1
|
Kamrul-Hasan A, Alam MS, Mustari M, Hannan MA, Chowdhury EUR, Chowdhury SR, Gaffar MAJ, Singha SK, Mohana CA, Mondal E, Rahman MS, Rahman MM, Sarker S, Hoque MA, Islam MR, Robel MAB, Ahmad S, Raunak AIB, Nur-A-Musabber, Kaisar MM, Selim S. Cardiovascular risk in newly diagnosed patients with type 2 diabetes mellitus: a nationwide, facility-based, cross-sectional study in Bangladesh. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2025; 25:200399. [PMID: 40248520 PMCID: PMC12005924 DOI: 10.1016/j.ijcrp.2025.200399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/16/2025] [Accepted: 04/02/2025] [Indexed: 04/19/2025]
Abstract
Aims Evidence on cardiovascular (CV) risk stratification in Bangladeshi patients with type 2 diabetes mellitus (T2DM) who are asymptomatic for cardiovascular disease (CVD) is limited. This study aimed to assess the 10-year CV risk in newly diagnosed patients with T2DM. Methods In 2023, a cross-sectional study was carried out at endocrinology clinics in tertiary hospitals throughout Bangladesh, involving newly diagnosed patients with T2DM aged 25 to 84 who had no prior history of CVD and were asymptomatic for the condition. CV risk was assessed and classified using QRISK3. Results 1617 newly diagnosed patients with T2DM (age 44.92 ± 11.84 years, male 49.5 %) were analyzed. Their median QRISK3 score was 11.0 %, with 46.5 % at low, 25.7 % at moderate, and 27.8 % at high 10-year CV risk, respectively. The QRISK3 score increased with age for both men and women, with men consistently scoring higher than women in every age group. Among the age groups 25-39, 40-64, and 65-84, the percentages of patients with high 10-year CV risk were 3.3 %, 34.0 %, and 94.5 %, respectively. The median relative risk (RR) of CVD was 4.3. RR decreased with age for both sexes, and men had a lower RR than women across all age groups. A sleep duration of 6-9 h was associated with a lower 10-year CV risk. Conclusions Many newly diagnosed Bangladeshi patients with T2DM have substantial CV risk. QRISK3 can assist clinicians in predicting 10-year CV risk and choosing appropriate treatments to prevent CVD.
Collapse
Affiliation(s)
- A.B.M. Kamrul-Hasan
- Department of Endocrinology, Mymensingh Medical College, Mymensingh, Bangladesh
| | - Muhammad Shah Alam
- Department of Medicine, Army Medical College Cumilla, Cumilla, Bangladesh
| | - Marufa Mustari
- Department of Endocrinology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | | | | | - Sumon Rahman Chowdhury
- Department of Diabetes, Endocrinology and Metabolism, Chittagong Diabetic General Hospital, Chattogram, Bangladesh
| | | | | | - Choman Abdullah Mohana
- Department of Endocrinology, Mymensingh Medical College Hospital, Mymensingh, Bangladesh
| | - Ershad Mondal
- Department of Endocrinology, Mymensingh Medical College Hospital, Mymensingh, Bangladesh
| | - Md. Shahinur Rahman
- Department of Diabetes and Endocrinology, Diabetic Association Hospital, Pabna, Bangladesh
| | | | - Sourav Sarker
- Department of Medicine, Boalkhali Upazila Health Complex, Chattogram, Bangladesh
| | - Md. Azizul Hoque
- Department of Endocrinology, Shaheed Tajuddin Ahmad Medical College, Gazipur, Bangladesh
| | | | - Md. Abdul Bari Robel
- Department of Endocrinology, Cumilla Medical College Hospital, Cumilla, Bangladesh
| | - Shahryar Ahmad
- Department of Endocrinology, Comilla Medical College, Cumilla, Bangladesh
| | - Ahmed Ifrad Bin Raunak
- Department of Endocrinology, Shaheed Suhrawardy Medical College and Hospital, Dhaka, Bangladesh
| | - Nur-A-Musabber
- Department of Endocrinology, Mymensingh Medical College Hospital, Mymensingh, Bangladesh
| | - Md. Mostofa Kaisar
- Department of Endocrinology, Sheikh Fazilatunnessa Mujib Memorial KPJ Specialized Hospital, Kasimpur, Gazipur, Bangladesh
| | - Shahjada Selim
- Department of Endocrinology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| |
Collapse
|
2
|
Mohammadnia N, Wesselink BE, Bax WA, Nidorf SM, Mosterd A, Fiolet ATL, Cetinyurek-Yavuz A, Thompson PL, Bangdiwala SI, Eikelboom JW, Cornel JH, El Messaoudi S. Cardiovascular Benefit of Colchicine in Relation to Baseline Risk: A Secondary Analysis of the LoDoCo2 Trial. J Am Heart Assoc 2025; 14:e038687. [PMID: 40371626 DOI: 10.1161/jaha.124.038687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/04/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND The LoDoCo2 (Low-Dose Colchicine 2) trial showed that colchicine reduced the risk for cardiovascular events in patients with chronic coronary syndrome. Current guidelines recommend colchicine use in selected high-risk patients. The aim of this secondary analysis was to explore the relative and absolute benefits of colchicine according to baseline risk. METHODS The LoDoCo2 trial randomized 5522 patients to colchicine 0.5 mg or placebo. The primary end point was a composite of cardiovascular death, spontaneous myocardial infarction, ischemic stroke, or ischemia-driven coronary revascularization. First, a LoDoCo2 risk score was developed by Cox regression to identify high-risk features for the primary end point. Second, the Thrombolysis in Myocardial Infarction Risk Score for Secondary Prevention was applied to explore robustness of findings. RESULTS In the LoDoCo2 risk score, high-risk features were age ≥75, diabetes, and current smoker. In high-risk (≥1 high-risk feature), compared with low-risk (0 high-risk features) patients, colchicine was associated with consistent relative (high risk: hazard ratio [HR], 0.72 [95% CI, 0.56-0.94] versus low risk: HR, 0.67 [95% CI, 0.52-0.88]; P for interaction=0.73) and absolute benefits (high risk: HR, -1.33 [95% CI, -2.38 to -0.27] versus low risk: HR, -0.93 [95% CI -1.57 to -0.30] events per 100 person-years). Using the Thrombolysis in Myocardial Infarction Risk Score for Secondary Prevention, consistent relative and absolute benefits were found in high-, intermediate-, and low-risk patients. CONCLUSIONS In patients with chronic coronary syndrome, the relative and absolute benefits of colchicine were consistent in those at high, intermediate, and low risk for cardiovascular events. These findings support the use of colchicine across the spectrum of baseline risk. REGISTRATION URL: https://www.anzctr.org.au; Unique identifier: 12614000093684.
Collapse
Affiliation(s)
| | - Britta E Wesselink
- Department of Cardiology Radboud University Medical Center Nijmegen The Netherlands
| | - Willem A Bax
- Department of Internal Medicine Northwest Clinics Alkmaar Netherlands
| | - Stefan M Nidorf
- Heart and Vascular Research Institute of Western Australia Perth WA Australia
| | - Arend Mosterd
- Dutch Network for Cardiovascular Research (WCN) Utrecht The Netherlands
- Department of Cardiology Meander Medical Center Amersfoort The Netherlands
| | - Aernoud T L Fiolet
- Dutch Network for Cardiovascular Research (WCN) Utrecht The Netherlands
- Department of Cardiology University Medical Center Utrecht Utrecht The Netherlands
| | | | - Peter L Thompson
- Heart and Vascular Research Institute of Western Australia Perth WA Australia
- School of Medicine University of Western Australia Perth WA Australia
- Sir Charles Gairdner Hospital Perth WA Australia
| | - Shrikant I Bangdiwala
- Department of Health Research Methods, Evidence and Impact McMaster University Hamilton ON Canada
- Statistics Department Population Health Research Institute, McMaster University Hamilton ON Canada
| | | | - Jan H Cornel
- Department of Cardiology Radboud University Medical Center Nijmegen The Netherlands
- Dutch Network for Cardiovascular Research (WCN) Utrecht The Netherlands
- Department of Cardiology Northwest Clinics Alkmaar Netherlands
| | - Saloua El Messaoudi
- Department of Cardiology Radboud University Medical Center Nijmegen The Netherlands
| |
Collapse
|
3
|
Rochmawati ID, Deo S, Lees JS, Mark PB, Sattar N, Celis-Morales C, Pell JP, Welsh P, Ho FK. Adding traditional and emerging biomarkers for risk assessment in secondary prevention: a prospective cohort study of 20 656 patients with cardiovascular disease. Eur J Prev Cardiol 2025; 32:585-595. [PMID: 39474888 DOI: 10.1093/eurjpc/zwae352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/26/2024] [Accepted: 10/14/2024] [Indexed: 05/13/2025]
Abstract
AIMS This study aims to explore whether conventional and emerging biomarkers could improve risk discrimination and calibration in the secondary prevention of recurrent atherosclerotic cardiovascular disease (ASCVD), based on a model using predictors from SMART2 (Secondary Manifestations of ARTerial Disease). METHODS AND RESULTS In a cohort of 20 658 UK Biobank participants with medical history of ASCVD, we analysed any improvement in C indices and net reclassification index (NRI) for future ASCVD events, following addition of lipoprotein A (LP-a), apolipoprotein B, Cystatin C, Hemoglobin A1c (HbA1c), gamma-glutamyl transferase (GGT), aspartate aminotransferase (AST), alanine aminotransferase, and alkaline phosphatase (ALP), to a model with predictors used in SMART2 for the outcome of recurrent major cardiovascular event. We also examined any improvement in C indices and NRIs replacing creatinine-based estimated glomerular filtration rate (eGFR) with Cystatin C-based estimates. Calibration plots between different models were also compared. Compared with the baseline model (C index = 0.663), modest increments in C indices were observed when adding HbA1c (ΔC = 0.0064, P < 0.001), Cystatin C (ΔC = 0.0037, P < 0.001), GGT (ΔC = 0.0023, P < 0.001), AST (ΔC = 0.0007, P < 0.005) or ALP (ΔC = 0.0010, P < 0.001) or replacing eGFRCr with eGFRCysC (ΔC = 0.0036, P < 0.001) or eGFRCr-CysC (ΔC = 0.00336, P < 0.001). Similarly, the strongest improvements in NRI were observed with the addition of HbA1c (NRI = 0.014) or Cystatin C (NRI = 0.006) or replacing eGFRCr with eGFRCr-CysC (NRI = 0.001) or eGFRCysC (NRI = 0.002). There was no evidence that adding biomarkers modified calibration. CONCLUSION Adding several biomarkers, most notably Cystatin C and HbA1c, but not LP-a, in a model using SMART2 predictors modestly improved discrimination.
Collapse
Affiliation(s)
- Ike Dhiah Rochmawati
- School of Health and Wellbeing, University of Glasgow, 90 Byres Road, Glasgow G12 8TB, UK
- Department of Clinical and Community Pharmacy, Faculty of Pharmacy, University of Surabaya, Jalan Raya Kalirungkut, Surabaya 60293, Indonesia
| | - Salil Deo
- Louis Stokes Cleveland VA Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Jennifer S Lees
- School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Patrick B Mark
- School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Carlos Celis-Morales
- School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
- Human Performance Lab, Education, Physical Activity and Health Research Unit, University Católica del Maule, San Miguel Avenue 3605, Talca 3466706, Chile
- Centro de Investigacion en Medicina de Altura (CEIMA), Universidad Arturo Prat, Avenue Arturo Prat, Iquique 2120, Chile
| | - Jill P Pell
- School of Health and Wellbeing, University of Glasgow, 90 Byres Road, Glasgow G12 8TB, UK
| | - Paul Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Frederick K Ho
- School of Health and Wellbeing, University of Glasgow, 90 Byres Road, Glasgow G12 8TB, UK
| |
Collapse
|
4
|
van Steenkiste J, van Dorst P, Dohmen D, Boersma C. Prerequisites for Cost-Effective Home Blood Pressure Telemonitoring: Early Health Economic Analysis. JMIR Cardio 2025; 9:e64386. [PMID: 40340846 PMCID: PMC12080967 DOI: 10.2196/64386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 05/10/2025] Open
Abstract
Background Home blood pressure telemonitoring (HBPT) has been proposed to enhance adherence and optimize health care delivery, yet its prerequisites for cost-effective implementation remain unclear. Objective This study aims to quantify the potential cost-effectiveness of HBPT and identify prerequisites for cost-effective implementation of HBPT in comparison to standard hypertension management, using an early health economic analysis from a societal perspective. Methods A decision-analytic Markov model with a lifetime horizon (30 years) and a willingness-to-pay threshold of €20,000 (€1=US $1.09) per quality-adjusted life year (QALY) was developed to assess the cost-effectiveness of HBPT compared to standard of care (SOC). The HBPT intervention was based on an existing HBPT program applied by the Maasstad Hospital, Rotterdam, the Netherlands. The model incorporated 12 health states: 7 blood pressure states, 1 cardiovascular (CV) event, 1 recurrent CV event, 1 postrecurrent CV event, 1 all-cause death, and 1 CV disease-related death. A hypothetical cohort of 1000 patients (average age 65.3 years) was modeled, and results were reported in costs, QALYs, and the incremental cost-effectiveness ratio (ICER). The model assumed 3 in-person outpatient department (OPD) consultations in the SOC group and 1.5 in the HBPT group. Extensive sensitivity analyses were performed to identify important variables for the cost-effective implementation of HBPT. Results Following the base-case analysis, HBPT was not cost-effective with an ICER of €20,386 per QALY. Sensitivity analyses indicated that reducing the number of in-person OPD consultations resulted in a more favorable ICER. Specifically, reducing the number of in-person OPD consultations to 1.48 annually resulted in an ICER below the willingness-to-pay threshold. Reducing the in-person OPD consultations to an average of 1.18 per year would make HBPT cost-saving. Scenario analyses revealed that extending the duration of HBPT's clinical effect to 2 or 3 years substantially improved the ICER. Additionally, targeting HBPT toward patients aged 64 years or below further improved the ICER. Conclusions HBPT could result in cost-effective or cost-saving outcomes with only minor reductions in in-person OPD consultations. These findings highlight the potential of HBPT to transform hypertension management by replacing traditional hypertension management with more efficient care using remote patient monitoring.
Collapse
Affiliation(s)
- Job van Steenkiste
- Department of Internal Medicine, Maasstad Hospital, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Faculty of Management Sciences, Open University, Valkenburgerweg 177, Heerlen, 6419 AT, The Netherlands
| | - Pim van Dorst
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Health-Ecore Ltd, Zeist, The Netherlands
| | - Daan Dohmen
- Faculty of Management Sciences, Open University, Valkenburgerweg 177, Heerlen, 6419 AT, The Netherlands
| | - Cornelis Boersma
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Health-Ecore Ltd, Zeist, The Netherlands
| |
Collapse
|
5
|
Chen M, Ma X, Xue Y, Wang X, Zhao Y, Yan P, Liu X, Du Y, Qiu C, Sun Q. Association of Sleep Duration With Incident Carotid Plaque: A Prospective Cohort Study. J Am Heart Assoc 2025; 14:e039215. [PMID: 40314385 DOI: 10.1161/jaha.124.039215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/08/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Sleep is an important determinant of cardiovascular health. We sought to investigate the longitudinal association between sleep duration and incident carotid plaque in a rural Chinese population. METHODS This population-based prospective cohort study included 1004 rural residents (age ≥40 years) who were free of carotid plaque and had no history of clinical stroke and transient ischemic attack at baseline (2017). Incident carotid plaques were detected by carotid ultrasound images at follow-up (2021). Multivariable Cox regression was used to associate sleep duration with the presence and severity of incident carotid plaques. Restricted cubic splines analyses were conducted to assess dose-response association between sleep duration and incident carotid plaques. RESULTS During the mean follow-up of 3.95 (SD=0.14) years, 214 (21.3%) of the 1004 participants were found to have incident carotid plaques. A short sleep duration (<7 versus 7-9 hours) was associated with multivariable-adjusted hazard ratio (95% CI) of 1.58 (1.10-2.28) for carotid plaques, 2.96 (1.38-6.36) for greater carotid plaque thickness, and 2.57 (1.33-4.97) for multiple carotid plaques; those associations remained significant in participants with low-to-intermediate traditional cardiovascular disease risk. Long sleep duration (>9 versus 7-9 hours) was not significantly associated with carotid plaques. Restricted cubic splines supported the association of short, but not long, sleep duration with increased risk of incident carotid plaques. CONCLUSIONS A short sleep duration is a risk factor for carotid plaques, even among individuals with low-to-intermediate cardiovascular disease risk. This suggests that short sleep duration may be a potential target for early interventions to delay carotid atherosclerosis. REGISTRATION URL: https://www.chictr.org.cn; Unique Identifier: ChiCTR1800017197.
Collapse
Affiliation(s)
- Meijie Chen
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong P. R. China
| | - Xiaotong Ma
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong P. R. China
| | - Yuan Xue
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong P. R. China
| | - Xiang Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong P. R. China
| | - Yuanyuan Zhao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong P. R. China
| | - Peng Yan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong P. R. China
| | - Xiaohui Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong P. R. China
| | - Yifeng Du
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong P. R. China
| | - Chengxuan Qiu
- Aging Research Center, Department of Neurobiology Care Sciences and Society, Karolinska Institutet-Stockholm University Stockholm Sweden
| | - Qinjian Sun
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong P. R. China
| |
Collapse
|
6
|
Yang S, Pan Y, Zheng W. Baseline High-Sensitivity C-Reactive Protein as a Predictor of Adverse Clinical Events in Patients with Coronary Artery Disease Undergoing Percutaneous Coronary Intervention: A Meta-Analysis. Cardiol Rev 2025; 33:227-238. [PMID: 37754679 DOI: 10.1097/crd.0000000000000604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Inflammation in patients with coronary artery disease (CAD) has been linked to adverse clinical outcomes. A useful biomarker for measuring inflammation levels, high-sensitivity C-reactive protein (hs-CRP) in the blood can be used to detect the presence of low-grade inflammation. This study sought to assess the predictive value of baseline hs-CRP levels for adverse clinical events in CAD patients undergoing percutaneous coronary intervention (PCI). To investigate this topic, a meta-analysis was performed. We conducted a systematic search of PubMed, Embase, and the Cochrane Library for original articles reporting the correlation between hs-CRP levels and adverse clinical events in CAD patients undergoing PCI. We followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines and conducted a meta-analysis by extracting relevant data. Our pooled calculations yielded hazard ratios or odds ratios with 95% confidence intervals. A total of 28 studies comprising 60544 patients were included in this analysis. High baseline hs-CRP levels predicted increased risk for major adverse cardiac events ( P = 0.037), major adverse cardiac and cerebrovascular events ( P = 0.020), all-cause mortality ( P = 0.001), cardiovascular mortality ( P < 0.001), death and/or myocardial infarction ( P = 0.017) in patients, as well as restenosis ( P < 0.001). However, there was no association between elevated baseline hs-CRP levels and thrombosis. In conclusion, in CAD patients undergoing PCI, baseline hs-CRP levels are reliable predictors of major adverse cardiac events, major adverse cardiac and cerebrovascular events, all-cause mortality, cardiovascular mortality, death and/or myocardial infarction, and restenosis. Therefore, hs-CRP can effectively assist in prognosis determination for CAD patients undergoing PCI.
Collapse
Affiliation(s)
- Shanshan Yang
- From the Department of Cardiovascular Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yuxiang Pan
- Department of Critical Care Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Wan Zheng
- From the Department of Cardiovascular Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
7
|
Qureshi AI, Baskett WI, Bhatti IA, Ovbiagele B, Siddiq F, Ford DE, Gomez CR, Hanley DF, Shyu CR. Post 90-day outcomes of acute ischemic stroke patients following thrombectomy: analysis of real-world data. Front Neurol 2025; 16:1543101. [PMID: 40371087 PMCID: PMC12074927 DOI: 10.3389/fneur.2025.1543101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/09/2025] [Indexed: 05/16/2025] Open
Abstract
Background Previous studies have focused on 90-day outcomes in acute ischemic stroke patients who undergo thrombectomy, although long-term outcomes are not well understood. We compared the long-term rates of survival and new stroke recurrence among acute ischemic stroke patients who did and did not undergo thrombectomy. Methods Using the Oracle Real-World Data (a de-identified large data source of multicenter electronic health records covering the period of January 2016 to January 2023), we analyzed 3,934 acute ischemic stroke patients who underwent thrombectomy and 3,934 propensity-matched controls of acute ischemic stroke patients who did not undergo thrombectomy. The risk of death or palliative care and new stroke following >90 days post-admission was ascertained using Cox proportional hazards regression analysis to adjust for potential confounders. We also estimated the rate of new stroke and palliative care-free survival using Kaplan-Meier survival analysis. Results Among 3,934 acute ischemic stroke patients who underwent thrombectomy, 2,660 patients either died or received palliative care or developed new stroke (median follow-up period of 775 days post-initial stroke admission; interquartile range Q1 = 356 days, Q3 = 1,341 days). The 2-year new stroke and palliative care-free survival were 36.6 and 45.8% among patients who did and did not undergo thrombectomy, respectively (adjusted hazard ratio [HR], 1.19, 95% confidence interval [CI], 1.12-1.26). The risk of palliative care or death was not different (adjusted HR, 0.89, 95% CI, 0.77-1.02) between both groups, but the risk of new stroke was higher among patients who underwent thrombectomy (adjusted HR, 1.25, 95% CI, 1.18-1.33). Conclusion Acute ischemic stroke patients who undergo thrombectomy are at greater risk of new stroke, palliative care, or death after 90 days, primarily driven by the occurrence of stroke. There is a need for closer surveillance and enhanced recurrent stroke prevention in this high-risk group.
Collapse
Affiliation(s)
- Adnan I. Qureshi
- Zeenat Qureshi Stroke Institutes, Columbia, MO, United States
- Department of Neurology, University of Missouri, Columbia, MO, United States
| | - William I. Baskett
- Department of Data Science and Informatics, University of Missouri, Columbia, MO, United States
| | - Ibrahim A. Bhatti
- Zeenat Qureshi Stroke Institutes, Columbia, MO, United States
- Department of Neurology, University of Missouri, Columbia, MO, United States
- Department of Neurosurgery, University of Missouri, Columbia, MO, United States
| | - Bruce Ovbiagele
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Farhan Siddiq
- Department of Neurosurgery, University of Missouri, Columbia, MO, United States
| | | | - Camilo R. Gomez
- Department of Neurology, University of Missouri, Columbia, MO, United States
| | | | - Chi-Ren Shyu
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, United States
| |
Collapse
|
8
|
Ricco JB, Hostalrich A. Patients with Abdominal Aortic Aneurysm: Are We Missing Something? Eur J Vasc Endovasc Surg 2025:S1078-5884(25)00276-X. [PMID: 40188864 DOI: 10.1016/j.ejvs.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/17/2025] [Accepted: 04/01/2025] [Indexed: 05/09/2025]
Affiliation(s)
- Jean-Baptiste Ricco
- Research and Anatomy Laboratory, ABS LAB, University of Poitiers, Poitiers, France; Department of Vascular Surgery, University Hospital of Rangueil, Toulouse, France.
| | - Aurélien Hostalrich
- Department of Vascular Surgery, University Hospital of Rangueil, Toulouse, France
| |
Collapse
|
9
|
Wang Y, Aivalioti E, Stamatelopoulos K, Zervas G, Mortensen MB, Zeller M, Liberale L, Di Vece D, Schweiger V, Camici GG, Lüscher TF, Kraler S. Machine learning in cardiovascular risk assessment: Towards a precision medicine approach. Eur J Clin Invest 2025; 55 Suppl 1:e70017. [PMID: 40191920 DOI: 10.1111/eci.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/22/2025] [Indexed: 04/24/2025]
Abstract
Cardiovascular diseases remain the leading cause of global morbidity and mortality. Validated risk scores are the basis of guideline-recommended care, but most scores lack the capacity to integrate complex and multidimensional data. Limitations inherent to traditional risk prediction models and the growing burden of residual cardiovascular risk highlight the need for refined strategies that go beyond conventional paradigms. Artificial intelligence and machine learning (ML) provide unique opportunities to refine cardiovascular risk assessment and surveillance through the integration of diverse data types and sources, including clinical, electrocardiographic, imaging and multi-omics derived data. In fact, ML models, such as deep neural networks, can handle high-dimensional data through which phenotyping and cardiovascular risk assessment across diverse patient populations become much more precise, fostering a paradigm shift towards more personalized care. Here, we review the role of ML in advancing cardiovascular risk assessment and discuss its potential to identify novel therapeutic targets and to improve prevention strategies. We also discuss key challenges inherent to ML, such as data quality, standardized reporting, model transparency and validation, and discuss barriers in its clinical translation. We highlight the transformative potential of ML in precision cardiology and advocate for more personalized cardiovascular prevention strategies that go beyond previous notions.
Collapse
Affiliation(s)
- Yifan Wang
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Evmorfia Aivalioti
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Georgios Zervas
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Martin Bødtker Mortensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marianne Zeller
- Department of Cardiology, CHU Dijon Bourgogne, Dijon, France
- Physiolopathologie et Epidémiologie Cérébro-Cardiovasculaire (PEC2), EA 7460, Univ Bourgogne, Dijon, France
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Davide Di Vece
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Victor Schweiger
- Deutsches Herzzentrum der Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals GSTT and Cardiovascular Academic Group, King's College, London, UK
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Internal Medicine and Cardiology, Cantonal Hospital Baden, Baden, Switzerland
| |
Collapse
|
10
|
Popiolek-Kalisz J, Mazur M, Perone F. The Role of Dietary Education in Cardiac Rehabilitation. Nutrients 2025; 17:1082. [PMID: 40292477 PMCID: PMC11945974 DOI: 10.3390/nu17061082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Cardiovascular disease remains a leading cause of death globally; however, most cases could be prevented by addressing modifiable risk factors, such as unhealthy lifestyle factors, including diet. These aspects are also crucial in secondary prevention. Cardiac rehabilitation programs are vital in improving cardiovascular outcomes, and apart from recommended pharmacotherapy, they focus on lifestyle modifications, including exercise, a healthy diet, and smoking cessation. The aim of this review was to summarize the evidence on the role of dietary education in cardiac rehabilitation programs. The available data show that nutritional recommendations play an important role in cardiac rehabilitation programs, with the Mediterranean diet being widely recommended for its cardiovascular benefits. Adherence to dietary recommendations in the course of cardiac rehabilitation has been linked to improved metabolic and cardiovascular outcomes; however, further studies with long-term follow-up are needed. Moreover, while challenges in following dietary recommendations exist, individualized care and support are essential for successful outcomes in cardiac rehabilitation programs. Including dietary education is an important part of cardiac rehabilitation after myocardial infarction; however, more studies are needed to investigate the role of individualized dietary support and personalized education in cardiac rehabilitation.
Collapse
Affiliation(s)
- Joanna Popiolek-Kalisz
- Department of Clinical Dietetics, Medical University of Lublin, ul. Chodzki 7, 20-093 Lublin, Poland; (J.P.-K.)
- Department of Cardiology, Cardinal Wyszynski Hospital in Lublin, al. Krasnicka 100, 20-718 Lublin, Poland
| | - Michal Mazur
- Department of Clinical Dietetics, Medical University of Lublin, ul. Chodzki 7, 20-093 Lublin, Poland; (J.P.-K.)
| | - Francesco Perone
- Cardiac Rehabilitation Unit, Rehabilitation Clinic ‘Villa delle Magnolie’, 81020 Castel Morrone, Caserta, Italy;
| |
Collapse
|
11
|
Campbell MK, Chew NWS, Mehta A. Beyond Cholesterol: Unraveling Residual Lipidomic Risk in Cardiovascular Health. Curr Atheroscler Rep 2025; 27:37. [PMID: 40095146 DOI: 10.1007/s11883-025-01284-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE OF REVIEW This paper reviews the existing literature on lipidomics as a tool for improved cardiovascular risk estimation in both primary and secondary prevention populations. RECENT FINDINGS Detailed lipidomic signatures identified by mass spectrometry have been shown to enhance risk estimation for clinical CAD and the presence of subclinical CAD on CTCA in multiple large cohort populations. In patients with established atherosclerotic disease, ceramide and phospholipid-based risk scores improve prediction for recurrent cardiovascular events and cardiovascular death. Lipidomic profiles and lipidomic-enhanced risk scores have been shown to improve prediction of incident cardiovascular disease, recurrent cardiovascular events and cardiovascular death independent of traditional risk factors. Simplified risk scores utilizing the ratios of several ceramide species improve clinical utility, however resources and infrastructure limit widespread implementation. There are currently no therapeutics to address lipidomic risk aside from traditional risk factor modification.
Collapse
Affiliation(s)
- Matthew K Campbell
- Department of Internal Medicine, Virginia Commonwealth University Health, Virginia Commonwealth University School of Medicine, Richmond, USA
| | - Nicholas W S Chew
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anurag Mehta
- VCU Health Pauley Heart Center, Virginia Commonwealth University School of Medicine, PO Box 980036, 1200 East Broad Street, VA, 23298, Richmond, USA.
| |
Collapse
|
12
|
Foo CY, Nadia Mansor NA, Erng T, Mohamed MS, Mahadevan G, Lau G, Ranga A, Ong TK. Exploring the Potential Health and Economic Benefits of Optimized Low-Density Lipoprotein Cholesterol Management in Malaysia's Atherosclerotic Cardiovascular Disease Population: A Model-Based Analysis. Value Health Reg Issues 2025; 46:101059. [PMID: 39580723 DOI: 10.1016/j.vhri.2024.101059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/20/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVES This study quantified the health and economic benefits of improving low-density lipoprotein cholesterol (LDL-C) control in Malaysian patients with established atherosclerotic cardiovascular disease (ASCVD). It aimed to inform policy discussions and healthcare planning for effective ASCVD management. METHODS A deterministic, prevalence-based model was used to project the annual health burden and direct medical costs associated with recurrent ASCVD events over a 10-year horizon. The target population included adults (≥30 years) with established ASCVD and uncontrolled LDL-C levels (>1.8 mmol/L). The model comprised 3 modules: population size projection, recurrent ASCVD risk calculation (by means of the Secondary Manifestations of ARTerial disease [SMART] risk model), and direct medical and productivity cost estimation. The current status quo and a scenario with a 50% improvement in mean LDL-C were compared. RESULTS We projected over 800 000 adults with established ASCVD in 2023, increasing to approximately 1.4 million by 2032. Under the status quo, about 55 000 recurrent ASCVD events were expected within 10 years, with significant direct medical costs and productivity losses. Improved LDL-C control could potentially reduce recurrent events by 7000 cases (13% reduction), prevent 2100 premature deaths, and save approximately 32 400 years of life. Economically, this could lead to a reduction of approximately 72 million MYR in direct medical costs and a gain of approximately 132.4 million MYR in productivity over a decade. CONCLUSIONS Optimizing LDL-C control in high-risk patients with ASCVD presents a critical opportunity to reduce health and economic burdens in Malaysia.
Collapse
Affiliation(s)
- Chee Yoong Foo
- Real-World Solutions, IQVIA, Petaling Jaya, Selangor, Malaysia.
| | | | - Thurston Erng
- Department of Cardiology, Serdang Hospital, Kajang, Selangor, Malaysia
| | - Mohd Sapawi Mohamed
- Cardiology Unit, Hospital Raja Perempuan Zainab II, Kota Bharu, Kelantan, Malaysia
| | - Gurudevan Mahadevan
- Department of Cardiology, Hospital Sultanah Aminah, Johor Bahru, Johor, Malaysia
| | - Glendon Lau
- Department of Cardiology, Serdang Hospital, Kajang, Selangor, Malaysia
| | - Asri Ranga
- Department of Cardiology, Serdang Hospital, Kajang, Selangor, Malaysia
| | - Tiong Kiam Ong
- Department of Cardiology, Sarawak Heart Centre, Kota Samarahan, Sarawak, Malaysia
| |
Collapse
|
13
|
Banerjee S, Weideman RA, Jacob DA, DiGregorio HR, Kelly KC, Banerjee A, Ravishankar M, Strickland PT, Abraham HM, Minniefield NE, Grimsley BR, Schussler JM, Parmar RJ, Stoler RC, Brilakis ES, Little BB. Estimating the Risk of Cardiovascular Events in U.S. Veterans Using the SMART Risk Score. JACC. ADVANCES 2025; 4:101459. [PMID: 39759440 PMCID: PMC11697775 DOI: 10.1016/j.jacadv.2024.101459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 01/07/2025]
Abstract
Background Estimation of long-term risk for cardiovascular events using the SMART (Secondary Manifestations of Arterial Disease) risk score can be potentially valuable in devising risk mitigation strategies. Objectives The objective of this study was to apply the SMART risk score to compute the risk for major adverse cardiovascular events (MACE) in the U.S. Veteran patient population. Methods We used the Veterans Affairs (VA) informatics and computing infrastructure to identify patients referred for an initial outpatient cardiology evaluation between the years 2003 and 2010 to estimate 10-year risk for composite MACE (all-cause death, ischemic stroke, and nonfatal myocardial infarction). Cox regression and survival curves were used to develop and validate the VA SMART score. Results The study population included 472,702 patients (mean age 60 ± 8.9 years, 96% male) who were allocated into development (n = 94,091) and test cohorts (n = 378,611). The median follow-up time was 7.9 years (IQR: 6.0-9.9). The VA-SMART score allowed accurate estimation of MACE. Patients were stratified in low (<10%), moderate (10% to 20%), high (20% to 30%), and very high (≥30%) risk groups with observed events rates of 6.8%, 17.9%, 28.5%, and 49.5%, respectively, in the test cohort (P < 0.0001 for all intergroup comparisons). Most MACE events were all-cause death, with nonfatal myocardial infarction and stroke also being high, especially in the very high-risk group. The VA SMART score performed similar to other established risk prediction models (C-statistic = 0.67). Conclusions The VA SMART risk score can estimate the long-term risk of recurrent cardiovascular events in U.S. Veterans and could help implement individualized risk mitigation strategies.
Collapse
Affiliation(s)
- Subhash Banerjee
- Baylor Scott & White Heart and Vascular Hospital, Dallas, Texas, USA
- Baylor University Medical Center, Dallas, Texas, USA
- Veterans Affairs North Texas Healthcare System, Dallas, Texas, USA
| | - Rick A. Weideman
- Veterans Affairs North Texas Healthcare System, Dallas, Texas, USA
| | - David A. Jacob
- Veterans Affairs North Texas Healthcare System, Dallas, Texas, USA
| | | | - Kevin C. Kelly
- Veterans Affairs North Texas Healthcare System, Dallas, Texas, USA
| | | | | | | | | | - Nicole E. Minniefield
- Veterans Affairs North Texas Healthcare System, Dallas, Texas, USA
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bradley R. Grimsley
- Baylor Scott & White Heart and Vascular Hospital, Dallas, Texas, USA
- Baylor University Medical Center, Dallas, Texas, USA
| | - Jeffrey M. Schussler
- Baylor Scott & White Heart and Vascular Hospital, Dallas, Texas, USA
- Baylor University Medical Center, Dallas, Texas, USA
| | - Rohit J. Parmar
- Baylor Scott & White Heart and Vascular Hospital, Dallas, Texas, USA
- Baylor University Medical Center, Dallas, Texas, USA
| | - Robert C. Stoler
- Baylor Scott & White Heart and Vascular Hospital, Dallas, Texas, USA
- Baylor University Medical Center, Dallas, Texas, USA
| | - Emmanouil S. Brilakis
- Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Minneapolis, Minnesota, USA
| | - Bertis B. Little
- Veterans Affairs North Texas Healthcare System, Dallas, Texas, USA
- University of Louisville School of Public Health and Information Sciences, Louisville, Kentucky, USA
| |
Collapse
|
14
|
Gallo A, Le Goff W, Santos RD, Fichtner I, Carugo S, Corsini A, Sirtori C, Ruscica M. Hypercholesterolemia and inflammation-Cooperative cardiovascular risk factors. Eur J Clin Invest 2025; 55:e14326. [PMID: 39370572 PMCID: PMC11628670 DOI: 10.1111/eci.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Maintaining low concentrations of plasma low-density lipoprotein cholesterol (LDLc) over time decreases the number of LDL particles trapped within the artery wall, slows the progression of atherosclerosis and delays the age at which mature atherosclerotic plaques develop. This substantially reduces the lifetime risk of atherosclerotic cardiovascular disease (ASCVD) events. In this context, plaque development and vulnerability result not only from lipid accumulation but also from inflammation. RESULTS Changes in the composition of immune cells, including macrophages, dendritic cells, T cells, B cells, mast cells and neutrophils, along with altered cytokine and chemokine release, disrupt the equilibrium between inflammation and anti-inflammatory mechanisms at plaque sites. Considering that it is not a competition between LDLc and inflammation, but instead that they are partners in crime, the present narrative review aims to give an overview of the main inflammatory molecular pathways linked to raised LDLc concentrations and to describe the impact of lipid-lowering approaches on the inflammatory and lipid burden. Although remarkable changes in LDLc are driven by the most recent lipid lowering combinations, the relative reduction in plasma C-reactive protein appears to be independent of the magnitude of LDLc lowering. CONCLUSION Identifying clinical biomarkers of inflammation (e.g. interleukin-6) and possible targets for therapy holds promise for monitoring and reducing the ASCVD burden in suitable patients.
Collapse
Affiliation(s)
- Antonio Gallo
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Wilfried Le Goff
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Raul D. Santos
- Academic Research Organization Hospital Israelita Albert Einstein and Lipid Clinic Heart Institute (InCor)University of Sao Paulo Medical School HospitalSao PauloBrazil
| | - Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Stefano Carugo
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Cesare Sirtori
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
15
|
Bruun K, Mortensen MB. Rethinking atherosclerotic cardiovascular disease prevention in the era of expanding therapies: could plaque stabilization reduce the need for lifelong treatments and polypharmacy? Curr Opin Cardiol 2025; 40:50-55. [PMID: 39436382 DOI: 10.1097/hco.0000000000001188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
PURPOSE OF REVIEW This review examines current evidence on pharmacologically induced plaque stabilization in the context of a growing range of new therapies. It explores the potential for a paradigm shift in atherosclerotic cardiovascular disease (ASCVD) prevention, where treatments may not need to be lifelong to achieve lasting benefits. RECENT FINDINGS Since 2015, over 14 novel therapies have been introduced, each shown to reduce ASCVD risk when added to standard care with statins and aspirin. More than 80% of ischemic heart disease patients are now eligible for one or more of these treatments, increasing the risk of polypharmacy, treatment burden, and adverse side effects. As more therapies become available, this challenge is expected to grow. Many of these treatments have demonstrated plaque regression and stabilization, as evidenced by both intravascular ultrasound and computed tomography angiography, which likely explains much of their efficacy. SUMMARY The increasing number of novel therapies presents challenges in preventing ASCVD without leading to lifelong polypharmacy and increased patient burden. Since many of these drugs act through plaque stabilization, a new approach may be feasible - using these treatments for shorter durations to induce plaque regression, followed by less intensive maintenance therapies to preserve stability. This approach warrants further investigation in future studies.
Collapse
Affiliation(s)
- Kathrine Bruun
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Martin Bødtker Mortensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Wu XD, Ye XY, Liu XY, Lin Y, Lin X, Li YY, Ye BH, Sun JC. Benefits of intensive lipid-lowering therapies in patients with acute coronary syndrome: a systematic review and meta-analysis. Ann Med 2024; 56:2389470. [PMID: 39126262 PMCID: PMC11318487 DOI: 10.1080/07853890.2024.2389470] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/23/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Previous meta-analyses have investigated the efficacy of lipid-lowering therapies for atherosclerotic cardiovascular disease; however, few have focused on patients with acute coronary syndrome (ACS). This meta-analysis aimed to compare the benefits of intensive lipid-lowering therapy with those of background statin therapy in patients with ACS. METHODS Searches were performed on PubMed, Embase, the Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov databases for articles published until April 13, 2023. Randomized controlled trials that compared intensive lipid-lowering therapies and background statin therapies in patients with prior ACS and recorded the outcome of three-point major cardiovascular events (MACE) were included. The risk ratio (RR) with 95% confidence interval (CI) was used as a measure of primary and secondary outcomes. RESULTS Nine trials involving 38,640 patients with ACS were identified. Pooled results suggested that intensive lipid-lowering therapies are associated with a reduction in the risk of three-point MACE (RR, 0.88; 95% CI, 0.83-0.94; p < 0.001), recurrent ACS (RR, 0.82; 95% CI, 0.71-0.96; p = 0.013), nonfatal myocardial infarction (MI) (RR, 0.87; 95% CI, 0.81-0.93; p < 0.001), stroke (RR, 0.83; 95% CI, 0.73-0.94; p = 0.003), and unstable angina-related hospitalization (RR, 0.57; 95% CI, 0.33-0.99; p = 0.046), but not all-cause mortality (RR, 0.94; 95% CI, 0.82-1.07; p = 0.329), cardiovascular disease-related mortality (RR, 0.96; 95% CI, 0.88-1.06; p = 0.457) or coronary revascularization (RR, 0.89; 95% CI, 0.79-1.00; p = 0.057). CONCLUSIONS Intensive lipid-lowering therapies may reduce the risk of three-point MACE, recurrent ACS, nonfatal MI, stroke, and hospitalization for unstable angina in patients with ACS undergoing background statin therapy. These results may assist in clinical decision-making for the secondary prevention of cardiovascular events to initiate intensive lipid-lowering therapies immediately after ACS.
Collapse
Affiliation(s)
- Xian-Dan Wu
- Department of General Medicine, The First people’s hospital of Wenling, Taizhou, Zhejiang, China
| | - Xin-Yue Ye
- Shanghai Jiao Tong University, Shanghai, China
| | - Xuan-Yan Liu
- Department of General Medicine, The First people’s hospital of Wenling, Taizhou, Zhejiang, China
| | - Yue Lin
- Department of General Medicine, The First people’s hospital of Wenling, Taizhou, Zhejiang, China
| | - Xian Lin
- Department of General Medicine, The First people’s hospital of Wenling, Taizhou, Zhejiang, China
| | - Yan-Yan Li
- Department of General Medicine, The First people’s hospital of Wenling, Taizhou, Zhejiang, China
| | - Bin-Hua Ye
- Department of General Medicine, The First people’s hospital of Wenling, Taizhou, Zhejiang, China
| | - Jing-Chao Sun
- Department of Cardiology, Taizhou Municipal Hospital, Taizhou, Zhejiang, China
| |
Collapse
|
17
|
De Cock E, Kautbally S, Timmermans F, Bogaerts K, Hanet C, Desmet W, Gurné O, Vranckx P, Hiltrop N, Dujardin K, Vanduynhoven P, Vermeersch P, Pirlet C, Hermans K, Van Reet B, Ferdinande B, Aminian A, Dewilde W, Guédès A, Simon F, De Roeck F, De Vroey F, Jukema JW, Sinnaeve P, Buysschaert I. Low-dose colchicine for the prevention of cardiovascular events after percutaneous coronary intervention: Rationale and design of the COL BE PCI trial. Am Heart J 2024; 278:61-71. [PMID: 39233210 DOI: 10.1016/j.ahj.2024.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION Patients with coronary artery disease (CAD) remain vulnerable to future major atherosclerotic events after revascularization, despite effective secondary prevention strategies. Inflammation plays a central role in the pathogenesis of CAD and recurrent events. To date, there is no specific anti-inflammatory medicine available with proven effective, cost-efficient, and favorable benefit-risk profile, except for colchicine. Initial studies with colchicine have sparked major interest in targeting atherosclerotic events with anti-inflammatory agents, but further studies are warranted to enforce the role of colchicine role as a major treatment pillar in CAD. Given colchicine's low cost and established acceptable long-term safety profile, confirming its efficacy through a pragmatic trial holds the potential to significantly impact the global burden of cardiovascular disease. METHODS The COL BE PCI trial is an investigator-initiated, multicenter, double-blind, event-driven trial. It will enroll 2,770 patients with chronic or acute CAD treated with percutaneous coronary intervention (PCI) at 19 sites in Belgium, applying lenient in- and exclusion criteria and including at least 30% female participants. Patients will be randomized between 2 hours and 5 days post-PCI to receive either colchicine 0.5 mg daily or placebo on top of contemporary optimal medical therapy and without run-in period. All patients will have baseline hsCRP measurements and a Second Manifestations of Arterial Disease (SMART) risk score calculation. The primary endpoint is the time from randomization to the first occurrence of a composite endpoint consisting of all-cause death, spontaneous non-fatal myocardial infarction, non-fatal stroke, or coronary revascularization. The trial is event-driven and will continue until 566 events have been reached, providing 80% power to detect a 21 % reduction in the primary endpoint taking a premature discontinuation of 15% into account. We expect a trial duration of approximately 44 months. CONCLUSION The COL BE PCI Trial aims to assess the effectiveness and safety of administering low-dose colchicine for the secondary prevention in patients with both chronic and acute coronary artery disease undergoing PCI. TRIAL REGISTRATION ClinicalTrials.gov: NCT06095765.
Collapse
Affiliation(s)
- Emmanuel De Cock
- Department of Cardiology, AZ Sint-Jan Brugge AV, Bruges, Belgium; Department of Cardiology, Ghent University Hospital, Ghent, Belgium
| | - Shakeel Kautbally
- Department of Cardiology, Cliniques Universitaires St-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Frank Timmermans
- Department of Cardiology, Ghent University Hospital, Ghent, Belgium
| | - Kris Bogaerts
- Department of Public Health and Primary Care, KU Leuven, I-BioStat, Leuven, Belgium and UHasselt, I-BioStat, Diepenbeek, Belgium
| | - Claude Hanet
- Department of Cardiology, Université Catholique de Louvain, Mont Godinne, Yvoir, Belgium
| | - Walter Desmet
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium; Belgium & Department of Cardiovascular Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Olivier Gurné
- Department of Cardiology, Cliniques Universitaires St-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Pascal Vranckx
- Department of Cardiology and Intensive Care Medicine, Jessa Ziekenhuis, Hasselt, Belgium
| | - Nick Hiltrop
- Department of Cardiology, AZ Groeninge, Kortrijk, Belgium
| | - Karl Dujardin
- Department of Cardiology, AZ Delta, Roeselare, Belgium
| | | | - Paul Vermeersch
- Department of Cardiology, ZNA (Ziekenhuis Netwerk Antwerpen) Middelheim, Antwerp, Belgium
| | - Charles Pirlet
- Department of Cardiology, Citadelle Liège, Liège, Belgium
| | - Kurt Hermans
- Department of Cardiology, AZ Sint-Lucas Ghent, Ghent, Belgium
| | - Bert Van Reet
- Department of Cardiology, AZ Turnhout, Turnhout, Belgium
| | - Bert Ferdinande
- Department of Cardiology, Hospital Oost-Limburg, Genk, Belgium
| | - Adel Aminian
- Department of Cardiology, Centre Hospitalier Universitaire de Charleroi, Charleroi, Belgium
| | - Willem Dewilde
- Department of Cardiology, Imelda Hospital Bonheiden, Bonheiden, Belgium
| | - Antoine Guédès
- Department of Cardiology, Université Catholique de Louvain, Mont Godinne, Yvoir, Belgium
| | - François Simon
- Department of Cardiology, Clinique Saint-Luc Bouge, Namur, Belgium
| | - Frederic De Roeck
- Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| | - Frédéric De Vroey
- Department of Cardiology, Grand Hôpital de Charleroi, Charleroi, Belgium
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands; Netherlands Heart Institute, Utrecht, The Netherlands
| | - Peter Sinnaeve
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium; Belgium & Department of Cardiovascular Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Ian Buysschaert
- Department of Cardiology, AZ Sint-Jan Brugge AV, Bruges, Belgium.
| |
Collapse
|
18
|
Evans W, Akyea RK, Simms A, Kai J, Qureshi N. Opportunities and challenges for identifying undiagnosed Rare Disease patients through analysis of primary care records: long QT syndrome as a test case. J Community Genet 2024; 15:687-698. [PMID: 39405009 PMCID: PMC11645366 DOI: 10.1007/s12687-024-00742-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/02/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Patients with rare genetic diseases frequently experience significant diagnostic delays. Routinely collected data in the electronic health record (EHR) may be used to help identify patients at risk of undiagnosed conditions. Long QT syndrome (LQTS) is a rare inherited cardiac condition associated with significant morbidity and premature mortality. In this study, we examine LQTS as an exemplar disease to assess if clinical features recorded in the primary care EHR can be used to develop and validate a predictive model to aid earlier detection. METHODS 1495 patients with an LQTS diagnostic code and 7475 propensity-score matched controls were identified from 10.5 million patients' electronic primary care records in the UK's Clinical Practice Research Datalink (CPRD). Associated clinical features recorded before diagnosis (with p < 0.05) were incorporated into a multivariable logistic regression model, the final model was determined by backwards regression and validated by bootstrapping to determine model optimism. RESULTS The mean age at LQTS diagnosis was 58.4 (SD 19.41). 18 features were included in the final model. Discriminative accuracy, assessed by area under the curve (AUC), was 0.74, (95% CI 0.73, 0.75) (optimism 6%). Features occurring at significantly greater frequency before diagnosis included: epilepsy, palpitations, syncope, collapse, mitral valve disease and irritable bowel syndrome. CONCLUSION This study demonstrates the potential to develop primary care prediction models for rare conditions, like LQTS, in routine primary care records and highlights key considerations including disease suitability, finding an appropriate linked dataset, the need for accurate case ascertainment and utilising an approach to modelling suitable for rare events.
Collapse
Affiliation(s)
- William Evans
- Primary Care Stratified Medicine (PRISM), Centre for Academic Primary Care, School of Medicine, University of Nottingham, Applied Health Research Building [42], University Park, Nottingham, NG7 2RD, UK.
| | - Ralph K Akyea
- Primary Care Stratified Medicine (PRISM), Centre for Academic Primary Care, School of Medicine, University of Nottingham, Applied Health Research Building [42], University Park, Nottingham, NG7 2RD, UK
| | - Alex Simms
- Department of Cardiology, Leeds Teaching Hospital NHS Trust, Leeds, UK
| | - Joe Kai
- Primary Care Stratified Medicine (PRISM), Centre for Academic Primary Care, School of Medicine, University of Nottingham, Applied Health Research Building [42], University Park, Nottingham, NG7 2RD, UK
| | - Nadeem Qureshi
- Primary Care Stratified Medicine (PRISM), Centre for Academic Primary Care, School of Medicine, University of Nottingham, Applied Health Research Building [42], University Park, Nottingham, NG7 2RD, UK
| |
Collapse
|
19
|
Ferrante G, Bacallao BA, Gioia F, Del Monaco G, Amata F. 2024 European Society of Cardiology Guidelines on Peripheral Arterial and Aortic Diseases. Eur Cardiol 2024; 19:e24. [PMID: 39764064 PMCID: PMC11702006 DOI: 10.15420/ecr.2024.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 05/02/2025] Open
Affiliation(s)
- Giuseppe Ferrante
- Department of Biomedical Sciences, Humanitas UniversityMilan, Italy
- Department of Cardiovascular Medicine, Humanitas Research Hospital-IRCCSMilan, Italy
| | - Brittany A Bacallao
- Department of Cardiovascular Medicine, Humanitas Research Hospital-IRCCSMilan, Italy
| | - Francesco Gioia
- Department of Biomedical Sciences, Humanitas UniversityMilan, Italy
- Department of Cardiovascular Medicine, Humanitas Research Hospital-IRCCSMilan, Italy
| | - Guido Del Monaco
- Department of Biomedical Sciences, Humanitas UniversityMilan, Italy
- Department of Cardiovascular Medicine, Humanitas Research Hospital-IRCCSMilan, Italy
| | - Francesco Amata
- Department of Biomedical Sciences, Humanitas UniversityMilan, Italy
- Department of Cardiovascular Medicine, Humanitas Research Hospital-IRCCSMilan, Italy
| |
Collapse
|
20
|
Angiolillo DJ, Galli M, Alexopoulos D, Aradi D, Bhatt DL, Bonello L, Capodanno D, Cavallari LH, Collet JP, Cuisset T, Ferreiro JL, Franchi F, Geisler T, Gibson CM, Gorog DA, Gurbel PA, Jeong YH, Marcucci R, Siller-Matula JM, Mehran R, Neumann FJ, Pereira NL, Rizas KD, Rollini F, So DYF, Stone GW, Storey RF, Tantry US, Berg JT, Trenk D, Valgimigli M, Waksman R, Sibbing D. International Consensus Statement on Platelet Function and Genetic Testing in Percutaneous Coronary Intervention: 2024 Update. JACC Cardiovasc Interv 2024; 17:2639-2663. [PMID: 39603778 DOI: 10.1016/j.jcin.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 11/29/2024]
Abstract
Current evidence indicates that dual antiplatelet therapy with aspirin plus a P2Y12 inhibitor is essential for the prevention of thrombotic events after percutaneous coronary interventions. However, dual antiplatelet therapy is associated with increased bleeding which may outweigh the benefits. This has set the foundations for customizing antiplatelet treatments to the individual patient. However, bleeding and ischemic risks are often present in the same patient, making it difficult to achieve this balance. The fact that oral P2Y12 inhibitors (clopidogrel, prasugrel, and ticagrelor) have diverse pharmacodynamic profiles that affect clinical outcomes supports the rationale for using platelet function and genetic testing to individualize antiplatelet treatment regimens. Indeed, up to one-third of patients treated with clopidogrel, but a minority of those treated with prasugrel or ticagrelor, exhibit high residual platelet reactivity resulting in an increased thrombotic risk. On the other hand, prasugrel and ticagrelor are frequently associated with low platelet reactivity and increased bleeding risk compared with clopidogrel without providing any additional reduction in ischemic events compared with patients who adequately respond to clopidogrel. The use of platelet function and genetic testing may allow for a guided selection of oral P2Y12 inhibitors. However, the nonuniform results of randomized controlled trials have led guidelines to provide limited recommendations on the implementation of these tests in patients undergoing percutaneous coronary intervention. In light of recent advancements in the field, this consensus document by a panel of international experts fills in the guideline gap by providing updates on the latest evidence in the field as well as recommendations for clinical practice.
Collapse
Affiliation(s)
- Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, USA.
| | - Mattia Galli
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Dimitrios Alexopoulos
- 7th Department of Cardiology, Hygeia Hospital, Athens, Greece; State Hospital for Cardiology, Balatonfüred, Hungary
| | - Daniel Aradi
- State Hospital for Cardiology, Balatonfüred, Hungary; Hungary and Heart and Vascular Centre, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Laurent Bonello
- Intensive Care Unit, Hopital Universitaire Nord, Aix-Marseille University, Marseille, France
| | - Davide Capodanno
- Azienda Ospedaliero-Universitaria Policlinico G. Rodolico-San Marco, University of Catania, Catania, Italy
| | - Larisa H Cavallari
- Center for Pharmacogenomics and Precision Medicine, Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Jean-Philippe Collet
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Thomas Cuisset
- Department of Cardiology, La Timone Hospital, Marseille, France
| | - Jose Luis Ferreiro
- Department of Cardiology, Joan XXIII University Hospital, IISPV, Rovira i Virgili University, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Tarragona, Spain
| | - Francesco Franchi
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Tobias Geisler
- Department of Cardiology and Angiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - C Michael Gibson
- Baim Institute of Clinical Research, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Diana A Gorog
- Cardiovascular Division, Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom; Centre for Health Services and Clinical Research, Postgraduate Medical School, University of Hertfordshire, Hertfordshire, United Kingdom
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, Maryland, USA
| | - Young-Hoon Jeong
- CAU Thrombosis and Biomarker Center, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, South Korea; Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Jolanta M Siller-Matula
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Roxana Mehran
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School Medicine at Mount Sinai, New York, New York, USA
| | - Franz-Josef Neumann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Naveen L Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Konstantinos D Rizas
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Ludwig-Maximilians University, Munich, Germany; German Center for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany
| | - Fabiana Rollini
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Derek Y F So
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Gregg W Stone
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Robert F Storey
- Cardiovascular Research Unit, Division of Clinical Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, Maryland, USA
| | - Jurrien Ten Berg
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, the Netherlands; Department of Cardiology, University Medical Center Maastricht, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - Dietmar Trenk
- Clinical Pharmacology, Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Valgimigli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland; Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland; University of Bern, Bern, Switzerland
| | - Ron Waksman
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Dirk Sibbing
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Ludwig-Maximilians University, Munich, Germany; Privatklinik Lauterbacher Mühle am Ostsee, Seeshaupt, Germany
| |
Collapse
|
21
|
Mazhar F, Faucon AL, Fu EL, Szummer KE, Mathisen J, Gerward S, Reuter SB, Marx N, Mehran R, Carrero JJ. Systemic inflammation and health outcomes in patients receiving treatment for atherosclerotic cardiovascular disease. Eur Heart J 2024; 45:4719-4730. [PMID: 39211962 PMCID: PMC11578643 DOI: 10.1093/eurheartj/ehae557] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS The burden and outcomes of inflammation in patients with atherosclerotic cardiovascular disease (ASCVD) are not well defined beyond the controlled settings of trials and research cohorts. METHODS This was an observational study of ASCVD adults undergoing C-reactive protein testing in Stockholm's healthcare (2007-21). After excluding C-reactive protein tests associated with acute illness or medications/conditions that bias C-reactive protein interpretation, systemic inflammation was evaluated over a 3-month ascertainment window. Determinants of C-reactive protein ≥ 2 mg/L were explored with logistic regression. C-reactive protein categories were compared via negative-binomial/Cox regression for subsequent healthcare resource utilization and occurrence of major adverse cardiovascular events, heart failure hospitalization, and death. RESULTS A total of 84 399 ASCVD adults were included (46% female, mean age 71 years, 59% with C-reactive protein ≥ 2 mg/L). Female sex, older age, lower kidney function, albuminuria, diabetes, hypertension, and recent anaemia were associated with higher odds of C-reactive protein ≥ 2 mg/L. The use of renin-angiotensin system inhibitors, antiplatelets, and lipid-lowering therapy was associated with lower odds. Over a median of 6.4 years, compared with C-reactive protein < 2 mg/L, patients with C-reactive protein ≥ 2 mg/L had higher rates of hospitalizations, days spent in hospital, outpatient consultations, and dispensed medications (P < .05 for all). They also had a higher rate of major adverse cardiovascular events [hazard ratio (HR) 1.30; 95% confidence interval (CI) 1.27-1.33], heart failure (HR 1.24; 95% CI 1.20-1.30), and death (HR 1.35; 95% CI 1.31-1.39). Results were consistent across subgroups and granular C-reactive protein categories and robust to the exclusion of extreme C-reactive protein values or early events. CONCLUSIONS Three in five adults with ASCVD have systemic inflammation, which is associated with excess healthcare resource utilization and increased rates of cardiovascular events and death.
Collapse
Affiliation(s)
- Faizan Mazhar
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
| | - Anne-Laure Faucon
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
| | - Edouard L Fu
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Karolina E Szummer
- Department of Cardiology, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | | | | | | | - Nikolaus Marx
- Department of Internal Medicine I, RWTH Aachen University, Aachen, Germany
| | - Roxana Mehran
- Mount Sinai School of Medicine, Mount Sinai Health System, New York City, NY, USA
| | - Juan-Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
- Division of Nephrology, Department of Clinical Sciences, Danderyd Hospital, Danderyd, Sweden
| |
Collapse
|
22
|
McEvoy JW, McCarthy CP, Bruno RM, Brouwers S, Canavan MD, Ceconi C, Christodorescu RM, Daskalopoulou SS, Ferro CJ, Gerdts E, Hanssen H, Harris J, Lauder L, McManus RJ, Molloy GJ, Rahimi K, Regitz-Zagrosek V, Rossi GP, Sandset EC, Scheenaerts B, Staessen JA, Uchmanowicz I, Volterrani M, Touyz RM. 2024 ESC Guidelines for the management of elevated blood pressure and hypertension. Eur Heart J 2024; 45:3912-4018. [PMID: 39210715 DOI: 10.1093/eurheartj/ehae178] [Citation(s) in RCA: 322] [Impact Index Per Article: 322.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
|
23
|
Kwon OC, Lee HS, Jeon SY, Park MC. Incidence rate of recurrent cardiovascular events in patients with radiographic axial spondyloarthritis and the effect of tumor necrosis factor inhibitors. Arthritis Res Ther 2024; 26:174. [PMID: 39367448 PMCID: PMC11451105 DOI: 10.1186/s13075-024-03405-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Patients with radiographic axial spondyloarthritis (r-axSpA) are at increased risk of incident cardiovascular events. Tumor necrosis factor inhibitors (TNFi) have shown a protective effect against incident cardiovacular events. However, the incidence of recurrent cardiovascular events in patients with r-axSpA with a history of cardiovascular events, and the effect of TNFi on recurrent cardiovascular events remain unclear. We aimed to assess the incidence rate of recurrent cardiovascular events in patients with r-axSpA with a history of cardiovascular events and evaluate the effect of TNFi on the risk of recurrent cardiovascular events. METHODS This nationwide cohort study used data from the Korean National Claims Database. Data of patients with r-axSpA who had a history of cardiovascular events after being diagnosed with r-axSpA were extracted from the database. The outcome of interest was the recurrence of cardiovascular events (myocardial infarction or stroke). Patients were followed from the index date (date of the first cardiovascular event) to the date of cardiovascular event recurrence, the last date with claims data, or December 31, 2021, whichever occured first. The incidence rate of recurrent cardiovascular events was calculated. An inverse probability weighted Cox model was used to assess the effect of TNFi exposure on the risk of recurrent cardiovascular events. RESULTS This study included 413 patients (TNFi non-exposure, n = 338; TNFi exposure, n = 75). The incidence rate of recurrent cardiovascular events was 32 (95% confidence interval [CI] 22-42) per 1,000 person-years (TNFi non-exposure, 36 [95% CI 24-48] per 1,000 person-years; TNFi exposure, 19 [95% CI 2-35] per 1,000 person-years). In the inverse probability weighted Cox model, TNFi exposure was significantly associated with a lower risk of recurrent cardiovascular events (hazard ratio 0.33, 95% CI 0.12-0.94). CONCLUSIONS The incidence rate of recurrent cardiovascular events in patients with r-axSpA is substantial. TNFi exposure was associated with a lower risk of recurrent cardiovascular events.
Collapse
Affiliation(s)
- Oh Chan Kwon
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, South Korea
| | - So Young Jeon
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, South Korea
| | - Min-Chan Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea.
- Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonjuro, Gangnam-gu, Seoul, 06273, South Korea.
| |
Collapse
|
24
|
Mazzolai L, Teixido-Tura G, Lanzi S, Boc V, Bossone E, Brodmann M, Bura-Rivière A, De Backer J, Deglise S, Della Corte A, Heiss C, Kałużna-Oleksy M, Kurpas D, McEniery CM, Mirault T, Pasquet AA, Pitcher A, Schaubroeck HAI, Schlager O, Sirnes PA, Sprynger MG, Stabile E, Steinbach F, Thielmann M, van Kimmenade RRJ, Venermo M, Rodriguez-Palomares JF. 2024 ESC Guidelines for the management of peripheral arterial and aortic diseases. Eur Heart J 2024; 45:3538-3700. [PMID: 39210722 DOI: 10.1093/eurheartj/ehae179] [Citation(s) in RCA: 134] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
|
25
|
Yi J, Qu C, Li X, Gao H. Insulin resistance assessed by estimated glucose disposal rate and risk of atherosclerotic cardiovascular diseases incidence: the multi-ethnic study of atherosclerosis. Cardiovasc Diabetol 2024; 23:349. [PMID: 39342205 PMCID: PMC11439291 DOI: 10.1186/s12933-024-02437-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND To investigate the relationship between estimated glucose disposal rate (eGDR), a surrogate indicator of insulin resistance, and atherosclerotic cardiovascular diseases (ASCVD) incidence risk. METHODS This prospective cohort study utilized data from the 6026 participants from the Multi-Ethnic Study of Atherosclerosis. The eGDR (mg/kg/min) was computed as 21.158 - (0.09 × waist circumference [cm]) - (3.407 × hypertension [yes/no]) - (0.551 × HbA1c [%]). The population was categorized into four subgroups according to the quartiles (Q) of eGDR. Cox proportional hazard models were applied to assess the associations between eGDR and ASCVD incidence, and restricted cubic spine (RCS) was employed to examine the dose-response relationship. RESULTS The mean age of participants was 63.6 ± 10.1 years, comprising 3163 (52.5%) women. Over a median follow-up duration of 14.1 years, 565 (9.4%) developed ASCVD, including 256 (4.2%) myocardial infarctions, 234 (3.9%) strokes, and 358 (5.9%) fatal coronary heart disease. Compared to the lowest quartile, the adjusted hazard ratios (95% confidence intervals) for incident ASCVD for Q2-Q4 were 0.87 (0.68-1.10), 0.63 (0.47-0.84), and 0.43 (0.30-0.64), respectively. Per 1 standard deviation increase in eGDR was associated with a 30% (HR: 0.70, 95% CI 0.60-0.80) risk reduction of ASCVD, with the subgroup analyses indicating that age and hypertension modified the association (P for interaction < 0.05). RCS analysis indicated a significant and linear relationship between eGDR and ASCVD incidence risk. CONCLUSION eGDR level was negatively associated with incident ASCVD risk in a linear fashion among the general population. Our findings may contribute to preventive measures by improving ASCVD risk assessment.
Collapse
Affiliation(s)
- Jiayi Yi
- Center for Coronary Heart Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chao Qu
- Center for Coronary Heart Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiang Li
- Center for Coronary Heart Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hai Gao
- Center for Coronary Heart Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Gupte TP, Azizi Z, Kho PF, Zhou J, Chen ML, Panyard DJ, Guarischi-Sousa R, Hilliard AT, Sharma D, Watson K, Abbasi F, Tsao PS, Clarke SL, Assimes TL. A plasma proteomic signature for atherosclerotic cardiovascular disease risk prediction in the UK Biobank cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.13.24313652. [PMID: 39314942 PMCID: PMC11419231 DOI: 10.1101/2024.09.13.24313652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Background While risk stratification for atherosclerotic cardiovascular disease (ASCVD) is essential for primary prevention, current clinical risk algorithms demonstrate variability and leave room for further improvement. The plasma proteome holds promise as a future diagnostic and prognostic tool that can accurately reflect complex human traits and disease processes. We assessed the ability of plasma proteins to predict ASCVD. Method Clinical, genetic, and high-throughput plasma proteomic data were analyzed for association with ASCVD in a cohort of 41,650 UK Biobank participants. Selected features for analysis included clinical variables such as a UK-based cardiovascular clinical risk score (QRISK3) and lipid levels, 36 polygenic risk scores (PRSs), and Olink protein expression data of 2,920 proteins. We used least absolute shrinkage and selection operator (LASSO) regression to select features and compared area under the curve (AUC) statistics between data types. Randomized LASSO regression with a stability selection algorithm identified a smaller set of more robustly associated proteins. The benefit of plasma proteins over standard clinical variables, the QRISK3 score, and PRSs was evaluated through the derivation of Δ AUC values. We also assessed the incremental gain in model performance using proteomic datasets with varying numbers of proteins. To identify potential causal proteins for ASCVD, we conducted a two-sample Mendelian randomization (MR) analysis. Result The mean age of our cohort was 56.0 years, 60.3% were female, and 9.8% developed incident ASCVD over a median follow-up of 6.9 years. A protein-only LASSO model selected 294 proteins and returned an AUC of 0.723 (95% CI 0.708-0.737). A clinical variable and PRS-only LASSO model selected 4 clinical variables and 20 PRSs and achieved an AUC of 0.726 (95% CI 0.712-0.741). The addition of the full proteomic dataset to clinical variables and PRSs resulted in a Δ AUC of 0.010 (95% CI 0.003-0.018). Fifteen proteins selected by a stability selection algorithm offered improvement in ASCVD prediction over the QRISK3 risk score [Δ AUC: 0.013 (95% CI 0.005-0.021)]. Filtered and clustered versions of the full proteomic dataset (consisting of 600-1,500 proteins) performed comparably to the full dataset for ASCVD prediction. Using MR, we identified 11 proteins as potentially causal for ASCVD. Conclusion A plasma proteomic signature performs well for incident ASCVD prediction but only modestly improves prediction over clinical and genetic factors. Further studies are warranted to better elucidate the clinical utility of this signature in predicting the risk of ASCVD over the standard practice of using the QRISK3 score.
Collapse
Affiliation(s)
- Trisha P. Gupte
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Zahra Azizi
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Pik Fang Kho
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jiayan Zhou
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ming-Li Chen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel J. Panyard
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Rodrigo Guarischi-Sousa
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research (PAVIR), Stanford, CA, USA
| | - Austin T. Hilliard
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research (PAVIR), Stanford, CA, USA
| | - Disha Sharma
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathleen Watson
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Fahim Abbasi
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Philip S. Tsao
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Shoa L. Clarke
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Themistocles L. Assimes
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
27
|
Deshotels MR, Kotta PA, Rico Mesa JS, Oyenubi OA, Nambi V. When Does Primary Prevention Encroach on Secondary Prevention? Curr Atheroscler Rep 2024; 26:511-519. [PMID: 38976221 DOI: 10.1007/s11883-024-01227-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 07/09/2024]
Abstract
PURPOSE OF REVIEW The risk of incident atherosclerotic cardiovascular disease (ASCVD) in primary prevention is typically lower than in secondary prevention. However, there is a spectrum of risk among individuals undergoing primary prevention with the risk in some individuals approaching those of secondary prevention. We review the clinical conditions wherein the risk in primary prevention is similar to that observed in secondary prevention. RECENT FINDINGS Among individuals without established ASCVD, coronary artery calcium (CAC) scores ≥ 300 AU are associated with ASCVD event rates similar to secondary prevention populations. CAC score ≥ 1,000 AU are associated with an ASCVD risk seen in very high-risk secondary prevention populations. Interpretation of these observations must however consider differences in the risk reduction strategies. Current guidelines dichotomize ASCVD prevention into primary and secondary prevention, but certain primary prevention patients have an ASCVD risk equivalent to that of secondary prevention populations. Identifying higher risk primary prevention populations will allow for better risk mitigation strategies.
Collapse
Affiliation(s)
| | | | | | | | - Vijay Nambi
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Section of Cardiology and Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA.
- Michael E DeBakey Veterans Affairs Medical Center, Section of Cardiology, Houston, TX, USA.
- Center for Cardiovascular Disease Prevention (Clinic), 6655 Travis Street, Suite 320, Houston, TX, 77030, USA.
| |
Collapse
|
28
|
Duarte CK, Silva LDA, Andrade PMBD, Martins TMM, Ghisi GLDM. Barriers and facilitators to nutritional recommendations identified by participants of a cardiovascular rehabilitation program in a low resource context in Brazil. Nutrition 2024; 124:112451. [PMID: 38678640 DOI: 10.1016/j.nut.2024.112451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/24/2024] [Accepted: 04/01/2024] [Indexed: 05/01/2024]
Abstract
OBJECTIVES Nutritional recommendations, a core component of cardiovascular rehabilitation, play a vital role in managing cardiovascular diseases. However, adherence to these recommendations is complex, particularly in low-resource settings. This study explored the barriers and facilitators influencing adherence to nutritional recommendations among participants in a low-resource cardiovascular rehabilitation program in Brazil. METHODS A mixed-methods approach was employed. Sociodemographic data, the Mediterranean diet score, scale for assessing nutrition, and open-ended questions on adherence were collected. Those who completed the questionnaires (phase 1) were invited to participate in one focus group session (phase 2). The participants were characterized according to the responses provided in phase 1 (Mediterranean diet score and scale for assessing nutrition) in low adherence or high adherence to dietary practice. Descriptive statistics and thematic content analysis within the context of the theory of planned behavior were employed. RESULTS Seventy-four participants completed phase 1, with 41.9% classified into low adherence and 27.0% in high adherence; of those, 17 participated in phase 2. Focus group findings revealed 9 themes/29 subthemes. Barriers included food prices, income, knowledge, routine, food access, family patterns, disease, work, anxiety, eating habits, and food planning. Facilitators included affordable food, health considerations, taste preferences, knowledge, family/professional support, government assistance, personal willpower, income stability, easy food access, media influence, and a quiet eating place. CONCLUSIONS The study findings underscore the need for targeted interventions, including individualized meal planning, community engagement, and enhanced access to healthcare professionals, to optimize dietary adherence and improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Camila Kümmel Duarte
- Nutrition and Health Post-graduation program, Department of Nutrition, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana de Abreu Silva
- Nutrition and Health Post-graduation program, Department of Nutrition, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Gabriela Lima de Melo Ghisi
- KITE Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Physical Therapy, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
29
|
Xu L, Wang Y, Wang Y, Wang L, Du P, Cheng J, Zhang C, Jiao T, Xing L, Tapu MSR, Jia H, Li J. Early Use of PCSK9 Inhibitors in the Prognosis of Patients with Acute Coronary Syndrome by Protecting Vascular Endothelial Function. Pharmacology 2024; 110:1-14. [PMID: 38964284 DOI: 10.1159/000540083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) has a protective effect on acute coronary syndrome (ACS). However, most studies have shown that this protective effect is based on a decrease in low-density lipoprotein cholesterol, while other mechanisms remain limited. This study aimed to determine whether PCSK9i can improve the prognosis of ACS patients by protecting endothelial function. METHODS A total of 113 ACS patients were enrolled and randomly assigned to PCSK9i group (PCSK9i combined with statins) and control group (statins only). Blood lipids and endothelial function indicators were measured and analyzed 6 weeks before and after treatment. The effect of PCSK9i on the expression and secretion of endothelial function indicators in vascular endothelial cells were studied by cell experiments. RESULTS After 6 weeks of treatment, endothelial function indicators such as nitric oxide (NO), thrombomodulin, intercellular cell adhesion molecule-1, endothelin-1, and flow-mediated vasodilation were significantly improved in PCSK9i group compared with control group. Only the changes of NO and von Willebrand factor were associated with blood lipid levels, whereas the changes of other endothelial function indicators were not significantly associated with blood lipid levels. PCSK9i reduced the incidence of major adverse cardiovascular events in patients with ACS compared to those in the control group. In cell experiments, PCSK9i treatment significantly ameliorated LPS induced endothelial injury in HUVECs. CONCLUSION PCSK9i can protect vascular endothelial function partly independently of its lipid-lowering effect and ameliorate the prognosis of patients with ACS within 6 weeks. This mechanism may involve heat shock transcription factor 1/heat shock proteins -related signaling pathways. Early use of PCSK9i in patients with ACS should be strongly considered in clinical practice.
Collapse
Affiliation(s)
- Linghao Xu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanqi Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yiqiong Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liang Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peizhao Du
- Department of Cardiology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Cheng
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunsheng Zhang
- Department of Cardiology, East Hospital of Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Tiantian Jiao
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lijian Xing
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Md Sakibur Rahman Tapu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haonan Jia
- Laboratory of Molecular Neural Biology, School of Life Sciences and Institute of Systems Biology, Shanghai University, Shanghai, China
| | - Jiming Li
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
30
|
Wang P, Yuan D, Zhao X, Zhu P, Guo X, Jiang L, Xu N, Wang Z, Liu R, Wang Q, Chen Y, Zhang Y, Xu J, Liu Z, Song Y, Zhang Z, Yao Y, Feng Y, Tang X, Wang X, Gao R, Han Y, Yuan J. Inverse Association of Lipoprotein(a) on Long-Term Bleeding Risk in Patients with Coronary Heart Disease: Insight from a Multicenter Cohort in Asia. Thromb Haemost 2024; 124:684-694. [PMID: 37487540 PMCID: PMC11199048 DOI: 10.1055/s-0043-1771188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/08/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Lipoprotein(a), or Lp(a), has been recognized as a strong risk factor for atherosclerotic cardiovascular disease. However, the relationship between Lp(a) and bleeding remains indistinct, especially in the secondary prevention population of coronary artery disease (CAD). This investigation aimed to evaluate the association of Lp(a) with long-term bleeding among patients with CAD. METHODS Based on a prospective multicenter cohort of patients with CAD consecutively enrolled from January 2015 to May 2019 in China, the current analysis included 16,150 participants. Thus, according to Lp(a) quintiles, all subjects were divided into five groups. The primary endpoint was bleeding at 2-year follow-up, and the secondary endpoint was major bleeding at 2-year follow-up. RESULTS A total of 2,747 (17.0%) bleeding and 525 (3.3%) major bleeding were recorded during a median follow-up of 2.0 years. Kaplan-Meier survival analysis showed the highest bleeding incidence in Lp(a) quintile 1, compared with patients in Lp(a) quintiles 2 to 5 (p < 0.001), while the incidence of major bleeding seemed similar between the two groups. Moreover, restricted cubic spline analysis suggested that there was an L-shaped association between Lp(a) and 2-year bleeding after adjustment for potential confounding factors, whereas there was no significant association between Lp(a) and 2-year major bleeding. CONCLUSION There was an inverse and L-shaped association of Lp(a) with bleeding at 2-year follow-up in patients with CAD. More attention and effort should be made to increase the clinician awareness of Lp(a)'s role, as a novel marker for bleeding risk to better guide shared-decision making in clinical practice.
Collapse
Affiliation(s)
- Peizhi Wang
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Deshan Yuan
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xueyan Zhao
- Special Demand Medical Care Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Zhu
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Lin Jiang
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Xu
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhifang Wang
- Department of Cardiology, Xinxiang Central Hospital, Xinxiang, Henan Province, China
| | - Ru Liu
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qingsheng Wang
- Department of Cardiology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei Province, China
| | - Yan Chen
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongzhen Zhang
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Jingjing Xu
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenyu Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Song
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Zhang
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yi Yao
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingqing Feng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou, Guangdong Province, China
| | - Xiaofang Tang
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaozeng Wang
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning Province, China
| | - Runlin Gao
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaling Han
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning Province, China
| | - Jinqing Yuan
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Kinoshita D, Suzuki K, Yuki H, Niida T, Fujimoto D, Minami Y, Dey D, Lee H, McNulty I, Ako J, Ferencik M, Kakuta T, Ye JC, Jang IK. Coronary plaque phenotype associated with positive remodeling. J Cardiovasc Comput Tomogr 2024; 18:401-407. [PMID: 38677958 DOI: 10.1016/j.jcct.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/04/2024] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Positive remodeling is an integral part of the vascular adaptation process during the development of atherosclerosis, which can be detected by coronary computed tomography angiography (CTA). METHODS A total of 426 patients who underwent both coronary CTA and optical coherence tomography (OCT) were included. Four machine learning (ML) models, gradient boosting machine (GBM), random forest (RF), deep learning (DL), and support vector machine (SVM), were employed to detect specific plaque features. A total of 15 plaque features assessed by OCT were analyzed. The variable importance ranking was used to identify the features most closely associated with positive remodeling. RESULTS In the variable importance ranking, lipid index and maximal calcification arc were consistently ranked high across all four ML models. Lipid index and maximal calcification arc were correlated with positive remodeling, showing pronounced influence at the lower range and diminishing influence at the higher range. Patients with more plaques with positive remodeling throughout their entire coronary trees had higher low-density lipoprotein cholesterol levels and were associated with a higher incidence of cardiovascular events during 5-year follow-up (Hazard ratio 2.10 [1.26-3.48], P = 0.004). CONCLUSION Greater lipid accumulation and less calcium burden were important features associated with positive remodeling in the coronary arteries. The number of coronary plaques with positive remodeling was associated with a higher incidence of cardiovascular events.
Collapse
Affiliation(s)
- Daisuke Kinoshita
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keishi Suzuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Haruhito Yuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Takayuki Niida
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daichi Fujimoto
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yoshiyasu Minami
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Iris McNulty
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Junya Ako
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Maros Ferencik
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Tsunekazu Kakuta
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan.
| | - Jong Chul Ye
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea; Kim Jaechul Graduate School of Artificial Intelligence, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Wang KL, Balmforth C, Meah MN, Daghem M, Moss AJ, Tzolos E, Kwiecinski J, Molek-Dziadosz P, Craig N, Bularga A, Adamson PD, Dawson DK, Arumugam P, Sabharwal NK, Greenwood JP, Townend JN, Calvert PA, Rudd JHF, Verjans JW, Berman DS, Slomka PJ, Dey D, Mills NL, van Beek EJR, Williams MC, Dweck MR, Newby DE. Coronary Atherosclerotic Plaque Activity and Risk of Myocardial Infarction. J Am Coll Cardiol 2024; 83:2135-2144. [PMID: 38811091 PMCID: PMC11254330 DOI: 10.1016/j.jacc.2024.03.419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Total coronary atherosclerotic plaque activity across the entire coronary arterial tree is associated with patient-level clinical outcomes. OBJECTIVES We aimed to investigate whether vessel-level coronary atherosclerotic plaque activity is associated with vessel-level myocardial infarction. METHODS In this secondary analysis of an international multicenter study of patients with recent myocardial infarction and multivessel coronary artery disease, we assessed vessel-level coronary atherosclerotic plaque activity using coronary 18F-sodium fluoride positron emission tomography to identify vessel-level myocardial infarction. RESULTS Increased 18F-sodium fluoride uptake was found in 679 of 2,094 coronary arteries and 414 of 691 patients. Myocardial infarction occurred in 24 (4%) vessels with increased coronary atherosclerotic plaque activity and in 25 (2%) vessels without increased coronary atherosclerotic plaque activity (HR: 2.08; 95% CI: 1.16-3.72; P = 0.013). This association was not demonstrable in those treated with coronary revascularization (HR: 1.02; 95% CI: 0.47-2.25) but was notable in untreated vessels (HR: 3.86; 95% CI: 1.63-9.10; Pinteraction = 0.024). Increased coronary atherosclerotic plaque activity in multiple coronary arteries was associated with heightened patient-level risk of cardiac death or myocardial infarction (HR: 2.43; 95% CI: 1.37-4.30; P = 0.002) as well as first (HR: 2.19; 95% CI: 1.18-4.06; P = 0.013) and total (HR: 2.50; 95% CI: 1.42-4.39; P = 0.002) myocardial infarctions. CONCLUSIONS In patients with recent myocardial infarction and multivessel coronary artery disease, coronary atherosclerotic plaque activity prognosticates individual coronary arteries and patients at risk for myocardial infarction.
Collapse
Affiliation(s)
- Kang-Ling Wang
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; General Clinical Research Center, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Craig Balmforth
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Mohammed N Meah
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Marwa Daghem
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Alastair J Moss
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Evangelos Tzolos
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Jacek Kwiecinski
- Department of Interventional Cardiology and Angiology, Institute of Cardiology, Warsaw, Poland
| | - Patrycja Molek-Dziadosz
- Department of Coronary Artery Disease and Heart Failure, John Paul II Hospital, Kraków, Poland
| | - Neil Craig
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Anda Bularga
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Philip D Adamson
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Dana K Dawson
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Aberdeen, United Kingdom
| | - Parthiban Arumugam
- Manchester University National Health Service (NHS) Foundation Trust, Manchester, United Kingdom
| | - Nikant K Sabharwal
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - John P Greenwood
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom; Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom; The Baker Heart and Diabetes Institute, Monash University, and University of Melbourne, Melbourne, Victoria, Australia
| | - Jonathan N Townend
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Patrick A Calvert
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - James H F Rudd
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Johan W Verjans
- Australian Institute for Machine Learning, University of Adelaide, Adelaide, South Australia, Australia; Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | | | - Piotr J Slomka
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Damini Dey
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nicholas L Mills
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Edwin J R van Beek
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Michelle C Williams
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - David E Newby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
33
|
Sun W, Wang Y, Li C, Yao X, Wu X, He A, Zhao B, Huang X, Song H. Genetically predicted high serum sex hormone-binding globulin levels are associated with lower ischemic stroke risk: A sex-stratified Mendelian randomization study. J Stroke Cerebrovasc Dis 2024; 33:107686. [PMID: 38522757 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024] Open
Abstract
OBJECTIVE Cross-sectional and cohort studies have found insufficient evidence of a causal relationship between sex hormone-binding globulin and ischemic stroke, only associations. Here, we performed a sex-stratified, bidirectional, two-sample Mendelian randomization analysis to evaluate whether a causal relationship exists between sex hormone-binding globulin and ischemic stroke. METHODS Single-nucleotide polymorphisms associated with sex hormone-binding globulin and ischemic stroke were screened from genome-wide association studies summary data as instrumental variables to enable a bidirectional, two-sample Mendelian randomization study design. Inverse-variance weighted analysis was used as the main method to evaluate potential causality, and additional methods, including the weighted median and MR-Egger tests, were used to validate the Mendelian randomization results. Cochran's Q statistic, MR-Egger intercept test, and Mendelian Randomization-Pleiotropy Residual Sum and Outlier global test were used as sensitivity analysis techniques to assure the reliability of the results. Multivariable analysis was used to show the robustness of the results with key theorized confounders. RESULTS Inverse-variance weighted analysis showed that genetically predicted higher serum sex hormone-binding globulin levels were associated with significantly decreased risk of ischemic stroke in males (odds radio = 0.934, 95 % confidence interval = 0.885-0.985, P = 0.012) and females (odds radio = 0.924, 95 % confidence interval = 0.868-0.983, P = 0.013). In an analysis of ischemic stroke subtypes, genetically predicted higher serum sex hormone-binding globulin levels were also associated with significantly decreased risk of small-vessel occlusion in both males (odds radio = 0.849, 95 % confidence interval = 0.759-0.949, P = 0.004) and females (odds radio = 0.829, 95 % confidence interval = 0.724-0.949, P = 0.006). The association remained in sensitivity analyses and multivariable analyses. The reverse analysis suggested an association between genetically predicted risk of cardioembolism and increased serum sex hormone-binding globulin in females (Beta = 0.029 nmol/L, Standard Error = 0.010, P = 0.003). CONCLUSION Our findings provide new insight into the etiology of ischemic stroke and suggest that modulating serum sex hormone-binding globulin may be a therapeutic strategy to protect against ischemic stroke.
Collapse
Affiliation(s)
- Wei Sun
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Cancan Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xuefan Yao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiao Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Aini He
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Benke Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiaoqin Huang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Haiqing Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
34
|
Xu L, Wang L, Wang Y, Wang Y, Jiang Y, Du P, Cheng J, Zhang C, Wang R, Jiao T, Xing L, Ma J, Li J. PCSK9 inhibitors ameliorate arterial stiffness in ACS patients: evidences from Mendelian randomization, a retrospective study and basic experiments. Front Med (Lausanne) 2024; 11:1408760. [PMID: 38860206 PMCID: PMC11163136 DOI: 10.3389/fmed.2024.1408760] [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: 03/28/2024] [Accepted: 05/09/2024] [Indexed: 06/12/2024] Open
Abstract
Background Current evidences suggest that Proprotein Convertase Subtilisin/kexin Type 9 inhibitors (PCSK9i) exhibit a protective influence on acute coronary syndrome (ACS). Nevertheless, further investigation is required to comprehend the impact and mechanisms of these pharmaceutical agents on inflammatory factors and arterial stiffness (AS) in patients with ACS. Consequently, the objective of this study is to ascertain the influence of PCSK9i on arterial stiffness in ACS patients and elucidate the underlying mechanisms behind their actions. Methods This study employed Mendelian randomization (MR) analysis to examine the association between genetic prediction of PCSK9 inhibition and arterial stiffness. Data of 71 patients with ACS were retrospectively collected, including PCSK9i group (n = 36, PCSK9 inhibitors combined with statins) and control group (n = 35, statins only). Blood lipid levels, inflammatory markers and pulse wave velocity (PWV) data were collected before treatment and at 1 and 6 months after treatment for analysis. Additionally, cell experiments were conducted to investigate the impact of PCSK9i on osteogenesis of vascular smooth muscle cells (VSMCs), utilizing western blot (WB), enzyme-linked immunosorbent assay (ELISA), and calcification index measurements. Results The results of the MR analysis suggest that genetic prediction of PCSK9 inhibition has potential to reduce the PWV. Following treatment of statins combined with PCSK9 inhibitors for 1 and 6 months, the PCSK9i group exhibited significantly lower levels of total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), C-reactive protein (CRP), interleukin-6 (IL-6), fibrinogen (FIB) and procalcitonin (PCT) compared to the control group (p < 0.05). Additionally, PWV in the PCSK9i group demonstrated significant reduction after 6 months of treatment and was found to be associated with the circulating CRP level. In cell experiments, PCSK9i pretreatment ameliorated osteogenesis of VSMCs through reducing the deposition of calcium ions, alkaline phosphatase (ALP) activity, and expression of runt-related transcription factor 2 (RUNX2). Conclusion PCSK9i have potential to enhance arterial stiffness in ACS patients. Specifically, at the clinical level, this impact may be attributed to alterations in circulating CRP levels. At the cellular level, it is associated with the signaling pathway linked to RUNX2.
Collapse
Affiliation(s)
- Linghao Xu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liang Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanqi Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yiqiong Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanzhen Jiang
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Peizhao Du
- Department of Cardiology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Cheng
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunsheng Zhang
- Department of Cardiology, East Hospital of Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Ruijie Wang
- Department of Cardiology, Harbin Medical University First Affiliated Hospital, Harbin, China
| | - Tiantian Jiao
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lijian Xing
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiangping Ma
- School of Medicine, Tongji University, Shanghai, China
| | - Jiming Li
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
35
|
Nurmohamed NS, van Rosendael AR, Danad I, Ngo-Metzger Q, Taub PR, Ray KK, Figtree G, Bonaca MP, Hsia J, Rodriguez F, Sandhu AT, Nieman K, Earls JP, Hoffmann U, Bax JJ, Min JK, Maron DJ, Bhatt DL. Atherosclerosis evaluation and cardiovascular risk estimation using coronary computed tomography angiography. Eur Heart J 2024; 45:1783-1800. [PMID: 38606889 PMCID: PMC11129796 DOI: 10.1093/eurheartj/ehae190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/13/2024] [Accepted: 03/13/2024] [Indexed: 04/13/2024] Open
Abstract
Clinical risk scores based on traditional risk factors of atherosclerosis correlate imprecisely to an individual's complex pathophysiological predisposition to atherosclerosis and provide limited accuracy for predicting major adverse cardiovascular events (MACE). Over the past two decades, computed tomography scanners and techniques for coronary computed tomography angiography (CCTA) analysis have substantially improved, enabling more precise atherosclerotic plaque quantification and characterization. The accuracy of CCTA for quantifying stenosis and atherosclerosis has been validated in numerous multicentre studies and has shown consistent incremental prognostic value for MACE over the clinical risk spectrum in different populations. Serial CCTA studies have advanced our understanding of vascular biology and atherosclerotic disease progression. The direct disease visualization of CCTA has the potential to be used synergistically with indirect markers of risk to significantly improve prevention of MACE, pending large-scale randomized evaluation.
Collapse
Affiliation(s)
- Nick S Nurmohamed
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit
Amsterdam, Amsterdam, The
Netherlands
- Department of Vascular Medicine, Amsterdam UMC, University of
Amsterdam, Amsterdam, The
Netherlands
- Division of Cardiology, The George Washington University School of
Medicine, Washington, DC, United States
| | | | - Ibrahim Danad
- Department of Cardiology, University Medical Center Utrecht,
Utrecht, The Netherlands
- Department of Cardiology, Radboud University Medical Center,
Nijmegen, The Netherlands
| | - Quyen Ngo-Metzger
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson
School of Medicine, Pasadena, CA, United States
| | - Pam R Taub
- Section of Cardiology, Department of Medicine, University of
California, San Diego, CA, United States
| | - Kausik K Ray
- Department of Primary Care and Public Health, Imperial College
London, London, United
Kingdom
| | - Gemma Figtree
- Faculty of Medicine and Health, University of Sydney,
Australia, St Leonards, Australia
| | - Marc P Bonaca
- Department of Medicine, University of Colorado School of
Medicine, Aurora, CO, United States
| | - Judith Hsia
- Department of Medicine, University of Colorado School of
Medicine, Aurora, CO, United States
| | - Fatima Rodriguez
- Department of Medicine, Stanford University School of
Medicine, Stanford, CA, United States
| | - Alexander T Sandhu
- Department of Medicine, Stanford University School of
Medicine, Stanford, CA, United States
| | - Koen Nieman
- Department of Medicine, Stanford University School of
Medicine, Stanford, CA, United States
| | - James P Earls
- Cleerly, Inc., Denver, CO, United States
- Department of Radiology, The George Washington University School of
Medicine, Washington, DC, United States
| | | | - Jeroen J Bax
- Department of Cardiology, Leiden University Medical Center,
Leiden, The Netherlands
| | | | - David J Maron
- Department of Medicine, Stanford University School of
Medicine, Stanford, CA, United States
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount
Sinai, 1 Gustave Levy Place, Box 1030, New York, NY
10029, United States
| |
Collapse
|
36
|
Holtrop J, Bhatt DL, Ray KK, Mach F, Smulders YM, Carballo D, Steg PG, Visseren FLJ, Dorresteijn JAN. Impact of the 2021 European Society for Cardiology prevention guideline's stepwise approach for cardiovascular risk factor treatment in patients with established atherosclerotic cardiovascular disease. Eur J Prev Cardiol 2024; 31:754-762. [PMID: 38324720 DOI: 10.1093/eurjpc/zwae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/24/2024] [Accepted: 01/27/2024] [Indexed: 02/09/2024]
Abstract
AIMS This study aimed to evaluate the stepwise approach for cardiovascular (CV) risk factor treatment as outlined by the European Society for Cardiology 2021 guidelines on CV disease (CVD) prevention in patients with established atherosclerotic CVD (ASCVD). METHODS AND RESULTS In patients with ASCVD, included in UCC-SMART (n = 8730) and European parts of the REACH registry (n = 18 364), the 10-year CV risk was estimated using SMART2. Treatment effects were derived from meta-analyses and trials. Step 1 recommendations were LDL cholesterol (LDLc) < 1.8 mmol/L, systolic blood pressure (SBP) < 140 mmHg, using any antithrombotic medication, sodium-glucose co-transporter 2 (SGLT2) inhibition, and smoking cessation. Step 2 recommendations were LDLc < 1.4 mmol/L, SBP < 130 mmHg, dual-pathway inhibition (DPI, aspirin plus low-dose rivaroxaban), colchicine, glucagon-like peptide (GLP)-1 receptor agonists, and eicosapentaenoic acid. Step 2 was modelled accounting for Step 1 non-attainment. With current treatment, residual CV risk was 22%, 32%, and 60% in the low, moderate, and pooled (very) high European risk regions, respectively. Step 2 could prevent up to 198, 223 and 245 events per 1000 patients treated, respectively. Intensified LDLc reduction, colchicine, and DPI could be applied to most patients, preventing up to 57, 74, and 59 events per 1000 patients treated, respectively. Following Step 2, the number of patients with a CV risk of <10% could increase from 20%, 6.4%, and 0.5%, following Step 1, to 63%, 48%, and 12%, in the respective risk regions. CONCLUSION With current treatment, residual CV risk in patients with ASCVD remains high across all European risk regions. The intensified Step 2 treatment options result in marked further reduction of residual CV risk in patients with established ASCVD. KEY FINDINGS Guideline-recommended intensive treatment of patients with cardiovascular disease could prevent additional 198-245 new cardiovascular events for every 1000 patients treated.
Collapse
Affiliation(s)
- Joris Holtrop
- Department of Vascular Medicine, University Medical Centre Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai Health System, New York, NY, USA
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, ICTU-Global, Imperial College London, London, UK
| | - François Mach
- Division of Cardiology, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Yvo M Smulders
- Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - David Carballo
- Division of Cardiology, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Gabriel Steg
- Department of Cardiology, Université Paris-Cité, FACT (French Alliance for Cardiovascular Trials) NSERM1148/LVTS, AP-HP, Hôpital Bichat, Paris, France
| | - Frank L J Visseren
- Department of Vascular Medicine, University Medical Centre Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Jannick A N Dorresteijn
- Department of Vascular Medicine, University Medical Centre Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| |
Collapse
|
37
|
He J, Yang M, Song C, Zhang R, Yuan S, Li J, Dou K. Lipoprotein(a) is associated with recurrent cardiovascular events in patients with coronary artery disease and prediabetes or diabetes. J Endocrinol Invest 2024; 47:883-894. [PMID: 37777699 DOI: 10.1007/s40618-023-02203-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/17/2023] [Indexed: 10/02/2023]
Abstract
PURPOSE Elevated lipoprotein(a) [Lp(a)] and diabetes mellitus (DM) are both associated with adverse events in high-risk patients with established coronary artery disease (CAD). Currently, the association between Lp(a) levels and recurrent cardiovascular (CV) events (CVEs) remained undetermined in patients with different glucose status. Therefore, this study aimed to investigate the prognostic significance of Lp(a) levels for recurrent CVEs in high-risk CAD patients who suffered from first CVEs according to different glycemic metabolism. METHODS We recruited 5257 consecutive patients with prior CVEs and followed up for recurrent CVEs, including CV death, non-fatal myocardial infarction (MI), and non-fatal stroke. Patients were assigned to low, medium, and high groups according to Lp(a) levels and further stratified by glucose status. RESULTS During a median 37-month follow-up, 225 (4.28%) recurrent CVEs occurred. High Lp(a) was independently associated with recurrent CVEs [adjusted Hazard Ratio (HR), 1.57; 95% confidence interval (CI) 1.12-2.19; P = 0.008]. When participants were classified according to Lp(a) levels and glycemic status, high Lp(a) levels were associated with an increased risk of recurrent CVEs in pre-DM (adjusted HR, 2.96; 95% CI 1.24-7.05; P = 0.014). Meanwhile, medium and high Lp(a) levels were both associated with an increased risk for recurrent CVEs in DM (adjusted HR, 3.09; 95% CI 1.30-7.34; P = 0.010 and adjusted HR, 3.13, 95% CI 1.30-7.53; P = 0.011, respectively). CONCLUSIONS This study demonstrated that elevated Lp(a) levels were associated with an increased recurrent CVE risk in patients with CAD, particularly among those with pre-DM and DM, indicating that Lp(a) may provide incremental value in risk stratification in this population.
Collapse
Affiliation(s)
- J He
- State Key Laboratory of Cardiovascular Disease, Beijing, China
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
- Department of Cardiology, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - M Yang
- Department of Cardiology, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - C Song
- State Key Laboratory of Cardiovascular Disease, Beijing, China
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
- Department of Cardiology, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - R Zhang
- State Key Laboratory of Cardiovascular Disease, Beijing, China
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
- Department of Cardiology, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - S Yuan
- State Key Laboratory of Cardiovascular Disease, Beijing, China
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
- Department of Cardiology, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - J Li
- State Key Laboratory of Cardiovascular Disease, Beijing, China.
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China.
| | - K Dou
- State Key Laboratory of Cardiovascular Disease, Beijing, China.
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China.
- Department of Cardiology, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China.
| |
Collapse
|
38
|
Hoes LLF, Geleijnse JM, Bonekamp NE, Dorresteijn JAN, van der Meer MG, van der Schouw YT, Visseren FLJ, Koopal C. Prevalence and determinants of self-reported low-fat-, low-salt-, and vegetarian diets in patients with cardiovascular disease between 1996 and 2019. Nutr Metab Cardiovasc Dis 2024; 34:935-943. [PMID: 38403481 DOI: 10.1016/j.numecd.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/01/2023] [Accepted: 01/10/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND AND AIMS Guidelines no longer recommend low-fat diets and currently recommend more plant-based diets to reduce atherosclerotic cardiovascular disease (ASCVD) risk. Furthermore, these guidelines have consistently recommended salt-reduced diets. This article describes current self-reported use and time-trends in the self-reported use of low-fat, low-salt and vegetarian diets in ASCVD patients and examines patient characteristics associated with each diet. METHODS AND RESULTS 9005 patients with ASCVD included between 1996 and 2019 in the UCC-SMART cohort were studied. The prevalence of self-reported diets was assessed and multi-variable logistic regression was used to identify the determinants of each diet. Between 1996-1997 and 2018-2019, low-fat diets declined from 22.4 % to 3.8 %, and low-salt diets from 14.7 % to 4.6 %. The prevalence of vegetarian diets increased from 1.1 % in 1996-1997 to 2.3 % in 2018-2019. Patients with cerebrovascular disease (CeVD) and peripheral artery disease or an abdominal aortic aneurysm (PAD/AAA) were less likely to report a low-salt diet than coronary artery disease (CAD) patients (OR 0.62 [95%CI 0.49-0.77] and 0.55 [95%CI 0.41-0.72]). CONCLUSION In the period 1996 to 2019 amongst patients with ASCVD, the prevalence of self-reported low-fat diets was low and decreased in line with changes in recommendations in major guidelines. The prevalence of self-reported vegetarian diets was low but increased in line with societal and guideline changes. The prevalence of self-reported low-salt diets was low, especially in CeVD and PAD/AAA patients compared to CAD patients, and decreased over time. Renewed action is needed to promote low-salt diets in ASCVD patients.
Collapse
Affiliation(s)
- L L F Hoes
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - J M Geleijnse
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, the Netherlands
| | - N E Bonekamp
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - J A N Dorresteijn
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - M G van der Meer
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Frank L J Visseren
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| | - Charlotte Koopal
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
39
|
Hu Z, Cao X, Jing P, Zhang B, Shi Y, Siegrist J, Li J, Zhang M. Work stress and changes in heart rate variability among employees after first acute coronary syndrome: a hospital-based longitudinal cohort study. Front Public Health 2024; 12:1336065. [PMID: 38601505 PMCID: PMC11005455 DOI: 10.3389/fpubh.2024.1336065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/12/2024] [Indexed: 04/12/2024] Open
Abstract
Background Work stress is considered as a risk factor for coronary heart disease, but its link with heart rate variability (HRV) among heart attack survivors is unknown yet. The aim of this study was to investigate associations between baseline work stress and the changes of HRV over one-year after onset of acute coronary syndrome (ACS). Methods Hundred and twenty-two patients with regular paid work before their first ACS episode were recruited into this hospital-based longitudinal cohort study. During hospitalization (baseline), all patients underwent assessments of work stress by job strain (JS) and effort-reward imbalance (ERI) models, and were assigned into low or high groups; simultaneously, sociodemographic and clinical data, as well depression, anxiety, and job burnout, were collected. Patients were followed up 1, 6, and 12 months after discharge, with HRV measurements at baseline and each follow-up point. Generalized estimating equations were used to analyze the effects of baseline work stress on HRV over the following 1 year. Results After adjusting for baseline characteristics and clinical data, anxiety, depression, and burnout scores, high JS was not associated with any HRV measures during follow-up (all p > 0.10), whereas high ERI was significantly related to slower recovery of 5 frequency domain HRV measures (TP, HF, LF, VLF, and ULF) (all p < 0.001), and marginally associated with one time domain measure (SDNN) (p = 0.069). When mutually adjusting for both work stress models, results of ERI remained nearly unchanged. Conclusion Work stress in terms of ERI predicted lower HRV during the one-year period after ACS, especially frequency domain measures.
Collapse
Affiliation(s)
- Zhao Hu
- Cardiology Department, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingyu Cao
- Cardiology Department, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Pan Jing
- Cardiology Department, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bangying Zhang
- Cardiology Department, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunke Shi
- Cardiology Department, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Johannes Siegrist
- Institute of Medical Sociology, Centre for Health and Society, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jian Li
- Departments of Environmental Health Sciences and Epidemiology, Fielding School of Public Health, School of Nursing, University of California, Los Angeles, Los Angeles, CA, United States
| | - Min Zhang
- Cardiology Department, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
40
|
Castelijns MC, Hageman SHJ, Teraa M, van der Meer MG, Westerink J, Ten Berg J, Visseren FLJ. Generalisability of trials on antithrombotic treatment intensification in patients with cardiovascular disease. Heart 2024; 110:482-490. [PMID: 38182277 DOI: 10.1136/heartjnl-2023-323519] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024] Open
Abstract
OBJECTIVE Assessment of generalisability of guideline-informing trials on antithrombotic treatment intensification to real-world patients with cardiovascular disease (CVD). METHODS Inclusion and exclusion criteria of the Cardiovascular Outcomes for People Using Anticoagulation Strategies (COMPASS), Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management and Avoidance (CHARISMA), Prevention of Cardiovascular events in Patients with Prior Heart Attack Using Ticagrelor Compared to Placebo on a Background of Aspirin-Thrombolysis in Myocardial Infarction (PEGASUS-TIMI) and Dual Antiplatelet Therapy (DAPT) study were applied to coronary artery disease (CAD) and/or peripheral artery disease (PAD) patients from Utrecht Cardiovascular Cohort-Second Manifestations of Arterial Disease (UCC-SMART) to determine real-world eligibility. Eligible and ineligible patients were compared on baseline characteristics, cardiovascular events, major bleeding and mortality. RESULTS Eligibility ranged from 11%-94% for CAD to 75%-90% for patients with PAD. Cardiovascular, bleeding and mortality risks were higher in COMPASS-eligible patients with CAD (rate ratios (RR) 1.98 (95% CI 1.74 to 2.26), 2.02 (95% CI 1.47 to 2.78) and 3.11 (95% CI 2.71 to 3.57), respectively) and CHARISMA-eligible patients (RR 1.51 (95% CI 1.12 to 2.06), 2.25 (95% CI 1.01 to 6.21) and 4.43 (95% CI 2.79 to 7.51), respectively), and lower in COMPASS-eligible patients with PAD (RR 0.45 (95% CI 0.36 to 0.56), 0.29 (95% CI 0.18 to 0.46) and 0.45 (95% CI 0.38 to 0.54), respectively) and DAPT-eligible patients with CAD (RR CVD 0.49 (95% CI 0.34 to 0.69) and mortality 0.67 (95% CI 0.48 to 0.94)) than ineligible patients. After adjustment for trial eligibility criteria, only higher cardiovascular and mortality risks in COMPASS-eligible patients with CAD and lower cardiovascular risks in CHARISMA-eligible and DAPT-eligible patients persisted with CAD. CONCLUSION A large proportion of contemporary CVD patients would be eligible for intensified antithrombotic treatment trials, with mostly similar adjusted event risks to ineligible patients. Trial-based guideline recommendations are largely applicable to real-world patients.
Collapse
Affiliation(s)
- Maria C Castelijns
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Steven H J Hageman
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martin Teraa
- Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Manon G van der Meer
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan Westerink
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Internal Medicine, Isala Clinics Zwolle, Zwolle, The Netherlands
| | - Jurrien Ten Berg
- Department of Cardiology, Sint Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Frank L J Visseren
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
41
|
Zeng G, Zhu P, Yuan D, Wang P, Li T, Li Q, Xu J, Tang X, Song Y, Chen Y, Zhang C, Jia S, Liu R, Jiang L, Song L, Gao R, Yang Y, Zhao X, Yuan J. Renal function alters the association of lipoprotein(a) with cardiovascular outcomes in patients undergoing percutaneous coronary intervention: a prospective cohort study. Clin Kidney J 2024; 17:sfae032. [PMID: 38435350 PMCID: PMC10906361 DOI: 10.1093/ckj/sfae032] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Indexed: 03/05/2024] Open
Abstract
Background and hypothesis Lipoprotein(a) [Lp(a)] and renal dysfunction are both independent risk factors for cardiovascular disease. However, it remains unclear whether renal function mediates the association between Lp(a) and cardiovascular outcomes in patients undergoing percutaneous coronary intervention (PCI). Methods From a large prospective cohort study, 10 435 eligible patients undergoing PCI from January 2013 to December 2013 were included in our analysis. Patients were stratified into three renal function groups according to their baseline estimated glomerular filtration rate (eGFR) (<60; 60-90; ≥90 ml/min/1.73 m2). The primary endpoint was a composite of all-cause death, nonfatal MI, ischemic stroke, and unplanned revascularization [major adverse cardiac and cerebrovascular events (MACCE)]. Results Over a median follow-up of 5.1 years, a total of 2144 MACCE events occurred. After multivariable adjustment, either eGFR <60 ml/min/1.73 m2 or elevated Lp(a) conferred a significantly higher MACCE risk. Higher Lp(a) was significantly associated with an increased risk of MACCE in patients with eGFR <60 ml/min/1.73 m2. However, this association was weakened in subjects with only mild renal impairment and diminished in those with normal renal function. A significant interaction for MACCE between renal categories and Lp(a) was observed (P = 0.026). Patients with concomitant Lp(a) ≥30 mg/dl and eGFR <60 ml/min/1.73 m2 experienced worse cardiovascular outcomes compared with those without. Conclusion The significant association between Lp(a) and cardiovascular outcomes was mediated by renal function in patients undergoing PCI. Lp(a)-associated risk was more pronounced in patients with worse renal function, suggesting close monitoring and aggressive management are needed in this population.
Collapse
Affiliation(s)
- Guyu Zeng
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Zhu
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Deshan Yuan
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peizhi Wang
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianyu Li
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qinxue Li
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Xu
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaofang Tang
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Song
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Chen
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ce Zhang
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sida Jia
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ru Liu
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Jiang
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Song
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Runlin Gao
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xueyan Zhao
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinqing Yuan
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
42
|
Nurmohamed NS, Bom MJ, Jukema RA, de Groot RJ, Driessen RS, van Diemen PA, de Winter RW, Gaillard EL, Sprengers RW, Stroes ESG, Min JK, Earls JP, Cardoso R, Blankstein R, Danad I, Choi AD, Knaapen P. AI-Guided Quantitative Plaque Staging Predicts Long-Term Cardiovascular Outcomes in Patients at Risk for Atherosclerotic CVD. JACC Cardiovasc Imaging 2024; 17:269-280. [PMID: 37480907 DOI: 10.1016/j.jcmg.2023.05.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/17/2023] [Accepted: 05/30/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND The recent development of artificial intelligence-guided quantitative coronary computed tomography angiography analysis (AI-QCT) has enabled rapid analysis of atherosclerotic plaque burden and characteristics. OBJECTIVES This study set out to investigate the 10-year prognostic value of atherosclerotic burden derived from AI-QCT and to compare the spectrum of plaque to manually assessed coronary computed tomography angiography (CCTA), coronary artery calcium scoring (CACS), and clinical risk characteristics. METHODS This was a long-term follow-up study of 536 patients referred for suspected coronary artery disease. CCTA scans were analyzed with AI-QCT and plaque burden was classified with a plaque staging system (stage 0: 0% percentage atheroma volume [PAV]; stage 1: >0%-5% PAV; stage 2: >5%-15% PAV; stage 3: >15% PAV). The primary major adverse cardiac event (MACE) outcome was a composite of nonfatal myocardial infarction, nonfatal stroke, coronary revascularization, and all-cause mortality. RESULTS The mean age at baseline was 58.6 years and 297 patients (55%) were male. During a median follow-up of 10.3 years (IQR: 8.6-11.5 years), 114 patients (21%) experienced the primary outcome. Compared to stages 0 and 1, patients with stage 3 PAV and percentage of noncalcified plaque volume of >7.5% had a more than 3-fold (adjusted HR: 3.57; 95% CI 2.12-6.00; P < 0.001) and 4-fold (adjusted HR: 4.37; 95% CI: 2.51-7.62; P < 0.001) increased risk of MACE, respectively. Addition of AI-QCT improved a model with clinical risk factors and CACS at different time points during follow-up (10-year AUC: 0.82 [95% CI: 0.78-0.87] vs 0.73 [95% CI: 0.68-0.79]; P < 0.001; net reclassification improvement: 0.21 [95% CI: 0.09-0.38]). Furthermore, AI-QCT achieved an improved area under the curve compared to Coronary Artery Disease Reporting and Data System 2.0 (10-year AUC: 0.78; 95% CI: 0.73-0.83; P = 0.023) and manual QCT (10-year AUC: 0.78; 95% CI: 0.73-0.83; P = 0.040), although net reclassification improvement was modest (0.09 [95% CI: -0.02 to 0.29] and 0.04 [95% CI: -0.05 to 0.27], respectively). CONCLUSIONS Through 10-year follow-up, AI-QCT plaque staging showed important prognostic value for MACE and showed additional discriminatory value over clinical risk factors, CACS, and manual guideline-recommended CCTA assessment.
Collapse
Affiliation(s)
- Nick S Nurmohamed
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Division of Cardiology, The George Washington University School of Medicine, Washington, DC, USA. https://twitter.com/NickNurmohamed
| | - Michiel J Bom
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Ruurt A Jukema
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Robin J de Groot
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Roel S Driessen
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Pepijn A van Diemen
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Ruben W de Winter
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Emilie L Gaillard
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Ralf W Sprengers
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | | | - James P Earls
- Division of Cardiology, The George Washington University School of Medicine, Washington, DC, USA; Cleerly Inc, Denver, Colorado, USA
| | - Rhanderson Cardoso
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ron Blankstein
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ibrahim Danad
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Andrew D Choi
- Division of Cardiology, The George Washington University School of Medicine, Washington, DC, USA.
| | - Paul Knaapen
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
43
|
Groh LA, Willems LH, Fintelman P, Reijnen MMPJ, El Messaoudi S, Warlé MC. Dual-Pathway Inhibition with Rivaroxaban and Low-Dose Aspirin Does Not Alter Immune Cell Responsiveness and Distribution in Patients with Coronary Artery Disease. Cardiol Ther 2024; 13:233-242. [PMID: 38055176 PMCID: PMC10899137 DOI: 10.1007/s40119-023-00342-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023] Open
Abstract
INTRODUCTION Cardiovascular diseases (CVD) are the leading cause of death globally. Inflammation is an important driver of CVD where tissue damage may lead to the formation of deadly thrombi. Therefore, antithrombotic drugs, such as platelet inhibitors, are crucial for secondary risk prevention in coronary artery disease (CAD) and peripheral artery disease (PAD). For severe forms of the disease, dual-pathway inhibition (DPI) where low-dose aspirin is combined with rivaroxaban has shown improved efficacy in reducing cardiovascular mortality. METHODS Given this greater improvement in mortality, and the importance of inflammation in driving atherosclerosis, the potential for off-target inflammation-lowering effects of these drugs was evaluated by looking at the change in immune cell distribution and responsiveness to ex vivo lipopolysaccharide (LPS) stimulation after 3 months of DPI in patients with CAD. RESULTS We observed no changes in whole blood or peripheral blood mononuclear cell (PBMC) immune cell responsiveness to LPS after 3 months of DPI. Additionally, we did not observe any changes in the distribution of total white blood cells, monocytes, neutrophils, lymphocytes, or platelets during the study course. Signs of systemic inflammation were studied using Olink proteomics in 33 patients with PAD after 3 months of DPI. No changes were observed in any of the inflammatory proteins measured after the treatment period, suggesting that the state of chronic inflammation was not altered in these subjects. CONCLUSION Three months of DPI does not result in any meaningful change in immune cell responsiveness and distribution in patients with CAD or PAD. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT05210725.
Collapse
Affiliation(s)
- Laszlo A Groh
- Department of Surgery (Internal Address 618), Radboud University Medical Center, Postal Address 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Loes H Willems
- Department of Surgery (Internal Address 618), Radboud University Medical Center, Postal Address 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Paula Fintelman
- Department of Surgery (Internal Address 618), Radboud University Medical Center, Postal Address 9101, 6500 HB, Nijmegen, The Netherlands
| | - Michel M P J Reijnen
- Department of Surgery, Rijnstate Hospital, Arnhem, The Netherlands
- Multi-Modality Medical Imaging Group, University of Twente, Enschede, The Netherlands
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michiel C Warlé
- Department of Surgery (Internal Address 618), Radboud University Medical Center, Postal Address 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
44
|
Saba PS, Al Kindi S, Nasir K. Redefining Cardiovascular Risk Assessment as a Spectrum: From Binary to Continuous. J Am Coll Cardiol 2024; 83:574-576. [PMID: 38296401 DOI: 10.1016/j.jacc.2023.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 02/08/2024]
Affiliation(s)
- Pier Sergio Saba
- Clinical and Interventional Cardiology, Sassari University Hospital, Sassari, Italy.
| | - Sadeer Al Kindi
- Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas, USA. https://twitter.com/sadeer_alkindi
| | - Khurram Nasir
- Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas, USA. https://twitter.com/khurramn1
| |
Collapse
|
45
|
Vijayaraghavan K, Baum S, Desai NR, Voyce SJ. Intermediate and long-term residual cardiovascular risk in patients with established cardiovascular disease treated with statins. Front Cardiovasc Med 2024; 10:1308173. [PMID: 38288054 PMCID: PMC10822878 DOI: 10.3389/fcvm.2023.1308173] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
Introduction Statins remain the first-line treatment for secondary prevention of cardiovascular (CV) events, with lowering of low-density lipoprotein cholesterol (LDL-C) being their therapeutic target. Although LDL-C reduction significantly lowers CV risk, residual risk persists, even in patients with well-controlled LDL-C; thus, statin add-on agents that target pathways other than LDL-C, such as the omega-3 fatty acid eicosapentaenoic acid, may help to further reduce persistent CV risk in patients with established CV disease. Methods This narrative review examines the contemporary literature assessing intermediate- and long-term event rates in patients with established CV disease treated with statins. Results CV event rates among patients treated with statins who have established CV disease, including coronary artery disease, cerebrovascular disease, or peripheral arterial disease, accumulate over time, with a cumulative incidence of CV events reaching up to approximately 40% over 10 years. Recurrent stroke occurs in up to 19% of patients seven years after a first cerebrovascular event. Repeat revascularization and CV-related death occurs in up to 38% and 33% of patients with peripheral artery disease after three years, respectively. Discussion Additional treatment strategies, such as eicosapentaenoic acid, are needed to reduce persistent CV risk in patients with established CV disease treated with statins.
Collapse
Affiliation(s)
- K. Vijayaraghavan
- Division of Cardiology, University of Arizona College of Medicine, Phoenix, AZ, United States
| | - S. Baum
- Flourish Research, Boca Raton, FL, United States
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - N. R. Desai
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - S. J. Voyce
- Clinical Cardiology Research, Geisinger Heart Institute, Scranton, PA, United States
| |
Collapse
|
46
|
Yuan D, Xu N, Song Y, Zhang Z, Xu J, Liu Z, Tang X, Han Y, Chen Y, Zhang Y, Zhu P, Guo X, Wang Z, Liu R, Wang Q, Yao Y, Feng Y, Zhao X, Yuan J. Association Between Free Fatty Acids and Cardiometabolic Risk in Coronary Artery Disease: Results From the PROMISE Study. J Clin Endocrinol Metab 2023; 109:125-134. [PMID: 37540767 DOI: 10.1210/clinem/dgad416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Indexed: 08/06/2023]
Abstract
CONTEXT The association between free fatty acids (FFAs) and unfavorable clinical outcomes has been reported in the general population. However, evidence in the secondary prevention population is relatively scarce. OBJECTIVE We aimed to examine the relationship between FFA and cardiovascular risk in patients with coronary artery disease (CAD). METHODS This study was based on a multicenter cohort of patients with CAD enrolled from January 2015 to May 2019. The primary outcome was all-cause death. Secondary outcomes included cardiac death and major adverse cardiovascular events (MACE), a composite of death, myocardial infarction, and unplanned revascularization. RESULTS During a follow-up of 2 years, there were 468 (3.0%) all-cause deaths, 335 (2.1%) cardiac deaths, and 1279 (8.1%) MACE. Elevated FFA levels were independently associated with increased risks of all-cause death, cardiac death, and MACE (all P < .05). Moreover, When FFA were combined with an original model derived from the Cox regression, there were significant improvements in discrimination and reclassification for prediction of all-cause death (net reclassification improvement [NRI] 0.245, P < .001; integrated discrimination improvement [IDI] 0.004, P = .004), cardiac death (NRI 0.269, P < .001; IDI 0.003, P = .006), and MACE (NRI 0.268, P < .001; IDI 0.004, P < .001). Notably, when stratified by age, we found that the association between FFA with MACE risk appeared to be stronger in patients aged ≥60 years compared with those aged <60 years. CONCLUSION In patients with CAD, FFAs are associated with all-cause death, cardiac death, and MACE. Combined evaluation of FFAs with other traditional risk factors could help identify high-risk individuals who may require closer monitoring and aggressive treatment.
Collapse
Affiliation(s)
- Deshan Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Na Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Ying Song
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Zheng Zhang
- Department of Cardiology, the First Hospital of Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Jingjing Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Zhenyu Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Xiaofang Tang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Yaling Han
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang 110000, People's Republic of China
| | - Yan Chen
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Yongzhen Zhang
- Department of Cardiology, Peking University Third Hospital, Beijing 100037, People's Republic of China
| | - Pei Zhu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Xiaogang Guo
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine (FAHZU), Hangzhou 310000, People's Republic of China
| | - Zhifang Wang
- Department of Cardiology, Xinxiang Central Hospital, Xinxiang 453000, People's Republic of China
| | - Ru Liu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Qingsheng Wang
- Department of Cardiology, the First Hospital of Qinhuangdao, Qinhuangdao 066000, People's Republic of China
| | - Yi Yao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Yingqing Feng
- Department of Cardiology, Guangdong Provincial People's Hospital, Guangzhou 510000, People's Republic of China
| | - Xueyan Zhao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Jinqing Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| |
Collapse
|
47
|
Mensing LA, van Tuijl RJ, de Kort GA, van der Schaaf IC, Visseren FL, Rinkel GJE, Velthuis BK, Ruigrok YM. Screening for intracranial aneurysms in persons ⩾35 years with hypertension and atherosclerotic disease who smoke(d). Eur Stroke J 2023; 8:1071-1078. [PMID: 37585730 PMCID: PMC10683722 DOI: 10.1177/23969873231193296] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/21/2023] [Indexed: 08/18/2023] Open
Abstract
INTRODUCTION Lifetime risk of aneurysmal subarachnoid haemorrhage (aSAH) is high (7%) in persons ⩾35 years with hypertension who smoke(d). Whether screening for intracranial aneurysms (IAs) to prevent aSAH is effective in these patients is unknown. PATIENTS AND METHODS Participants were retrieved from a cohort of patients with clinically manifest atherosclerotic vascular disease included between 2012 and 2019 at the University Medical Centre Utrecht (SMART-ORACLE, NCT01932671) in whom CT-angiography (CTA) of intracranial arteries was performed. We selected patients ⩾35 years with hypertension who smoke(d). CTAs were reviewed for the presence of IAs by experienced neuroradiologists. Patients with IAs were offered follow-up imaging to detect aneurysmal growth. We determined aneurysm prevalence and developed a diagnostic model for IA risk at screening using multivariable logistic regression. RESULTS IA were found in 25 of 500 patients (5.0% prevalence, 95%CI: 3.3%-7.3%). Median 5 year risk of rupture assessed with the PHASES score was 0.9% (IQR: 0.7%-1.3%). During a median follow-up of 57 months (IQR: 39-83 months) no patients suffered from aSAH. Aneurysmal growth was detected in one patient for whom preventive treatment was advised. IA risk at screening ranged between 1.6% and 13.4% with predictors being age, female sex and current smoking. DISCUSSION AND CONCLUSION IA prevalence in persons ⩾35 years with hypertension and atherosclerotic vascular disease who smoke(d) was 5%. Given the very small proportion of IA that needed preventive treatment, we currently do not advise screening for Caucasian persons older than 35 years of age who smoke and have hypertension in general. Whether screening may be effective for certain subgroups (e.g. women older than 50 years of age) or other ethnic populations should be the subject of future studies.
Collapse
Affiliation(s)
- Liselore A Mensing
- UMC Utrecht Brain Centre, Department of Neurology and Neurosurgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Rick J van Tuijl
- Department of Radiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Gerard A de Kort
- Department of Radiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | - Frank L Visseren
- Department of Vascular Medicine, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Gabriel JE Rinkel
- UMC Utrecht Brain Centre, Department of Neurology and Neurosurgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Birgitta K Velthuis
- Department of Radiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Ynte M Ruigrok
- UMC Utrecht Brain Centre, Department of Neurology and Neurosurgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
48
|
Hageman SHJ, Dorresteijn JAN, Pennells L, van Smeden M, Bots ML, Di Angelantonio E, Visseren FLJ. The relevance of competing risk adjustment in cardiovascular risk prediction models for clinical practice. Eur J Prev Cardiol 2023; 30:1741-1747. [PMID: 37338108 DOI: 10.1093/eurjpc/zwad202] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/17/2023] [Accepted: 06/03/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND Many models developed for predicting the risk of cardiovascular disease (CVD) are adjusted for the competing risk of non-CVD mortality, which has been suggested to reduce potential overestimation of cumulative incidence in populations where the risk of competing events is high. The objective was to evaluate and illustrate the clinical impact of competing risk adjustment when deriving a CVD prediction model in a high-risk population. METHODS AND RESULTS Individuals with established atherosclerotic CVD were included from the Utrecht Cardiovascular Cohort-Secondary Manifestations of ARTerial disease (UCC-SMART). In 8355 individuals, followed for a median of 8.2 years (IQR 4.2-12.5), two similar prediction models for the estimation of 10-year residual CVD risk were derived: with competing risk adjustment using a Fine and Gray model and without competing risk adjustment using a Cox proportional hazards model. On average, predictions were higher from the Cox model. The Cox model predictions overestimated the cumulative incidence [predicted-observed ratio 1.14 (95% CI 1.09-1.20)], which was most apparent in the highest risk quartiles and in older persons. Discrimination of both models was similar. When determining treatment eligibility on thresholds of predicted risks, more individuals would be treated based on the Cox model predictions. If, for example, individuals with a predicted risk > 20% were considered eligible for treatment, 34% of the population would be treated according to the Fine and Gray model predictions and 44% according to the Cox model predictions. INTERPRETATION Individual predictions from the model unadjusted for competing risks were higher, reflecting the different interpretations of both models. For models aiming to accurately predict absolute risks, especially in high-risk populations, competing risk adjustment must be considered.
Collapse
Affiliation(s)
- Steven H J Hageman
- Department of Vascular Medicine, University Medical Centre Utrecht, Heidelberglaan 100, Postbus 85500 3508 GA Utrecht, The Netherlands
| | - Jannick A N Dorresteijn
- Department of Vascular Medicine, University Medical Centre Utrecht, Heidelberglaan 100, Postbus 85500 3508 GA Utrecht, The Netherlands
| | - Lisa Pennells
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Papworth Road, Trumpington, Cambridge CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Trumpington, Cambridge CB2 0BB, UK
| | - Maarten van Smeden
- Julius Centre for Health Science and Primary Care, University Medical Centre Utrecht, University of Utrecht, Heidelberglaan 100, Postbus 85500 3508 GA Utrecht, The Netherlands
| | - Michiel L Bots
- Julius Centre for Health Science and Primary Care, University Medical Centre Utrecht, University of Utrecht, Heidelberglaan 100, Postbus 85500 3508 GA Utrecht, The Netherlands
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Papworth Road, Trumpington, Cambridge CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Trumpington, Cambridge CB2 0BB, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, CB2 0BB Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, CB2 0BB Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, CB10 1SA Cambridge, UK
- Health Data Science Research Centre, Human Technopole, 20157 Milan, Italy
| | - Frank L J Visseren
- Department of Vascular Medicine, University Medical Centre Utrecht, Heidelberglaan 100, Postbus 85500 3508 GA Utrecht, The Netherlands
| |
Collapse
|
49
|
Delialis D, Georgiopoulos G, Aivalioti E, Konstantaki C, Oikonomou E, Bampatsias D, Mavraganis G, Vardavas C, Liberopoulos E, Stellos K, Stamatelopoulos K. Remnant cholesterol in atherosclerotic cardiovascular disease: A systematic review and meta-analysis. Hellenic J Cardiol 2023; 74:48-57. [PMID: 37116829 DOI: 10.1016/j.hjc.2023.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Accumulating evidence suggests a substantial contribution of remnant cholesterol (RC) to residual risk for the development or relapse of atherosclerotic cardiovascular disease (ASCVD). We aimed to evaluate the association of RC levels with ASCVD risk by different risk categories and methods of RC assessment. We also assessed available evidence of the effects of lipid-lowering therapies (LLTs) on RC levels. METHODS English-language searches of Medline, PubMed, and Embase (inception to 31 January 2023); ClinicalTrials.gov (October 2022); and reference lists of studies and reviews. Studies reporting on the risk of the composite endpoint [all-cause mortality, cardiovascular mortality, and major adverse cardiac events (MACE)] by RC levels were included. Moreover, we searched for studies reporting differences in RC levels after the administration of LLT(s). RESULTS Among n = 29 studies with 257,387 participants, we found a pooled linear (pooled HR: 1.27 per 1-SD increase, 95% CI: 1.12-1.43, P < 0.001, I2 = 95%, n = 15 studies) and non-linear association (pooled HR: 1.59 per quartile increase, 95% CI: 1.35-1.85, P < 0.001, I2 = 87.9%, n = 15 studies) of RC levels and the risk of M ACE both in patients with and without established ASCVD. Interestingly, the risk of MACE was higher in studies with directly measured vs. calculated RC levels. In a limited number of studies and participants, LLTs reduced RC levels. CONCLUSION RC levels are associated with ASCVD risk both in primary and secondary prevention. Directly measured RC levels are associated with ASCVD risk more evidently. Available LLTs tend to decrease RC levels, although the clinical relevance of RC decrease merits further investigation. PROSPERO REGISTRATION CRD42022371346.
Collapse
Affiliation(s)
- Dimitrios Delialis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece; School of Biomedical Engineering and Imaging Sciences, King's College, London, UK
| | - Evmorfia Aivalioti
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Christina Konstantaki
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ermioni Oikonomou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dimitrios Bampatsias
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Georgios Mavraganis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Constantine Vardavas
- Department of Social Medicine, Faculty of Medicine, University of Crete, University Campus of Voutes, 700 13, Heraklion, Crete, Greece; Center for Global Tobacco Control, Department of Society, Human Development and Health, Harvard School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - Evangelos Liberopoulos
- 1(st) Department of Propedeutic Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Konstantinos Stellos
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece; Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
50
|
Weber JE, Ahmadi M, Boldt LH, Eckardt KU, Edelmann F, Gerhardt H, Grittner U, Haubold K, Hübner N, Kollmus-Heege J, Landmesser U, Leistner DM, Mai K, Müller DN, Nolte CH, Pieske B, Piper SK, Rattan S, Rauch G, Schmidt S, Schmidt-Ott KM, Schönrath K, Schulz-Menger J, Schweizerhof O, Siegerink B, Spranger J, Ramachandran VS, Witzenrath M, Endres M, Pischon T. Protocol of the Berlin Long-term Observation of Vascular Events (BeLOVE): a prospective cohort study with deep phenotyping and long-term follow up of cardiovascular high-risk patients. BMJ Open 2023; 13:e076415. [PMID: 37907297 PMCID: PMC10618970 DOI: 10.1136/bmjopen-2023-076415] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/22/2023] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION The Berlin Long-term Observation of Vascular Events is a prospective cohort study that aims to improve prediction and disease-overarching mechanistic understanding of cardiovascular (CV) disease progression by comprehensively investigating a high-risk patient population with different organ manifestations. METHODS AND ANALYSIS A total of 8000 adult patients will be recruited who have either suffered an acute CV event (CVE) requiring hospitalisation or who have not experienced a recent acute CVE but are at high CV risk. An initial study examination is performed during the acute treatment phase of the index CVE or after inclusion into the chronic high risk arm. Deep phenotyping is then performed after ~90 days and includes assessments of the patient's medical history, health status and behaviour, cardiovascular, nutritional, metabolic, and anthropometric parameters, and patient-related outcome measures. Biospecimens are collected for analyses including 'OMICs' technologies (e.g., genomics, metabolomics, proteomics). Subcohorts undergo MRI of the brain, heart, lung and kidney, as well as more comprehensive metabolic, neurological and CV examinations. All participants are followed up for up to 10 years to assess clinical outcomes, primarily major adverse CVEs and patient-reported (value-based) outcomes. State-of-the-art clinical research methods, as well as emerging techniques from systems medicine and artificial intelligence, will be used to identify associations between patient characteristics, longitudinal changes and outcomes. ETHICS AND DISSEMINATION The study was approved by the Charité-Universitätsmedizin Berlin ethics committee (EA1/066/17). The results of the study will be disseminated through international peer-reviewed publications and congress presentations. STUDY REGISTRATION First study phase: Approved WHO primary register: German Clinical Trials Register: https://drks.de/search/de/trial/DRKS00016852; WHO International Clinical Registry Platform: http://apps.who.int/trialsearch/Trial2.aspx?TrialID=DRKS00016852. Recruitment started on July 18, 2017.Second study phase: Approved WHO primary register: German Clinical Trials Register DRKS00023323, date of registration: November 4, 2020, URL: http://www.drks.de/ DRKS00023323. Recruitment started on January 1, 2021.
Collapse
Affiliation(s)
- Joachim E Weber
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Stroke Research (CSB), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Michael Ahmadi
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Leif-Hendrik Boldt
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Internal Medicine and Cardiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Kai-Uwe Eckardt
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Frank Edelmann
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Internal Medicine and Cardiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Holger Gerhardt
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ulrike Grittner
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Kathrin Haubold
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Norbert Hübner
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Jil Kollmus-Heege
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Ulf Landmesser
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Cardiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Department for Cardiology, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - David M Leistner
- Department of Cardiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Knut Mai
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Endocrinology and Metabolism, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Dominik N Müller
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Christian H Nolte
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Stroke Research (CSB), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Burkert Pieske
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Internal Medicine and Cardiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Sophie K Piper
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Institute of Medical Informatics, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Simrit Rattan
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Geraldine Rauch
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Sein Schmidt
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Stroke Research (CSB), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Kai M Schmidt-Ott
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Katharina Schönrath
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Jeanette Schulz-Menger
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Oliver Schweizerhof
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Bob Siegerink
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Joachim Spranger
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Endocrinology and Metabolism, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Vasan S Ramachandran
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Sections of Preventive Medicine and Epidemiology, and Cardiovascular Medicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Martin Witzenrath
- Division of Pulmonary Inflammation, and Department of Infectious Diseases and Respiratory Medicine, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Germany
| | - Matthias Endres
- Department of Neurology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Stroke Research (CSB), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), partner site Berlin, Berlin, Germany
- ExellenceCluster NeuroCure, Berlin, Germany
| | - Tobias Pischon
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Biobank Technology Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| |
Collapse
|