.001 for both). No significant differences of changes in PAV and TAV were observed across baseline quintiles of low-density lipoprotein cholesterol, HDL cholesterol, non-HDL cholesterol, triglycerides, or CRP. Moreover, across all measured lipoproteins and CRP, most patients demonstrated plaque regression (defined as any change from baseline in PAV or TAV <0). In conclusion, high-intensity statin therapy attenuated the natural progression of coronary atherosclerosis in all strata of patients with coronary artery disease irrespective of baseline lipoprotein or CRP levels. These findings provide support for the latest United States guideline recommendations for the broad use of high-intensity statin therapy in all patients with atherosclerosis, regardless of baseline lipid status.
Collapse
Affiliation(s)
- Rishi Puri
- Cleveland Clinic Coordinating Center for Clinical Research, Cleveland, Ohio; Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Steven E Nissen
- Cleveland Clinic Coordinating Center for Clinical Research, Cleveland, Ohio; Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Mingyuan Shao
- Cleveland Clinic Coordinating Center for Clinical Research, Cleveland, Ohio
| | - Kiyoko Uno
- Cleveland Clinic Coordinating Center for Clinical Research, Cleveland, Ohio
| | - Yu Kataoka
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Samir R Kapadia
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - E Murat Tuzcu
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
24
|
Albert BB, Derraik JGB, Brennan CM, Biggs JB, Smith GC, Garg ML, Cameron-Smith D, Hofman PL, Cutfield WS. Higher omega-3 index is associated with increased insulin sensitivity and more favourable metabolic profile in middle-aged overweight men. Sci Rep 2014; 4:6697. [PMID: 25331725 PMCID: PMC5381193 DOI: 10.1038/srep06697] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 10/01/2014] [Indexed: 01/13/2023] Open
Abstract
We assessed whether omega-3 index (red blood cell concentrations of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)) was associated with insulin sensitivity and other metabolic outcomes in 47 overweight men aged 46.5 ± 5.1 years. Participants were assessed twice, 16 weeks apart. Insulin sensitivity was assessed by the Matsuda method from an oral glucose tolerance test. Linear associations were examined; stratified analyses were carried out with participants separated according to the omega-3 index: lower tertiles (LOI; n = 31) and highest tertile (HOI; n = 16). Increasing omega-3 index was correlated with higher insulin sensitivity (r = 0.23; p = 0.025), higher disposition index (r = 0.20; p = 0.054), and lower CRP concentrations (r = −0.39; p < 0.0001). Insulin sensitivity was 43% higher in HOI than in LOI men (Matsuda index 6.83 vs 4.78; p = 0.009). Similarly, HOI men had disposition index that was 70% higher (p = 0.013) and fasting insulin concentrations 25% lower (p = 0.038). HOI men displayed lower nocturnal systolic blood pressure (−6.0 mmHg; p = 0.025) and greater systolic blood pressure dip (14.7 vs 10.8%; p = 0.039). Men in the HOI group also had lower concentrations of CRP (41% lower; p = 0.033) and free fatty acids (21% lower, p = 0.024). In conclusion, higher omega-3 index is associated with increased insulin sensitivity and a more favourable metabolic profile in middle-aged overweight men.
Collapse
Affiliation(s)
| | - José G B Derraik
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Janene B Biggs
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Greg C Smith
- Department of Pharmacology, University of New South Wales, Sydney, New South Wales, Australia
| | - Manohar L Garg
- Nutraceuticals Research Group, University of Newcastle, Callaghan, New South Wales, Australia
| | | | - Paul L Hofman
- 1] Liggins Institute, University of Auckland, Auckland, New Zealand [2] Gravida: National Centre for Growth and Development, Auckland, New Zealand
| | - Wayne S Cutfield
- 1] Liggins Institute, University of Auckland, Auckland, New Zealand [2] Gravida: National Centre for Growth and Development, Auckland, New Zealand
| |
Collapse
|
25
|
Abran M, Cloutier G, Cardinal MHR, Chayer B, Tardif JC, Lesage F. Development of a photoacoustic, ultrasound and fluorescence imaging catheter for the study of atherosclerotic plaque. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2014; 8:696-703. [PMID: 25350946 DOI: 10.1109/tbcas.2014.2360560] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Atherosclerotic cardiovascular diseases are a major cause of death in industrialized countries. Molecular imaging modalities are increasingly recognized to be a promising avenue towards improved diagnosis and for the evaluation of new drug therapies. In this work, we present an acquisition system and associated catheter enabling simultaneous photoacoustic, ultrasound and fluorescence imaging of arteries designed for in vivo imaging. The catheter performance is evaluated in tissue-mimicking phantoms. Simultaneous imaging with three modalities is demonstrated at frame rates of 30 images per second for ultrasound and fluorescence and 1 image per 13 seconds for photoacoustic. Acquired radio-frequency ultrasound data could be processed to obtain radial strain elastograms. With motorized pullback, 3D imaging of phantoms was performed using the three modalities.
Collapse
|
26
|
Dohi T, Maehara A, Moreno PR, Baber U, Kovacic JC, Limaye AM, Ali ZA, Sweeny JM, Mehran R, Dangas GD, Xu K, Sharma SK, Mintz GS, Kini AS. The relationship among extent of lipid-rich plaque, lesion characteristics, and plaque progression/regression in patients with coronary artery disease: a serial near-infrared spectroscopy and intravascular ultrasound study. Eur Heart J Cardiovasc Imaging 2014; 16:81-7. [PMID: 25190072 DOI: 10.1093/ehjci/jeu169] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
AIMS To evaluate the relationship between lipid content and plaque morphometry as well as the process of lesion progression and regression in patients with significant coronary artery disease. METHODS AND RESULTS The present study, using data from the YELLOW trial, was conducted in patients having significant coronary lesions (fractional flow reserve <0.8) who underwent serial intravascular ultrasound (IVUS) and near-infrared spectroscopy (NIRS) at baseline and after 7 weeks. For each coronary plaque (≥50% plaque burden that was ≥5 mm in length), we evaluated plaque characteristics and the extent of lipid-rich plaque [LRP, defined as the 4 mm long segment with the maximum lipid-core burden index (maxLCBI4 mm)] on NIRS. Among 66 patients (age 63.0 ± 10.1 years; 82% statin use at baseline), 94 plaques were identified. The extent of LRP at baseline was positively correlated with IVUS plaque burden (r = 0.317, P = 0.002). A large LRP (maxLCBI4 mm ≥500) was present only in plaques with a large plaque burden (≥70%). Multivariate analysis demonstrated that plaque burden was the best predictor of the extent of LRP (P < 0.001). In lesions with a large plaque burden and a large amount of LRP at baseline, a reduction in LRP was seen in all lesions in patients receiving intensive statin therapy (P = 0.004) without a significant change in plaque burden. CONCLUSIONS Coronary lesions containing a large amount of LRP also had a large plaque burden. Short-term regression of LRP (without a change in plaque burden) was observed mainly in plaques with a large plaque burden and a large amount of LRP at baseline. CLINICAL TRIAL REGISTRATION http://www.clinicaltrials.gov. Unique identifier: NCT01567826.
Collapse
Affiliation(s)
- Tomotaka Dohi
- Columbia University Medical Center, New York, NY, USA Cardiovascular Research Foundation, 111 East 59th Street, 12th Floor, New York, NY 10022, USA
| | - Akiko Maehara
- Columbia University Medical Center, New York, NY, USA Cardiovascular Research Foundation, 111 East 59th Street, 12th Floor, New York, NY 10022, USA
| | - Pedro R Moreno
- The Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Usman Baber
- The Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jason C Kovacic
- The Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Atul M Limaye
- The Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ziad A Ali
- Columbia University Medical Center, New York, NY, USA The Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph M Sweeny
- The Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roxana Mehran
- Cardiovascular Research Foundation, 111 East 59th Street, 12th Floor, New York, NY 10022, USA The Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George D Dangas
- Cardiovascular Research Foundation, 111 East 59th Street, 12th Floor, New York, NY 10022, USA The Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ke Xu
- Cardiovascular Research Foundation, 111 East 59th Street, 12th Floor, New York, NY 10022, USA
| | - Samin K Sharma
- The Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gary S Mintz
- Cardiovascular Research Foundation, 111 East 59th Street, 12th Floor, New York, NY 10022, USA
| | - Annapoorna S Kini
- The Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
27
|
Keshavarz-Motamed Z, Saijo Y, Majdouline Y, Riou L, Ohayon J, Cloutier G. Coronary artery atherectomy reduces plaque shear strains: An endovascular elastography imaging study. Atherosclerosis 2014; 235:140-9. [DOI: 10.1016/j.atherosclerosis.2014.04.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 04/16/2014] [Accepted: 04/16/2014] [Indexed: 01/26/2023]
|
28
|
Li B, Maafi F, Berti R, Pouliot P, Rhéaume E, Tardif JC, Lesage F. Hybrid FMT-MRI applied to in vivo atherosclerosis imaging. BIOMEDICAL OPTICS EXPRESS 2014; 5:1664-76. [PMID: 24877023 PMCID: PMC4026902 DOI: 10.1364/boe.5.001664] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 04/24/2014] [Accepted: 04/24/2014] [Indexed: 05/20/2023]
Abstract
Combining Fluorescent Molecular Tomography (FMT) with anatomical imaging, e.g. MRI facilitates interpreting functional information. Furthermore, using a heterogeneous model for light propagation has been shown in simulations to be superior to homogeneous modeling to quantify fluorescence. Here, we present a combined FMT-MRI system and apply it to heart and aorta molecular imaging, a challenging area due to strong tissue heterogeneity and the presence of air-voids due to lungs. First investigating performance in a phantom and mouse corpse, the MRI-enabled heterogeneous models resulted in an improved quantification of fluorescence reconstructions. The system was then used in mice for in vivo atherosclerosis molecular imaging. Results show that, when using the heterogeneous model, reconstructions were in agreement with the ex vivo measurements. Therefore, the proposed system might serve as a powerful imaging tool for atherosclerosis in mice.
Collapse
Affiliation(s)
- Baoqiang Li
- Institute of Biomedical Engineering, École Polytechnique de Montréal, Montreal, QC, H3C 3A7, Canada
- Montreal Heart Institute, Montreal, QC, H1T 1C8, Canada
| | - Foued Maafi
- Montreal Heart Institute, Montreal, QC, H1T 1C8, Canada
| | - Romain Berti
- Montreal Heart Institute, Montreal, QC, H1T 1C8, Canada
| | - Philippe Pouliot
- Institute of Biomedical Engineering, École Polytechnique de Montréal, Montreal, QC, H3C 3A7, Canada
- Montreal Heart Institute, Montreal, QC, H1T 1C8, Canada
| | - Eric Rhéaume
- Montreal Heart Institute, Montreal, QC, H1T 1C8, Canada
| | | | - Frederic Lesage
- Institute of Biomedical Engineering, École Polytechnique de Montréal, Montreal, QC, H3C 3A7, Canada
- Montreal Heart Institute, Montreal, QC, H1T 1C8, Canada
| |
Collapse
|
29
|
Dhindsa S, Jialal I. Potential anti-atherosclerotic effects of dipeptidyl peptidase-4 inhibitors in type 2 diabetes mellitus. Curr Diab Rep 2014; 14:463. [PMID: 24390468 DOI: 10.1007/s11892-013-0463-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality in patients with diabetes. Pharmacotherapy that can reduce hyperglycemia and also exhibit pleiotropic effects that can result in a reduction in cardiovascular disease will be a major advance. Recently, the dipeptidyl-peptidase-4 inhibitors were introduced as ant-hyperglycemic therapy. Studies from numerous groups have reported effects that could potentially result in a reduction in CVD. Some of the drugs in this class, especially vildagliptin and sitagliptin, have been shown to reduce postprandial hyperlipidemia following a fat load, improve endothelial function as evidenced by increased forearm blood flow, and also display anti-inflammatory effects. Their effects on platelet function, blood pressure, and oxidative stress are very preliminary and need to be confirmed. Finally, they have been shown to reduce subclinical atherosclerosis by reducing carotid intimal-medial thickness. However, the final arbiter with respect to a reduction in CVD will be the ongoing clinical trials.
Collapse
Affiliation(s)
- Sandeep Dhindsa
- Division of Endocrinology and Metabolism, Texas Tech University Health Sciences Center, Permian Basin Campus, Odessa, TX, 79763, USA
| | | |
Collapse
|
30
|
Mani V, Woodward M, Samber D, Bucerius J, Tawakol A, Kallend D, Rudd JHF, Abt M, Fayad ZA. Predictors of change in carotid atherosclerotic plaque inflammation and burden as measured by 18-FDG-PET and MRI, respectively, in the dal-PLAQUE study. Int J Cardiovasc Imaging 2014; 30:571-82. [PMID: 24458953 DOI: 10.1007/s10554-014-0370-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 01/15/2014] [Indexed: 12/17/2022]
Abstract
Baseline predictors of response to treatment of patients with coronary heart disease (CHD) with respect to vascular inflammation and atherosclerotic plaque burden are poorly understood. From post hoc analysis of the dal-PLAQUE study (NCT00655473), 18F-fluorodeoxyglucose-positron emission tomography (18-FDG-PET) imaging and carotid black blood magnetic resonance imaging (MRI) were used to track changes in these vascular parameters. Baseline demographics, imaging, and biomarkers were collected/measured in 130 patients with CHD or CHD risk-equivalents, and imaging follow-up at 6 months (PET) and 24 months (MRI) was performed. Using stepwise linear regression, predictors of change in carotid plaque inflammation by PET [target-to-background ratio (TBR), n = 92] and plaque burden by MRI [wall area (WA) and total vessel area (TVA), n = 89] were determined. Variables with p < 0.05 in multivariable models were considered independently significant. Interleukin-6, systolic blood pressure and standard deviation of wall thickness (WT) at baseline were independently positively associated with 18-FDG uptake (mean of maximum [MeanMax] TBR change over 6 months). Mean of mean TBR, phospholipase A2, apolipoprotein A-I, and high-sensitivity C-reactive protein at baseline were independently negatively associated with MeanMax TBR change over 6 months. Mean WT and plasminogen activator inhibitor-1 (PAI-1) activity at baseline, and age, were independently associated with change in WA over 24 months. For TVA changes; mean WA and PAI-1 activity at baseline, age, and female gender were independent predictors. These findings may help determine patients most suitable for clinical trials employing plaque inflammation or burden changes as endpoints.
Collapse
Affiliation(s)
- Venkatesh Mani
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Heinonen TM, Aamer M, Marshall C, Black DM, Tardif JC. Cardiovascular biomarkers and surrogate end points: key initiatives and clinical trial challenges. Expert Rev Cardiovasc Ther 2014; 10:989-94. [DOI: 10.1586/erc.12.84] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
32
|
Gebhard C, L'Allier PL, Tardif JC. Near-infrared spectroscopy for cardiovascular risk assessment? Not ready for prime time. Eur Heart J 2013; 35:263-5. [PMID: 24052602 DOI: 10.1093/eurheartj/eht361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
33
|
Puri R, Nissen SE, Shao M, Ballantyne CM, Barter PJ, Chapman MJ, Erbel R, Libby P, Raichlen JS, Uno K, Kataoka Y, Nicholls SJ. Coronary atheroma volume and cardiovascular events during maximally intensive statin therapy. Eur Heart J 2013; 34:3182-90. [DOI: 10.1093/eurheartj/eht260] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
34
|
Mahdy Ali K, Wonnerth A, Huber K, Wojta J. Cardiovascular disease risk reduction by raising HDL cholesterol--current therapies and future opportunities. Br J Pharmacol 2013; 167:1177-94. [PMID: 22725625 DOI: 10.1111/j.1476-5381.2012.02081.x] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Since the first discovery of an inverse correlation between high-density lipoprotein-cholesterol (HDL-C) levels and coronary heart disease in the 1950s the life cycle of HDL, its role in atherosclerosis and the therapeutic modification of HDL-C levels have been major research topics. The Framingham study and others that followed could show that HDL-C is an independent cardiovascular risk factor and that the increase of HDL-C of only 10 mg·L(-1) leads to a risk reduction of 2-3%. While statin therapy and therefore low-density lipoprotein-cholesterol (LDL-C) reduction could lower coronary heart disease considerably; cardiovascular morbidity and mortality still occur in a significant portion of subjects already receiving therapy. Therefore, new strategies and therapies are needed to further reduce the risk. Raising HDL-C was thought to achieve this goal. However, established drug therapies resulting in substantial HDL-C increase are scarce and their effect is controversial. Furthermore, it is becoming increasingly evident that HDL particle functionality is at least as important as HDL-C levels since HDL particles not only promote reverse cholesterol transport from the periphery (mainly macrophages) to the liver but also exert pleiotropic effects on inflammation, haemostasis and apoptosis. This review deals with the biology of HDL particles, the established and future therapeutic options to increase HDL-C and discusses the results and conclusions of the most important studies published in the last years. Finally, an outlook on future diagnostic tools and therapeutic opportunities regarding coronary artery disease is given.
Collapse
Affiliation(s)
- K Mahdy Ali
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
35
|
de Bock M, Derraik JGB, Brennan CM, Biggs JB, Morgan PE, Hodgkinson SC, Hofman PL, Cutfield WS. Olive (Olea europaea L.) leaf polyphenols improve insulin sensitivity in middle-aged overweight men: a randomized, placebo-controlled, crossover trial. PLoS One 2013; 8:e57622. [PMID: 23516412 PMCID: PMC3596374 DOI: 10.1371/journal.pone.0057622] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Accepted: 01/24/2013] [Indexed: 02/05/2023] Open
Abstract
Background Olive plant leaves (Olea europaea L.) have been used for centuries in folk medicine to treat diabetes, but there are very limited data examining the effects of olive polyphenols on glucose homeostasis in humans. Objective To assess the effects of supplementation with olive leaf polyphenols (51.1 mg oleuropein, 9.7 mg hydroxytyrosol per day) on insulin action and cardiovascular risk factors in middle-aged overweight men. Design Randomized, double-blinded, placebo-controlled, crossover trial in New Zealand. 46 participants (aged 46.4±5.5 years and BMI 28.0±2.0 kg/m2) were randomized to receive capsules with olive leaf extract (OLE) or placebo for 12 weeks, crossing over to other treatment after a 6-week washout. Primary outcome was insulin sensitivity (Matsuda method). Secondary outcomes included glucose and insulin profiles, cytokines, lipid profile, body composition, 24-hour ambulatory blood pressure, and carotid intima-media thickness. Results Treatment evaluations were based on the intention-to-treat principle. All participants took >96% of prescribed capsules. OLE supplementation was associated with a 15% improvement in insulin sensitivity (p = 0.024) compared to placebo. There was also a 28% improvement in pancreatic β-cell responsiveness (p = 0.013). OLE supplementation also led to increased fasting interleukin-6 (p = 0.014), IGFBP-1 (p = 0.024), and IGFBP-2 (p = 0.015) concentrations. There were however, no effects on interleukin-8, TNF-α, ultra-sensitive CRP, lipid profile, ambulatory blood pressure, body composition, carotid intima-media thickness, or liver function. Conclusions Supplementation with olive leaf polyphenols for 12 weeks significantly improved insulin sensitivity and pancreatic β-cell secretory capacity in overweight middle-aged men at risk of developing the metabolic syndrome. Trial Registration Australian New Zealand Clinical Trials Registry #336317.
Collapse
Affiliation(s)
- Martin de Bock
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | | | - Janene B. Biggs
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Philip E. Morgan
- Heart Research Institute, University of Sydney, Sydney, Australia
| | | | - Paul L. Hofman
- Liggins Institute, University of Auckland, Auckland, New Zealand
- Gravida: National Centre for Growth and Development, Auckland, New Zealand
| | - Wayne S. Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand
- Gravida: National Centre for Growth and Development, Auckland, New Zealand
- * E-mail:
| |
Collapse
|
36
|
Jialal I, Bajaj M. DPP-4 inhibitors and atherosclerosis: the promise. Atherosclerosis 2013; 227:224-5. [PMID: 23395524 DOI: 10.1016/j.atherosclerosis.2012.12.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 12/19/2012] [Indexed: 12/26/2022]
Affiliation(s)
- Ishwarlal Jialal
- Laboratory for Atherosclerosis and Metabolic Research, University of California Davis Medical Center, Sacramento, CA, USA.
| | | |
Collapse
|
37
|
Singh M, Bedi US. Is Atherosclerosis Regression a Realistic Goal of Statin Therapy and What Does That Mean? Curr Atheroscler Rep 2012; 15:294. [DOI: 10.1007/s11883-012-0294-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
38
|
Cocker MS, Mc Ardle B, Spence JD, Lum C, Hammond RR, Ongaro DC, McDonald MA, deKemp RA, Tardif JC, Beanlands RSB. Imaging atherosclerosis with hybrid [18F]fluorodeoxyglucose positron emission tomography/computed tomography imaging: what Leonardo da Vinci could not see. J Nucl Cardiol 2012; 19:1211-25. [PMID: 23073913 PMCID: PMC3510422 DOI: 10.1007/s12350-012-9631-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Prodigious efforts and landmark discoveries have led toward significant advances in our understanding of atherosclerosis. Despite significant efforts, atherosclerosis continues globally to be a leading cause of mortality and reduced quality of life. With surges in the prevalence of obesity and diabetes, atherosclerosis is expected to have an even more pronounced impact upon the global burden of disease. It is imperative to develop strategies for the early detection of disease. Positron emission tomography (PET) imaging utilizing [(18)F]fluorodeoxyglucose (FDG) may provide a non-invasive means of characterizing inflammatory activity within atherosclerotic plaque, thus serving as a surrogate biomarker for detecting vulnerable plaque. The aim of this review is to explore the rationale for performing FDG imaging, provide an overview into the mechanism of action, and summarize findings from the early application of FDG PET imaging in the clinical setting to evaluate vascular disease. Alternative imaging biomarkers and approaches are briefly discussed.
Collapse
Affiliation(s)
- Myra S. Cocker
- Molecular Function and Imaging Program, Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7 Canada
| | - Brian Mc Ardle
- Molecular Function and Imaging Program, Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7 Canada
| | - J. David Spence
- Stroke Prevention & Atherosclerosis Research Centre, Robarts Research Institute, University of Western Ontario, 1400 Western Road, London, ON Canada
| | - Cheemun Lum
- Interventional & Diagnostic Neuroradiology, Department of Radiology, The Ottawa
Hospital, University of Ottawa, Civic Campus, Diagnostic Imaging, K1Y 4E9 Ottawa, ON Canada
| | - Robert R. Hammond
- Departments of Pathology and Clinical Neurological Sciences, London Health Sciences Centre and University of Western Ontario, 339 Windermere Road, N6A 5A5 London, ON Canada
| | - Deidre C. Ongaro
- Molecular Function and Imaging Program, Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7 Canada
| | - Matthew A. McDonald
- Molecular Function and Imaging Program, Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7 Canada
| | - Robert A. deKemp
- Molecular Function and Imaging Program, Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7 Canada
| | | | - Rob S. B. Beanlands
- Molecular Function and Imaging Program, Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7 Canada
| |
Collapse
|
39
|
Abstract
Atherosclerotic cardiovascular disease (CVD) is a complex disorder that leads to premature death and hospitalization. Several drugs have been, or are currently being tested for their ability to reduce cardiovascular mortality and/or promote regression of atherosclerotic lesions. In addition to "hard end point" clinical trials in which total and cardiovascular mortality as well as risk of incident myocardial infarction are considered as outcomes, trials with surrogate end points using imaging biomarkers can rapidly assess the efficacy of new cardiovascular drugs. Low-density lipoprotein-based therapies with statins have been shown to promote atherosclerosis regression, and several other drugs targeting high-density lipoproteins or inflammation/oxidation are currently being tested in both outcomes and imaging trials in which atherosclerosis regression is anticipated. In this review, we focus on the latest progress in CVD and highlight novel drugs that tackle atherosclerosis as well as the currently used and upcoming imaging techniques to optimally measure atherosclerosis progression.
Collapse
Affiliation(s)
- Benoit J Arsenault
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Québec, Canada
| | | | | |
Collapse
|
40
|
Rhainds D, Arsenault BJ, Brodeur MR, Tardif JC. An update on the clinical development of dalcetrapib (RO4607381), a cholesteryl ester transfer protein modulator that increases HDL cholesterol levels. Future Cardiol 2012; 8:513-31. [DOI: 10.2217/fca.12.25] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
CETP is the target of CETP inhibitors such as anacetrapib and the modulator dalcetrapib. Both molecules have entered Phase III clinical trials, with the ultimate goal of reducing cardiovascular events by raising HDL cholesterol. At the 600-mg dose selected for the dal-OUTCOMES study, dalcetrapib is expected to inhibit CETP activity by approximately 30% and raise HDL-C by approximately 30% with limited effects on LDL cholesterol. Importantly, dalcetrapib does not raise blood pressure or aldosterone levels, two effects previously associated with the CETP inhibitor torcetrapib. Dalcetrapib has been well tolerated at the 600-mg dose. In the dal-PLAQUE atherosclerosis imaging study, dalcetrapib reduced the enlargement of total vessel area over time. In May 2012, following the results of the second interim analysis of dal-OUTCOMES, the Data and Safety Monitoring Board recommended stopping the study owing to a lack of clinically significant benefit, which was followed by Roche’s (Basel, Switzerland) decision to terminate the study and the dalcetrapib program (dal-HEART). Contrary to anacetrapib, a potent CETP inhibitor that markedly increases HDL cholesterol and significantly reduces LDL cholesterol, dalcetrapib has allowed us to test the hypothesis that an isolated, moderate elevation in HDL cholesterol prevents cardiovascular events.
Collapse
Affiliation(s)
- David Rhainds
- Atherosclerosis Research Group, Montreal Heart Institute, 5000 Belanger St., Montreal, Quebec, H1T 1C8, Canada
| | - Benoit J Arsenault
- Atherosclerosis Research Group, Montreal Heart Institute, 5000 Belanger St., Montreal, Quebec, H1T 1C8, Canada
| | - Mathieu R Brodeur
- Atherosclerosis Research Group, Montreal Heart Institute, 5000 Belanger St., Montreal, Quebec, H1T 1C8, Canada
| | - Jean-Claude Tardif
- Atherosclerosis Research Group, Montreal Heart Institute, 5000 Belanger St., Montreal, Quebec, H1T 1C8, Canada
- Faculty of Medicine, Université de Montréal, 2900, Boulevard Édouard-Montpetit Montréal, Québec H3T 1J4, Canada
| |
Collapse
|
41
|
A novel model of atherosclerosis in rabbits using injury to arterial walls induced by ferric chloride as evaluated by optical coherence tomography as well as intravascular ultrasound and histology. J Biomed Biotechnol 2012; 2012:121867. [PMID: 22665979 PMCID: PMC3361737 DOI: 10.1155/2012/121867] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 03/06/2012] [Accepted: 03/10/2012] [Indexed: 11/17/2022] Open
Abstract
This study aim was to develop a new model of atherosclerosis by FeCl3-induced injury to right common carotid arteries (CCAs) of rabbits. Right CCAs were induced in male New Zealand White rabbits (n = 15) by combination of a cholesterol-rich diet and FeCl3-induced injury to arterial walls. The right and left CCAs were evaluated by histology and in vivo intravascular ultrasound (IVUS) and optical coherence tomography (OCT) examinations of 24 hours (n = 3), 8 weeks (n = 6), and 12 weeks (n = 6) after injury. Each right CCA of the rabbits showed extensive white-yellow plaques. At eight and 12 weeks after injury, IVUS, OCT, and histological findings demonstrated that the right CCAs had evident eccentric plaques. Six plaques (50%) with evident positive remodeling were observed. Marked progression was clearly observed in the same plaque at 12 weeks after injury when it underwent repeat OCT and IVUS. We demonstrated, for the first time, a novel model of atherosclerosis induced by FeCl3. The model is simple, fast, inexpensive, and reproducible and has a high success rate. The eccentric plaques and remodeling of plaques were common in this model. We successfully carried out IVUS and OCT examinations twice in the same lesion within a relatively long period of time.
Collapse
|
42
|
Campbell KA, Lipinski MJ, Doran AC, Skaflen MD, Fuster V, McNamara CA. Lymphocytes and the adventitial immune response in atherosclerosis. Circ Res 2012; 110:889-900. [PMID: 22427326 DOI: 10.1161/circresaha.111.263186] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Although much of the research on atherosclerosis has focused on the intimal accumulation of lipids and inflammatory cells, there is an increasing amount of interest in the role of the adventitia in coordinating the immune response in atherosclerosis. In this review of the contributions of the adventitia and adventitial lymphocytes to the development of atherosclerosis, we discuss recent research on the formation and structural nature of adventitial immune aggregates, potential mechanisms of crosstalk between the intima, media, and adventitia, specific contributions of B lymphocytes and T lymphocytes, and the role of the vasa vasorum and surrounding perivascular adipose tissue. Furthermore, we highlight techniques for the imaging of lymphocytes in the vasculature.
Collapse
Affiliation(s)
- Kirsti A Campbell
- Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | | | | | | | | | | |
Collapse
|
43
|
Pancholi K. A review of imaging methods for measuring drug release at nanometre scale: a case for drug delivery systems. Expert Opin Drug Deliv 2012; 9:203-18. [DOI: 10.1517/17425247.2011.648374] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
44
|
Li B, Abran M, Matteau-Pelletier C, Rouleau L, Lam T, Sharma R, Rhéaume E, Kakkar A, Tardif JC, Lesage F. Low-cost three-dimensional imaging system combining fluorescence and ultrasound. JOURNAL OF BIOMEDICAL OPTICS 2011; 16:126010. [PMID: 22191927 DOI: 10.1117/1.3662455] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
In this paper, we present a dual-modality imaging system combining three-dimensional (3D) continuous-wave transillumination fluorescence tomography with 3D ultrasound (US) imaging. We validated the system with two phantoms, one containing fluorescent inclusions (Cy5.5) at different depths, and another varying-thickness semicylindrical phantom. Using raster scanning, the combined fluorescence/US system was used to collect the boundary fluorescent emission in the X-Y plane, as well as recovered the 3D surface and position of the inclusions from US signals. US images were segmented to provide soft priors for the fluorescence image reconstruction. Phantom results demonstrated that with priors derived from the US images, the fluorescent reconstruction quality was significantly improved. As further evaluation, we show pilot in vivo results using an Apo-E mouse to assess the feasibility and performance of this system in animal studies. Limitations and potential to be used in artherosclerosis studies are then discussed.
Collapse
Affiliation(s)
- Baoqiang Li
- École Polytechnique de Montréal, Institute of Biomedical Engineering, Montreal, H3C 3A7, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Fayad ZA, Mani V, Woodward M, Kallend D, Abt M, Burgess T, Fuster V, Ballantyne CM, Stein EA, Tardif JC, Rudd JHF, Farkouh ME, Tawakol A. Safety and efficacy of dalcetrapib on atherosclerotic disease using novel non-invasive multimodality imaging (dal-PLAQUE): a randomised clinical trial. Lancet 2011; 378:1547-59. [PMID: 21908036 PMCID: PMC4151875 DOI: 10.1016/s0140-6736(11)61383-4] [Citation(s) in RCA: 415] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Dalcetrapib modulates cholesteryl ester transfer protein (CETP) activity to raise high-density lipoprotein cholesterol (HDL-C). After the failure of torcetrapib it was unknown if HDL produced by interaction with CETP had pro-atherogenic or pro-inflammatory properties. dal-PLAQUE is the first multicentre study using novel non-invasive multimodality imaging to assess structural and inflammatory indices of atherosclerosis as primary endpoints. METHODS In this phase 2b, double-blind, multicentre trial, patients (aged 18-75 years) with, or with high risk of, coronary heart disease were randomly assigned (1:1) to dalcetrapib 600 mg/day or placebo for 24 months. Randomisation was done with a computer-generated randomisation code and was stratified by centre. Patients and investigators were masked to treatment. Coprimary endpoints were MRI-assessed indices (total vessel area, wall area, wall thickness, and normalised wall index [average carotid]) after 24 months and (18)F-fluorodeoxyglucose ((18)F-FDG) PET/CT assessment of arterial inflammation within an index vessel (right carotid, left carotid, or ascending thoracic aorta) after 6 months, with no-harm boundaries established before unblinding of the trial. Analysis was by intention to treat. This trial is registered at ClinicalTrials.gov, NCT00655473. FINDINGS 189 patients were screened and 130 randomly assigned to placebo (66 patients) or dalcetrapib (64 patients). For the coprimary MRI and PET/CT endpoints, CIs were below the no-harm boundary or the adverse change was numerically lower in the dalcetrapib group than in the placebo group. MRI-derived change in total vessel area was reduced in patients given dalcetrapib compared with those given placebo after 24 months; absolute change from baseline relative to placebo was -4·01 mm(2) (90% CI -7·23 to -0·80; nominal p=0·04). The PET/CT measure of index vessel most-diseased-segment target-to-background ratio (TBR) was not different between groups, but carotid artery analysis showed a 7% reduction in most-diseased-segment TBR in the dalcetrapib group compared with the placebo group (-7·3 [90% CI -13·5 to -0·8]; nominal p=0·07). Dalcetrapib did not increase office blood pressure and the frequency of adverse events was similar between groups. INTERPRETATION Dalcetrapib showed no evidence of a pathological effect related to the arterial wall over 24 months. Moreover, this trial suggests possible beneficial vascular effects of dalcetrapib, including the reduction in total vessel enlargement over 24 months, but long-term safety and clinical outcomes efficacy of dalcetrapib need to be analysed. FUNDING F Hoffmann-La Roche Ltd.
Collapse
Affiliation(s)
- Zahi A Fayad
- Translational and Molecular Imaging Institute and Department of Radiology, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|