1
|
Baratta F, Moscucci F, Lospinuso I, Cocomello N, Colantoni A, Di Costanzo A, Tramontano D, D'Erasmo L, Pastori D, Ettorre E, Del Ben M, Arca M, Desideri G. Lipid-Lowering Therapy and Cardiovascular Prevention in Elderly. Drugs 2025; 85:801-812. [PMID: 40338434 DOI: 10.1007/s40265-025-02182-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2025] [Indexed: 05/09/2025]
Abstract
The global population aged 80 years and older will reach approximately half a billion in the coming years, and cardiovascular prevention in this group of patients will become a global health challenge. In the era of evidence-based medicine, the use of lipid-lowering therapies (LLTs) in the elderly, particularly in primary and secondary cardiovascular prevention, remains an area of active research. Although there is broad consensus on the use of LLTs in the elderly to prevent recurrent cardiovascular events in secondary prevention, there is considerable debate about their use in primary prevention. Many efforts have been made to improve cardiovascular risk stratification in patients over 75 years of age in primary prevention. In recent years, some specific risk scores have been developed, including the Systematic Coronary Risk Evaluation 2 for Older Persons (SCORE2-OP). While there are very few specific warnings to consider for LLTs in the elderly, an important challenge in this patient population is to identify the turning point at which the disutility risk outweighs the potential benefits. However, despite the widespread recognition of the importance of this issue, there is a lack of guidance on how to identify patients who should be withdrawn from therapy. The aim of this narrative review is to examine the current state of knowledge regarding the indications for LLT in elderly patients, identify outstanding issues, and discuss future developments.
Collapse
Affiliation(s)
- Francesco Baratta
- Geriatric Unit, Department of Internal Medicine and Medical Specialties, AOU Policlinico Umberto I, Rome, Italy.
| | - Federica Moscucci
- Geriatric Unit, Department of Internal Medicine and Medical Specialties, AOU Policlinico Umberto I, Rome, Italy
| | - Ilaria Lospinuso
- Geriatric Unit, Department of Internal Medicine and Medical Specialties, AOU Policlinico Umberto I, Rome, Italy
| | - Nicholas Cocomello
- Department of Anatomical Sciences, Histological, Legal Medical and Locomotor, Sapienza University of Rome, Rome, Italy
- Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Alessandra Colantoni
- Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Alessia Di Costanzo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniele Tramontano
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Laura D'Erasmo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniele Pastori
- Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Evaristo Ettorre
- Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Maria Del Ben
- Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giovambattista Desideri
- Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
2
|
Jean-Gilles M, Gencer B. Therapeutic advances in the Lp(a) battle: what do we know and what are the most awaited novelties in the field? Curr Opin Lipidol 2025; 36:130-137. [PMID: 40127045 DOI: 10.1097/mol.0000000000000981] [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: 03/26/2025]
Abstract
PURPOSE OF REVIEW To review the latest advances in lipoprotein(a) [Lp(a)] treatment, focusing on the impact of currently available lipid-lowering therapies and highlighting the highly anticipated and most developed RNA-based therapies. RECENT FINDINGS Lp(a) is a key genetically determined cardiovascular risk modifier linked to myocardial infarction and calcific aortic stenosis development and progression. Conventional lipid-lowering therapies have no substantial effect on circulating Lp(a) levels, leading current guidelines to focus on managing traditional cardiovascular risk factors. New therapies, including antisense oligonucleotides and small interfering RNAs, target Lipoprotein(A) [LPA] gene translation to reduce apo(a) synthesis and Lp(a) particles formation. The most advanced candidates, pelacarsen, olpasiran, and lepodisiran, have shown promising Lp(a) reductions, ranging from -35% to -101% in Phase 1 and 2 trials. Phase 3 studies will clarify their effects on cardiovascular outcomes and address concerns about extremely low Lp(a) levels and safety. SUMMARY The RNA-based agents pelacarsen, olpasiran, and lepodisiran represent the most advanced developments in this field. Ongoing Phase 3 trials, expected to be finalized between 2025 and 2029, will be crucial in determining their efficacy in improving cardiovascular outcomes and their safety profiles.
Collapse
Affiliation(s)
| | - Baris Gencer
- Cardiology unit, Lausanne University Hospital, Lausanne
- Cardiology unit, Geneva University Hospitals, Genève, Switzerland
| |
Collapse
|
3
|
Kayzuka C, Rondon-Pereira VC, Nogueira Tavares C, Pacheco Pachado M, Monica FZ, Tanus-Santos JE, Lacchini R. Epigenetics is involved in the pleiotropic effects of statins. Expert Opin Drug Metab Toxicol 2025; 21:689-701. [PMID: 40208655 DOI: 10.1080/17425255.2025.2491732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 02/18/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025]
Abstract
INTRODUCTION Statins have significantly reduced mortality from cardiovascular diseases by lowering serum cholesterol levels. Beyond their lipid-lowering effects, statins improve vascular function, reduce inflammation, decrease reactive oxygen species (ROS) formation, and stabilize atherosclerotic plaques. However, the mechanisms underlying these pleiotropic effects remain unclear. AREA COVERED This narrative review summarizes and discusses epigenetic mechanisms that may explain part of the pleiotropic effects of statins. This approach allows for a reevaluation of statin use beyond its cholesterol-lowering benefits. A structured search was conducted in the PubMed and Scopus databases using specific search terms, including articles published up to August 2024. EXPERT OPINION The pleiotropic effects of statins, including those mediated by the isoprenoid pathway, partially explain their clinical benefits. By inhibiting histone deacetylases (HDACs, the 'erasers') and DNA methyltransferases (DNMTs, the 'writers'), statins promote increased histone acetylation and reduced DNA methylation at gene promoter regions. These epigenetic modifications enhance chromatin accessibility, facilitating gene transcription and protecting the cardiovascular system. Further investigation into these epigenetic mechanisms could support the repositioning of statins for broader therapeutic applications. Statins may have benefits extending beyond their role in managing hypercholesterolemia, as their pleiotropic effects contribute to the prevention of cardiovascular disease-related mortality through mechanisms independent of LDL cholesterol reduction.
Collapse
Affiliation(s)
- Cezar Kayzuka
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Cecilia Nogueira Tavares
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Mayra Pacheco Pachado
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Fabiola Zakia Monica
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Jose Eduardo Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
4
|
Caller T, Moore KJ, Lehmann LH, Wu SM, Leor J. Insights Into Heart-Tumor Interactions in Heart Failure. Circ Res 2025; 136:1262-1285. [PMID: 40403117 DOI: 10.1161/circresaha.124.325490] [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: 11/23/2024] [Revised: 02/10/2025] [Accepted: 02/27/2025] [Indexed: 05/24/2025]
Abstract
Heart failure (HF) often coexists with cancer. Beyond the known cardiotoxicity of some cancer treatments, HF itself has been associated with increased cancer incidence. The 2 conditions share common risk factors, mechanisms, and interactions that can worsen patient outcomes. The bidirectional relationship between HF and cancer presents a complex interplay of factors that are not fully understood. Recent preclinical evidence suggests that HF may promote tumor growth via the release of protumorigenic factors from the injured heart, revealing HF as a potentially protumorigenic condition. Our review discusses the biological crosstalk between HF and cancer, emphasizing the impact of HF on tumor growth, with inflammation, and modulating the immune system as central mechanisms. We further explore the clinical implications of this connection and propose future research directions. Understanding the mechanistic overlap and interactions between HF and cancer could lead to new biomarkers and therapies, addressing the growing prevalence of both conditions and enhancing approaches to diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Tal Caller
- Neufeld and Tamman Cardiovascular Research Institutes, Faculty of Medical and Health Sciences, Tel Aviv University, Israel (T.C., J.L.)
- Lev Leviev Cardiovascular and Thoracic Center, Sheba Medical Center, Tel Hashomer, Israel (T.C., J.L.)
| | - Kathryn J Moore
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine (K.J.M.)
| | - Lorenz H Lehmann
- Department of Cardiology, University Hospital Heidelberg, Germany (L.H.L.)
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg/Mannheim, Germany (L.H.L.)
- German Cancer Research Center (DKFZ), Heidelberg, Germany (L.H.L.)
| | - Sean M Wu
- Stanford Cardiovascular Institute (S.M.W.), Stanford University School of Medicine, CA
- Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA
| | - Jonathan Leor
- Neufeld and Tamman Cardiovascular Research Institutes, Faculty of Medical and Health Sciences, Tel Aviv University, Israel (T.C., J.L.)
- Lev Leviev Cardiovascular and Thoracic Center, Sheba Medical Center, Tel Hashomer, Israel (T.C., J.L.)
| |
Collapse
|
5
|
Goyal A, Maheshwari S, Mashkoor Y, Singh A, Rafique F, Sheikh AB, Bansal K. Impact of Periprocedural Statin Therapy on Mortality and Cardiovascular Outcomes in Transcatheter Aortic Valve Replacement: A Meta-Analysis and Meta-Regression. Catheter Cardiovasc Interv 2025. [PMID: 40391859 DOI: 10.1002/ccd.31607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/25/2025] [Accepted: 05/10/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Transcatheter aortic valve replacement (TAVR) stands as a notable alternative to surgical valve replacement for severe aortic stenosis (AS). Despite the established benefits of statins in cardiovascular pathologies, their specific impact in patients with severe AS undergoing TAVR remains uncertain. AIMS Our meta-analysis aims to assess whether periprocedural statin therapy improves survival and outcomes post-TAVR, thus addressing this gap in literature. METHODS A comprehensive literature search using various databases with relevant keywords terms was conducted to identify studies on the impact of periprocedural statin therapy on TAVR outcomes. We assessed the primary outcome of all-cause mortality alongside various secondary outcomes including stroke/transient ischemic attack (TIA), myocardial infarction, acute kidney injury (AKI), 30-day mortality, in-hospital mortality, rehospitalization, cardiovascular complications, and pacemaker requirement. A random-effects model using Comprehensive Meta Analysis Software was employed to analyze the data for each outcome. Statistical significance was set at a p < 0.05. RESULTS Our analysis of 19 observational studies revealed that periprocedural statin therapy significantly reduces all-cause mortality following TAVR surgery (OR = 0.71, 95% CI: 0.61-0.83, p < 0.001). However, the influence of statins on other outcomes remains inconclusive. These outcomes include stroke/TIA (OR = 0.90, 95% CI: 0.68-1.19, p = 0.455), risk of MI (OR = 1.72, 95% CI: 0.73-4.04, p = 0.214), AKI (OR = 0.99, 95% CI: 0.75-1.31, p = 0.968), 30-day mortality (OR = 0.71, 95% CI: 0.46-1.10, p = 0.126), in-hospital mortality (OR = 0.42, 95% CI: 0.13-1.38, p = 0.151), rehospitalization (OR = 0.92, 95% CI: 0.66-1.29, p = 0.645), cardiovascular complications (OR = 1.12, 95% CI: 0.91-1.37, p = 0.297), and pacemaker requirement (OR = 0.83, 95% CI: 0.65-1.06, p = 0.133). CONCLUSION Our meta-analysis indicates a potentially promising role for periprocedural statin therapy in enhancing patient outcomes post-TAVR surgery. We found a notable association between statin therapy and a reduction in all-cause mortality. However, the effects on secondary outcomes did not reach statistical significance, which warrants further investigation through larger, well-designed, randomized controlled trials.
Collapse
Affiliation(s)
- Aman Goyal
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Surabhi Maheshwari
- Department of Internal Medicine, University of Alabama at Montgomery, Montgomery, Alabama, USA
| | - Yusra Mashkoor
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Ajeet Singh
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Faryal Rafique
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Abu Baker Sheikh
- Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Kamna Bansal
- Department of Family and Community Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
6
|
Zimerman A, Kunzler ALF, Weber BN, Ran X, Murphy SA, Wang H, Honarpour N, Keech AC, Sever PS, Sabatine MS, Giugliano RP. Intensive Lowering of LDL Cholesterol Levels With Evolocumab in Autoimmune or Inflammatory Diseases: An Analysis of the FOURIER Trial. Circulation 2025; 151:1467-1476. [PMID: 40255182 DOI: 10.1161/circulationaha.124.072756] [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: 10/15/2024] [Accepted: 03/18/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Patients with an autoimmune or inflammatory disease (AIID) are at increased cardiovascular risk and may benefit more from statin therapy. In the FOURIER trial (Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk), the PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitor evolocumab lowered low-density lipoprotein cholesterol levels, but not hsCRP (high-sensitivity C-reactive protein) levels, and reduced the risk of cardiovascular events. METHODS FOURIER was a randomized trial of evolocumab versus placebo in 27 564 patients with stable atherosclerosis who were taking statins. This analysis focused on the effect of evolocumab in patients with or without an AIID, defined as any autoimmune or chronic inflammatory condition. The primary end point was a composite of cardiovascular death, myocardial infarction, stroke, unstable angina, or coronary revascularization. RESULTS At baseline, 889 patients (3.2%) had an AIID, most commonly rheumatoid arthritis (33.7%) or psoriasis (15.6%). Median (interquartile range) low-density lipoprotein cholesterol levels were 90.0 mg/dL (79.5-105.5) and 91.5 mg/dL (79.5-108.5) in patients with or without an AIID, respectively (P=0.025), and the placebo-adjusted percent reduction with evolocumab was consistent (60.2% versus 59.0%; P=0.57). Baseline hsCRP was higher in patients with an AIID (median 2.1 versus 1.7 mg/L; P<0.001) and did not significantly change with evolocumab in either group. Compared with placebo, evolocumab reduced the rate of the primary end point by 14% in patients without an AIID (hazard ratio, 0.86 [95% CI, 0.80-0.93]) and by 42% in patients with an AIID (hazard ratio, 0.58 [95% CI, 0.38-0.89]; Pinteraction=0.066). Likewise, evolocumab reduced the key secondary end point of cardiovascular death, myocardial infarction, or stroke by 19% in patients without an AIID (hazard ratio, 0.81 [95% CI, 0.74-0.89]) and 58% in those with an AIID (hazard ratio, 0.42 [95% CI, 0.24-0.74]; Pinteraction=0.022). CONCLUSIONS Intensive lowering of low-density lipoprotein cholesterol levels with evolocumab may lead to greater relative reduction in cardiovascular events in patients with an AIID. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT01764633.
Collapse
Affiliation(s)
- Andre Zimerman
- Hospital Moinhos de Vento, Moinhos de Vento College of Health Sciences, Porto Alegre, Brazil (A.Z., A.L.F.K.)
| | - Ana Laura F Kunzler
- Hospital Moinhos de Vento, Moinhos de Vento College of Health Sciences, Porto Alegre, Brazil (A.Z., A.L.F.K.)
| | - Brittany N Weber
- Division of Cardiovascular Medicine (B.N.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Xinhui Ran
- TIMI Study Group (X.R., S.A.M., M.S.S., R.P.G.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sabina A Murphy
- TIMI Study Group (X.R., S.A.M., M.S.S., R.P.G.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Huei Wang
- Biostatistics (H.W.), Amgen, Thousand Oaks, CA
| | | | - Anthony C Keech
- Sydney Medical School, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Australia (A.C.K.)
| | - Peter S Sever
- National Heart and Lung Institute, Imperial College London, UK (P.S.S.)
| | - Marc S Sabatine
- TIMI Study Group (X.R., S.A.M., M.S.S., R.P.G.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Robert P Giugliano
- TIMI Study Group (X.R., S.A.M., M.S.S., R.P.G.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
7
|
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
|
8
|
Arsha S, Tripathi A, Kangarlu J, Rehman B, Frishman WH, Aronow WS. Chemotherapy-Induced Cardiomyopathy: A Focus on the Utility of Statins. Cardiol Rev 2025:00045415-990000000-00492. [PMID: 40358411 DOI: 10.1097/crd.0000000000000942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Chemotherapy-induced cardiomyopathy (CICM) is a critical adverse consequence associated with chemotherapeutic treatments such as anthracyclines, taxanes, and alkylating agents. Cardiac dysfunction, characterized by left ventricular systolic dysfunction, is the primary effect found in these patients. This may result in heart failure, with heart failure related to chemotherapy resulting in a 3.5-fold increased risk of mortality compared with idiopathic cardiomyopathy alone. Multiple factors, including oxidative stress, inflammation, and disruption of key cellular pathways, are involved in cardiomyocyte damage and influence CICM pathophysiology. So far, dexrazoxane is the sole FDA-approved preventive therapy, but alternative interventions, such as beta-blockers, angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, and statins, have been studied for their cardioprotective potential. Statins, beyond their cholesterol-lowering capabilities, stand out for their pleiotropic effects, including antioxidant, anti-inflammatory, and endothelial-protective actions, which counteract inflammatory effects. Multiple studies and meta-analyses suggest that statin therapy may decrease both the incidence and severity of chemotherapy-related cardiotoxicity (CTX), as evidenced by smaller declines in left ventricular ejection fraction and lower rates of heart failure in statin-treated patients. However, not all investigations confirm these protective benefits; for instance, some trials, including SPARE-HF, reported no significant differences in cardiac outcomes. While these conflicting findings underscore the need for larger randomized trials, they also reflect the heterogeneity of cancer types, chemotherapy regimens, and patient profiles. Statins show promise as a cardioprotective strategy for individuals at risk of CICM. Enhancing patient selection and specifying the timing and duration of statin therapy are essential steps for incorporating these agents into standard care. Optimizing these parameters may reduce chemotherapy-related cardiac damage, improve long-term cardiac function, and enhance overall survival in cancer survivors.
Collapse
Affiliation(s)
- Sanjana Arsha
- From the Department of Internal Medicine, Westchester Medical Center, Vallhala, NY
| | - Ashish Tripathi
- From the Department of Internal Medicine, Westchester Medical Center, Vallhala, NY
| | - John Kangarlu
- From the Department of Internal Medicine, Westchester Medical Center, Vallhala, NY
| | - Bilal Rehman
- From the Department of Internal Medicine, Westchester Medical Center, Vallhala, NY
| | - William H Frishman
- Department of Cardiology, Westchester Medical Center, Valhalla, NY
- Department of Medicine, New York Medical College, Valhalla, NY
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center, Valhalla, NY
- Department of Medicine, New York Medical College, Valhalla, NY
| |
Collapse
|
9
|
Ghafoury R, Malek M, Ismail-Beigi F, Khamseh ME. Role of Residual Inflammation as a Risk Factor Across Cardiovascular-Kidney-Metabolic (CKM) Syndrome: Unpacking the Burden in People with Type 2 Diabetes. Diabetes Ther 2025:10.1007/s13300-025-01743-6. [PMID: 40343683 DOI: 10.1007/s13300-025-01743-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/14/2025] [Indexed: 05/11/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a global health crisis, with cardiovascular disease (CVD) accounting for 75% of mortality in this population. Despite advances in managing traditional risk factors, such as low-density lipoprotein cholesterol (LDL) cholesterol reduction (IMPROVE-IT, FOURIER), antithrombotic therapies (PEGASUS, COMPASS), and triglyceride-lowering agents (REDUCE-IT), a substantial residual cardiovascular risk persists, driven in part by chronic low-grade systemic inflammation. Chronic low-grade inflammation is a central driver of cardiovascular-kidney-metabolic (CKM) syndrome in T2DM, perpetuating residual cardiovascular risk despite optimal management of traditional risk factors. This narrative review synthesizes evidence on how inflammation accelerates coronary heart disease (CHD), heart failure (HF), stroke, diabetic kidney disease (DKD), and peripheral artery disease (PAD). We evaluate the anti-inflammatory mechanisms of current therapies such as statins, sodium-glucose cotransporter 2 (SGLT2) inhibitors, and glucagon-like peptide 1 (GLP-1) receptor agonists, as well as emerging agents like colchicine and interleukin (IL)-1β/IL-6 inhibitors, emphasizing their differential efficacy across CKM traits. By integrating pathophysiological insights with clinical trial data, we propose biomarker-guided strategies to target inflammation as a modifiable risk factor, offering a roadmap to bridge the gap in diabetes-related cardiovascular care.
Collapse
Affiliation(s)
- Roya Ghafoury
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), No. 10, Firoozeh St, Vali-asr Ave, Vali-asr Sq, Tehran, 1593716615, Iran
| | - Mojtaba Malek
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), No. 10, Firoozeh St, Vali-asr Ave, Vali-asr Sq, Tehran, 1593716615, Iran
| | | | - Mohammad E Khamseh
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), No. 10, Firoozeh St, Vali-asr Ave, Vali-asr Sq, Tehran, 1593716615, Iran.
| |
Collapse
|
10
|
De Spiegeleer A, Bronselaer A, Mahieu I, Vreys D, Haslbauer A, Leibfarth JP, Van Schoote L, Wakjira A, Petrovic M, Wynendaele E, De Spiegeleer B, Van Den Noortgate N, Kressig RW, Rössler R. The association between statins and gait speed reserve in older adults: effects of concomitant medication. GeroScience 2025:10.1007/s11357-025-01682-x. [PMID: 40332452 DOI: 10.1007/s11357-025-01682-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
Statins are frequently prescribed to older adults, yet their effects on ageing phenotypes such as frailty or physiological reserves remain poorly understood. Gait Speed Reserve (GSR), defined as the difference between maximal and usual gait speeds, serves as an indicator of physiological reserve, reflecting the body's ability to perform beyond baseline functional levels. Polypharmacy, prevalent in this population, may contribute to inconsistent findings through interactions between statins and concomitant medications. We aimed to investigate how concomitant medications moderate the association between statin use and GSR in older adults. To this end, we conducted a cross-sectional observational cohort study using data from the Mobility Center at the University Department of Geriatric Medicine FELIX PLATTER, Basel, Switzerland (n = 5519 adults aged ≥ 60 years). Moderation regression analyses with propensity score weighting were used to evaluate the effect of concomitant medications on the association between statin use and GSR. Results showed statin use was associated with a lower GSR compared to non-use (- 1.9 cm/s [95% CI, - 3.1 to - 0.72]). However, ACE inhibitors and aspirin significantly influenced this association. The GSR difference for statin users compared to non-users increased by 3.7 cm/s (from - 2.2 to 1.5 cm/s; 95% CI, 0.0 to 7.4) with concomitant ACE inhibitor use and by 5.8 cm/s (from - 3.4 to 2.3 cm/s; 95% CI, 2.5 to 9.1) with aspirin use. We found no statistically significant association between statin use and usual gait speed, the secondary outcome. In conclusion, ACE inhibitors and aspirin interacted with statins, reversing the negative association with GSR into a positive one when co-used. Future clinical trials are needed to determine causality and further investigate the impact of concomitant medication use on statin effects in aging populations. Meanwhile, our findings underscore the importance of considering concomitant medication use when assessing the effects of statins in older adults.
Collapse
Affiliation(s)
- Anton De Spiegeleer
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University, Ghent, Belgium
- Department of Geriatrics, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Antoon Bronselaer
- Department of Telecommunications & Information Processing, Faculty of Engineering and Architecture, Ghent University, Ghent, Belgium
| | - Ine Mahieu
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University, Ghent, Belgium
- Department of Geriatrics, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Dorien Vreys
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University, Ghent, Belgium
- Department of Geriatrics, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Aaron Haslbauer
- University Department of Geriatric Medicine FELIX PLATTER, Basel, Switzerland
| | - Jan-Philipp Leibfarth
- Department of Telecommunications & Information Processing, Faculty of Engineering and Architecture, Ghent University, Ghent, Belgium
| | - Lara Van Schoote
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University, Ghent, Belgium
- Department of Geriatrics, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Aster Wakjira
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University, Ghent, Belgium
- Jimma University, School of Pharmacy, Jimma, Ethiopia
| | - Mirko Petrovic
- Department of Geriatrics, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Evelien Wynendaele
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University, Ghent, Belgium
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Bart De Spiegeleer
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University, Ghent, Belgium
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Nele Van Den Noortgate
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University, Ghent, Belgium
- Department of Geriatrics, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Reto W Kressig
- University Department of Geriatric Medicine FELIX PLATTER, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Roland Rössler
- University Department of Geriatric Medicine FELIX PLATTER, Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
11
|
Romeo S, Vidal-Puig A, Husain M, Ahima R, Arca M, Bhatt DL, Diehl AM, Fontana L, Foo R, Frühbeck G, Kozlitina J, Lonn E, Pattou F, Plat J, Quaggin SE, Ridker PM, Rydén M, Segata N, Tuttle KR, Verma S, Roeters van Lennep J, Benn M, Binder CJ, Jamialahmadi O, Perkins R, Catapano AL, Tokgözoğlu L, Ray KK. Clinical staging to guide management of metabolic disorders and their sequelae: a European Atherosclerosis Society consensus statement. Eur Heart J 2025:ehaf314. [PMID: 40331343 DOI: 10.1093/eurheartj/ehaf314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
Obesity rates have surged since 1990 worldwide. This rise is paralleled by increases in pathological processes affecting organs such as the heart, liver, and kidneys, here termed systemic metabolic disorders (SMDs). For clinical management of SMD, the European Atherosclerosis Society proposes a pathophysiology-based system comprising three stages: Stage 1, where metabolic abnormalities such as dysfunctional adiposity and dyslipidaemia occur without detectable organ damage; Stage 2, which involves early organ damage manifested as Type 2 diabetes, asymptomatic diastolic dysfunction, metabolic-associated steatohepatitis (MASH), and chronic kidney disease (CKD); and Stage 3, characterized by more advanced organ damage affecting multiple organs. Various forms of high-risk obesity, driven by maintained positive energy balance, are the most common cause of SMD, leading to ectopic lipid accumulation and insulin resistance. This progression affects various organs, promoting comorbidities such as hypertension and atherogenic dyslipidaemia. Genetic factors influence SMD susceptibility, and ethnic disparities in SMD are attributable to genetic and socioeconomic factors. Key SMD features include insulin resistance, inflammation, pre-diabetes, Type 2 diabetes, MASH, hypertension, CKD, atherogenic dyslipidaemia, and heart failure. Management strategies involve lifestyle changes, pharmacotherapy, and metabolic surgery in severe cases, with emerging treatments focusing on genetic approaches. The staging system provides a structured approach to understanding and addressing the multi-faceted nature of SMD, which is crucial for improving health outcomes. Categorization of SMD abnormalities by presence and progression is aimed to improve awareness of a multi-system trait and encourage a tailored and global approach to treatment, ultimately aiming to reduce the burden of obesity-related comorbidities.
Collapse
Affiliation(s)
- Stefano Romeo
- Department of Medicine, H7 Medicin, Huddinge, H7 Endokrinologi och Diabetes Romeo, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital Huddinge, 141 57 Huddinge, Stockholm, Sweden
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Viale Europa, 88100 Catanzaro, Italy
| | - Antonio Vidal-Puig
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- Centro de Investigacion Principe Felipe, C/ d'Eduardo Primo Yufera, 3, 46012 Valencia, Spain
- Cambridge University Nanjing Centre of Technology and Innovation, No. 23, Rongyue Road, Jiangbei New Area, Nanjing, Jiangsu, China
| | - Mansoor Husain
- Ted Rogers Centre for Heart Research, Department of Medicine, University of Toronto, 661 University Avenue, Toronto, ON, Canada M5G 1M1
| | - Rexford Ahima
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
- Unit of Internal Medicine and Metabolic Diseases, Hospital Policlinico Umberto I, Rome, Italy
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, NC, USA
| | - Luigi Fontana
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Roger Foo
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, National University Health Systems, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Gema Frühbeck
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
- Metabolic Research Laboratory, CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Julia Kozlitina
- The Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eva Lonn
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Francois Pattou
- Department of Endocrine and Metabolic Surgery, CHU Lille, University of Lille, Inserm, Institut Pasteur Lille, Lille, France
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, NUTRIM School of Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Susan E Quaggin
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Nephrology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Katherine R Tuttle
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
- Providence Medical Research Center, Providence Inland Northwest Health, Spokane, WA, USA
| | - Subodh Verma
- Division of Cardiac Surgery, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Jeanine Roeters van Lennep
- Department of Internal Medicine, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marianne Benn
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Centre of Diagnostic Investigation, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Rosie Perkins
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Alberico L Catapano
- Center for the Study of Atherosclerosis, IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Lale Tokgözoğlu
- Department of Cardiology, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK
| |
Collapse
|
12
|
Borda MG, Ramírez-Vélez R, Botero-Rodriguez F, Patricio-Baldera J, de Lucia C, Pola I, Barreto GE, Khalifa K, Bergland AK, Kivipelto M, Cederholm T, Zetterberg H, Ashton NJ, Ballard C, Siow R, Aarsland D. Anthocyanin supplementation in adults at risk for dementia: a randomized controlled trial on its cardiometabolic and anti-inflammatory biomarker effects. GeroScience 2025:10.1007/s11357-025-01669-8. [PMID: 40314845 DOI: 10.1007/s11357-025-01669-8] [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: 02/20/2025] [Accepted: 04/15/2025] [Indexed: 05/03/2025] Open
Abstract
Anthocyanins are dietary flavonoids shown to have a therapeutic capacity to mitigate inflammation and oxidative stress. The present secondary analyses from the "Anthocyanins in People at Risk for Dementia Study" were aimed at (I) determining the intervention's effect on blood-based markers of cardiovascular disease and inflammation and (II) evaluating whether baseline factors such as age, sex, inflammation, or cardiometabolic score may moderate the intervention's effect on inflammatory status. This study was an ancillary, 24-week randomized, double-blind, placebo-controlled Phase II trial. Sub-sample participants (n = 99), aged 60-80 years with mild cognitive impairment or cardiometabolic disorders, were randomized to receive either 320 mg/day of anthocyanins or placebo. The biomarkers analyzed included inflammatory biomarker assessment (IL - 6, IL - 8, IL - 10, IL - 1b, TNF - α, IFN - γ), and C-reactive protein (CRP), as well as albumin, thrombocytes, cholesterol, LDL, HDL, and triglycerides, which were longitudinally compared between both groups. Baseline characteristics were balanced between the groups. ANCOVA analyses reveal 24-week differences favoring the anthocyanin treatment in LDL cholesterol levels (ƞp2 = 0.078; p = 0.015), cardiometabolic score (ƞp2 = 0.073; p = 0.021), CRP levels (ƞp2 = 0.417; p = 0.0001), IL - 6 (ƞp2 = 0.085; p = 0.015), IL - 1b (ƞp2 = 0.058; p = 0.037), and Inflam z-score 5 (ƞp2 = 0.059, p = 0.004). Moderation analysis demonstrated that the inflammatory score at baseline was a significant predictor of the effect of the intervention on the CRP levels. Anthocyanin supplementation reduces CRP and cardiovascular disease biomarkers in individuals at risk of dementia, especially when there is increased inflammation at baseline. ClinicalTrials.gov study identifier: NCT03419039.
Collapse
Affiliation(s)
- Miguel German Borda
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway.
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain.
- Centro de Investigación en Ciencias de La Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan Edo. de México, México.
| | - Robinson Ramírez-Vélez
- Navarrabiomed, IdiSNA, Hospital Universitario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Felipe Botero-Rodriguez
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- SynaptIA - Inteligencia artificial para la investigación en salud mental, Bogotá, Colombia
- Centro de Memoria y Cognición Intellectus, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Jonathan Patricio-Baldera
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Instituto de Investigación en Salud, Facultad de Ciencias de La Salud de La Universidad Autónoma de Santo Domingo, Santo Domingo, Dominican Republic
| | - Chiara de Lucia
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Ilaria Pola
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Khadija Khalifa
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
| | - Anne Katrine Bergland
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Tommy Cederholm
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, 62167, Uppsala, Sweden
- Theme Inflammation & Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK, Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Nicholas J Ashton
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Banner Alzheimer's Institute, University of Arizona, Phoenix, AZ, USA
| | - Clive Ballard
- Medical School, University of Exeter, University of Exeter, Exeter, UK
| | - Richard Siow
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Dag Aarsland
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| |
Collapse
|
13
|
Zhang Y, Li Y, Liu Y, Willett WC, Manson JE, Stampfer MJ, Hu FB, Giovannucci EL, Wang DD. Duration and type of statin use and long-term risk of type 2 diabetes among men and women with hypercholesterolaemia: findings from three prospective cohorts. Diabetologia 2025:10.1007/s00125-025-06441-3. [PMID: 40316730 DOI: 10.1007/s00125-025-06441-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/04/2025] [Indexed: 05/04/2025]
Abstract
AIMS/HYPOTHESIS Findings from RCTs and observational studies indicate a positive association between statin use and risk of type 2 diabetes. Mendelian randomisation studies provide evidence to support that the effect is causal. However, little is known about the long-term effects, and data on different types of statins remain limited. METHODS We analysed participants with hypercholesterolaemia from the Nurses' Health Study (NHS; 30,510 participants), the Nurses' Health Study II (NHSII; 21,547 participants) and the Health Professionals Follow-Up Study (HPFS; 9934 participants) who were free of diabetes, CVD and cancer at baseline. Statin use was assessed every 2 years starting in 2000 in the NHS and the HPFS and in 1999 in the NHSII. Incident cases of type 2 diabetes were confirmed by a validated supplementary questionnaire until the end of follow-up (31 January 2023). RESULTS We documented 6762 incident type 2 diabetes cases during up to 23 years of follow-up. Compared with non-users, statin users had a significantly higher risk of type 2 diabetes after adjustment for BMI and other potential confounding variables (pooled HR 1.40; 95% CI 1.33, 1.48). Compared with non-use, durations of statin use of 1-5, 6-10, 11-15 and >15 years were associated with HRs of 1.36 (95% CI 1.27, 1.44), 1.41 (95% CI 1.31, 1.52), 1.60 (95% CI 1.44, 1.78) and 1.76 (95% CI 1.50, 2.06), respectively; significant linear trends were observed when the comparison included non-users and within statin users only (both ptrend<0.001). Compared with non-users, the HRs for type 2 diabetes associated with 10 year use of specific types of statins were 1.99 (95% CI 1.45, 2.73) for rosuvastatin, 1.66 (95% CI 1.12, 2.47) for lovastatin, 1.62 (95% CI 1.39, 1.89) for atorvastatin, 1.44 (95% CI 1.06, 1.97) for pravastatin and 1.37 (95% CI 1.13, 1.66) for simvastatin. Use of a low-potency statin for 10 years was associated with a 34% higher risk of type 2 diabetes (HR 1.34; 95% CI 1.15, 1.56), while use of a high-potency statin for 10 years was associated with a 72% higher risk (HR 1.72; 95% CI 1.46, 2.04). The difference in the 10 year cumulative risk of type 2 diabetes comparing statin users vs non-users was most pronounced in participants with the least healthy lifestyles (4.5% vs 3.1%), while the smallest risk differential was observed among participants who adhered to the healthiest lifestyles (1.0% vs 0.4%). CONCLUSIONS/INTERPRETATION The positive association between statin use and type 2 diabetes was more pronounced with a longer duration of use, and the association varied across different types of statins. Adopting and maintaining a healthy lifestyle can serve as a viable approach to diabetes prevention during statin treatment.
Collapse
Affiliation(s)
- Yiwen Zhang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yanping Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yuxi Liu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Walter C Willett
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Meir J Stampfer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
14
|
Ki Y, Kim W, Lee KH, Han S, Kim Y, Doh J, Kim TN, Chung CH, Kim DY, Cho J, Yoon H, Jeong I, Park S, Song K, Yu CW, Cho D, Choi SH, Oh S, Shin S, Jeong H, Park Y, Kim H. Lipid-Lowering Effect and Safety of Ezetimibe and Atorvastatin 5 mg in Patients With Primary Hypercholesterolemia or Mixed Dyslipidemia: A Randomized, Double-Blind, Parallel, Multicenter, Phase 3 Clinical Trial. Clin Cardiol 2025; 48:e70138. [PMID: 40357888 PMCID: PMC12070249 DOI: 10.1002/clc.70138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/09/2025] [Accepted: 04/13/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVE This study aimed to compare the lipid-lowering effect and safety of low-intensity atorvastatin (5 mg) plus ezetimibe (10 mg) combination therapy (A5E10) with monotherapy regimens-atorvastatin 5 mg [A5], ezetimibe 10 mg [E10], and atorvastatin 10 mg [A10])-in dyslipidemia patients. METHODS A randomized, double-blind, placebo-controlled trial involving 252 dyslipidemia patients was conducted at 25 centers in South Korea (NCT05970679). Participants aged ≥ 19 years were randomized into four groups: A5E10, A5, E10, and A10. The primary endpoint was the percentage change in low-density lipoprotein cholesterol (LDL-C) levels from baseline to 8 weeks. Secondary endpoints included changes in other lipid parameters, lipid ratios, LDL-C goal achievement rates and safety assessments. RESULTS The mean age of the patients was 63 years, and 51.2% were male. The A5E10 group showed significantly greater LDL-C reduction (47.6%) compared with A5 (33.4%), E10 (19.4%), and A10 (40.1%) at 8 weeks (p < 0.0001). A5E10 also significantly reduced triglyceride, non-high-density lipoprotein cholesterol, and apolipoprotein B levels. In addition, a significant reduction in LDL-C levels was observed over the 4 weeks, with a 46.7% reduction in LDL-C levels after 4 weeks of A5E10 administration. No severe adverse events were observed in the A5E10 group. CONCLUSION The combination of low-intensity atorvastatin and ezetimibe was more effective than moderate-intensity atorvastatin monotherapy in lowering LDL-C levels and improving other lipid parameters. It was well-tolerated and demonstrated rapid benefits within a month, offering a promising alternative for patients with low to moderate cardiovascular risk who do not achieve adequate control with statin monotherapy.
Collapse
Affiliation(s)
- You‐Jeong Ki
- Cardiovascular Center, Department of Internal MedicineUijeongbu Eulji Medical CenterUijeongbuRepublic of Korea
| | - Weon Kim
- Cardiovascular Division, Department of Internal MedicineKyung Hee University Hospital, Kyung Hee UniversitySeoulRepublic of Korea
| | - Ki Hong Lee
- Cardiovascular Division, Department of Internal MedicineChonnam National University Medical School & HospitalGwangjuRepublic of Korea
| | - Sang‐Jin Han
- Division of Cardiology, Department of Internal MedicineHallym University Sacred Heart HospitalAnyangRepublic of Korea
| | - Yong‐Hyun Kim
- Department of EndocrinologyBundang Jesaeng HospitalBundang‐guGyeonggidoRepublic of Korea
| | - Joon‐Hyung Doh
- Inje University Ilsan Paik HospitalGoyangRepublic of Korea
| | - Tae Nyun Kim
- Division of Endocrinology and Metabolism, Department of Internal MedicineInje University Haeundae Paik HospitalBusanRepublic of Korea
| | - Choon Hee Chung
- Department of Internal Medicine and Research Institute of Metabolism and InflammationYonsei University Wonju College of MedicineWonjuRepublic of Korea
| | - Do Young Kim
- Division of Cardiology, Department of Internal MedicineAjou University Hospital and Ajou School of MedicineSuwonRepublic of Korea
| | - Jin‐Man Cho
- Department of Cardiovascular MedicineKyung Hee University Hospital at GangdongSeoulRepublic of Korea
| | - Hyuck‐Jun Yoon
- Cardiovascular CenterKeimyung University Dongsan HospitalDaeguRepublic of Korea
| | - In‐Kyung Jeong
- Division of Endocrinology and Metabolism, Department of Internal MedicineKyung Hee University Hospital at Gangdong, Kyung Hee University College of MedicineSeoulRepublic of Korea
| | - Sungha Park
- Cardiovascular Research InstituteYonsei University College of MedicineSeoulRepublic of Korea
| | - Kee‐Ho Song
- Division of Endocrinology and MetabolismKonkuk University Medical Center, Konkuk University School of MedicineRepublic of Korea
| | - Cheol Woong Yu
- Department of Cardiology, Cardiovascular CenterKorea University Anam HospitalSeoulRepublic of Korea
| | - Deok‐Kyu Cho
- Yongin Severance HospitalYonsei University College of MedicineRepublic of Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University College of MedicineSeoul National University Bundang HospitalSeongnam‐CityRepublic of Korea
| | - Seung‐Jin Oh
- Division of CardiologyNational Health Insurance Service Ilsan HospitalGoyangRepublic of Korea
| | - Sanghoon Shin
- Division of Cardiology, Department of Internal MedicineEwha Womans University Seoul HospitalSeoulRepublic of Korea
| | - Hyeonju Jeong
- Division of Cardiology, Department of internal medicine, Myongji HospitalHanyang University Medical CenterGoyangRepublic of Korea
| | - Yongwhi Park
- Division of Cardiology, Department of Internal MedicineGyeongsang National University Changwon HospitalChangwonRepublic of Korea
| | - Hyo‐Soo Kim
- Cardiovascular Center, Department of Internal MedicineSeoul National University HospitalSeoulRepublic of Korea
| |
Collapse
|
15
|
Masson W, Fernández-Villar G, Martinez-Elhelou S. Management of Atherosclerotic Cardiovascular Risk in Inflammatory Bowel Disease: Current Perspectives. Adv Ther 2025; 42:2118-2134. [PMID: 40146370 PMCID: PMC12006232 DOI: 10.1007/s12325-025-03154-2] [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: 12/13/2024] [Accepted: 02/24/2025] [Indexed: 03/28/2025]
Abstract
Inflammatory bowel disease (IBD) is a complex condition characterized by inflammation of the gastrointestinal system, encompassing Crohn's disease and ulcerative colitis. Patients diagnosed with IBD have an increased risk of atherosclerotic cardiovascular disease. This heightened risk can be attributed to a combination of mechanisms, including traditional risk factors, chronic inflammation, intestinal dysbiosis, increased risk of thrombosis, and the use of certain medications such as corticosteroids. There are significant gaps in current knowledge, particularly regarding the management of risk factors and the use of medications for cardiovascular disease prevention. Similarly, the cardiovascular effects of specific IBD therapies, particularly the newer ones, are not yet fully understood. This review focuses on the epidemiological evidence linking IBD with cardiovascular risk factors and cardiovascular disease. It describes the potential pathophysiological mechanisms underlying this association and examines the challenges involved in accurately assessing cardiovascular risk in these patients, including the utility of complementary tools such as subclinical atherosclerosis detection. Additionally, we consider the potential therapeutic implications for managing these patients. Finally, this review also underscores the importance of multidisciplinary collaboration. Effective teamwork among gastroenterologists, cardiologists, and general practitioners is essential for providing comprehensive care to patients with IBD.
Collapse
Affiliation(s)
- Walter Masson
- Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina.
| | | | | |
Collapse
|
16
|
Rai H, Reddi R, Coughlan JJ, Durand R, O'Callaghan D, Colleran R, Byrne RA. Pre-Percutaneous Coronary Intervention C-Reactive Protein Levels and In-Stent Restenosis: A Systematic Review and Meta-Analysis. Health Sci Rep 2025; 8:e70757. [PMID: 40309624 PMCID: PMC12040731 DOI: 10.1002/hsr2.70757] [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: 11/15/2024] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction C-reactive protein (CRP) is an inflammatory biomarker, implicated in the pathogenesis of atherosclerotic lesion formation, plaque rupture, and coronary thrombosis. The relationship between preprocedural CRP levels and subsequent development of in-stent restenosis (ISR) after percutaneous coronary intervention (PCI) however remains uncertain. Against this background, we performed a systematic review and meta-analysis, investigating the association between baseline CRP levels and the incidence of ISR after PCI. Methods Relevant published studies were identified following a PubMed, EMBASE, MEDLINE, Scopus and Web of Knowledge databases search until April 30, 2024. To be included, studies had to be original research with: (i) angiographically determined ISR group, (ii) angiographically or clinically determined no-ISR group, and (iii) CRP levels measured before the index PCI procedure for both groups. The mean difference in baseline CRP levels and associated 95% confidence interval (CI) between the ISR and no-ISR groups was ascertained for each included study. The pooled standard mean difference (SMD) and its 95% CI was derived after pooling study-level results using a random effects model, employing a Z-test. Begg's funnel plots and Egger's test were used for publication bias assessment. Results Out of a total of 1018 unique results, 19 studies, with a total sample size of 4744 patients (1154 restenosis cases/3590 controls), were included for quantitative synthesis. ISR group had higher baseline CRP levels (SMD = 0.41, 95% CI = 0.16, 0.66 mg/L, p = 0.001) in comparison to the no-ISR group. No evidence of publication bias was detected either visually by Begg's funnel plots or by Egger's test (p = 0.08). Our leave-one-out sensitivity analysis further attested our obtained associations. Conclusions The present systematic review and meta-analysis suggests a significant association of elevated pre-PCI CRP with subsequent angiographically confirmed ISR. These results warrant further validation in dedicated large cohorts, ideally in a prospective setting.
Collapse
Affiliation(s)
- Himanshu Rai
- Department of Cardiology and Cardiovascular Research Institute (CVRI) DublinMater Private NetworkDublinIreland
- School of Pharmacy and Biomolecular SciencesRCSI University of Medicine and Health SciencesDublinIreland
| | - Renitha Reddi
- School of MedicineRCSI University of Medicine and Health SciencesDublinIreland
| | - J. J. Coughlan
- Department of Cardiology and Cardiovascular Research Institute (CVRI) DublinMater Private NetworkDublinIreland
| | - Rory Durand
- Department of Cardiology and Cardiovascular Research Institute (CVRI) DublinMater Private NetworkDublinIreland
| | - Daniel O'Callaghan
- Department of Cardiology and Cardiovascular Research Institute (CVRI) DublinMater Private NetworkDublinIreland
| | - Roisin Colleran
- Department of Cardiology and Cardiovascular Research Institute (CVRI) DublinMater Private NetworkDublinIreland
- School of Pharmacy and Biomolecular SciencesRCSI University of Medicine and Health SciencesDublinIreland
| | - Robert A. Byrne
- Department of Cardiology and Cardiovascular Research Institute (CVRI) DublinMater Private NetworkDublinIreland
- School of Pharmacy and Biomolecular SciencesRCSI University of Medicine and Health SciencesDublinIreland
| |
Collapse
|
17
|
Marín-Jiménez I, Carpio D, Hernández V, Muñoz F, Zatarain-Nicolás E, Zabana Y, Mañosa M, Rodríguez-Moranta F, Barreiro-de Acosta M, Gutiérrez Casbas A. Spanish Working Group in Crohn's Disease and Ulcerative Colitis (GETECCU) position paper on cardiovascular disease in patients with inflammatory bowel disease. GASTROENTEROLOGIA Y HEPATOLOGIA 2025; 48:502314. [PMID: 39615874 DOI: 10.1016/j.gastrohep.2024.502314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 01/12/2025]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide. Therefore, it is essential to understand their relationship and prevalence in different diseases that may present specific risk factors for them. The objective of this document is to analyze the specific prevalence of CVD in patients with inflammatory bowel disease (IBD), describing the presence of classical and non-classical cardiovascular risk factors in these patients. Additionally, we will detail the pathophysiology of atherosclerosis in this patient group and the different methods used to assess cardiovascular risk, including the use of risk calculators in clinical practice and different ways to assess subclinical atherosclerosis and endothelial dysfunction. Furthermore, we will describe the potential influence of medication used for managing patients with IBD on cardiovascular risk, as well as the potential influence of commonly used drugs for managing CVD on the course of IBD. The document provides comments and evidence-based recommendations based on available evidence and expert opinion. An interdisciplinary group of gastroenterologists specialized in IBD management, along with a consulting cardiologist for this type of patients, participated in the development of these recommendations by the Spanish Group of Work on Crohn's Disease and Ulcerative Colitis (GETECCU).
Collapse
Affiliation(s)
- Ignacio Marín-Jiménez
- Sección de Gastroenterología, Servicio de Aparato Digestivo, Instituto de Investigación Sanitaria (IiSGM), Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España.
| | - Daniel Carpio
- Servicio de Aparato Digestivo, Complexo Hospitalario Universitario de Pontevedra, Pontevedra, España; Grupo de Investigación en Hepatología-Enfermedades Inflamatorias Intestinales, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Pontevedra, España
| | - Vicent Hernández
- Servicio de Aparato Digestivo, Complexo Hospitalario Universitario de Vigo (CHUVI), SERGAS, Vigo, Pontevedra, España; Grupo de Investigación en Patología Digestiva, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Pontevedra, España
| | - Fernando Muñoz
- Servicio de Digestivo. Complejo Asistencial Universitario de Salamanca, Salamanca, España
| | - Eduardo Zatarain-Nicolás
- Servicio de Cardiología, Instituto de Investigación Sanitaria (IiSGM), Hospital General Universitario Gregorio Marañón, Madrid; CIBERCV, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Universidad Complutense de Madrid, Madrid, España
| | - Yamile Zabana
- Servicio de Aparato Digestivo, Hospital Universitari Mútua Terrassa; Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Terrasa, Barcelona, España
| | - Míriam Mañosa
- Servicio de Aparato Digestivo, Hospital Universitari Germans Trias; Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Badalona, Barcelona, España
| | - Francisco Rodríguez-Moranta
- Servicio de Aparato Digestivo, Hospital Universitario de Bellvitge, IDIBELL, L'Hospitalet de Llobregat, Barcelona, España
| | - Manuel Barreiro-de Acosta
- Servicio de Gastroenterología, Hospital Clínico Universitario de Santiago, A Coruña, España; Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, España
| | - Ana Gutiérrez Casbas
- Servicio Medicina Digestiva, Hospital General Universitario Dr Balmis de Alicante, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), CIBERehd, Alicante, España
| |
Collapse
|
18
|
Hurwitz M, Agboola OJ, Gami A, Williams MS, Virani SS, Sharma GV, Patel J. Strategies for the Secondary Prevention of Atherosclerotic Cardiovascular Disease. US CARDIOLOGY REVIEW 2025; 19:e11. [PMID: 40342903 PMCID: PMC12060178 DOI: 10.15420/usc.2024.33] [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: 06/25/2024] [Accepted: 02/26/2025] [Indexed: 05/11/2025] Open
Abstract
Patients with atherosclerotic cardiovascular disease (ASCVD), such as those with a history of MI or stroke, are at high risk for morbidity and mortality associated with future cardiovascular events. Ideal management of these patients requires a multifactorial strategy for risk factor mitigation and prevention of additional cardiovascular events. Traditional management of secondary prevention patients involves lipid-lowering with statins, blood pressure control, and anti-platelet treatment. Several additional targets have been identified to optimize the secondary prevention of ASCVD, such as further lipid control, inflammation management, lifestyle and weight optimization, strict diabetes control, use of β-blockers, use of renin-angiotensin-aldosterone system inhibitors, vaccinations, and additional considerations of anti-thrombotic therapies. This review will describe the interventions associated with these targets, as well as the relevant research and indications for these therapies.
Collapse
Affiliation(s)
- Madelyn Hurwitz
- School of Medicine, University of VirginiaCharlottesville, VA
| | - Olayinka J Agboola
- Department of Cardiology, Inova Schar Heart and Vascular InstituteFalls Church, VA
| | - Abhishek Gami
- Division of Cardiology, Johns Hopkins University School of MedicineBaltimore, MD
| | - Marlene S Williams
- Division of Cardiology, Johns Hopkins University School of MedicineBaltimore, MD
| | - Salim S Virani
- Department of Medicine, The Aga Khan UniversityKarachi, Pakistan
- Texas Heart Institute and Baylor College of MedicineHouston, TX
| | - Garima V Sharma
- Department of Cardiology, Inova Schar Heart and Vascular InstituteFalls Church, VA
| | - Jaideep Patel
- Division of Cardiology, Johns Hopkins University School of MedicineBaltimore, MD
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of MedicineMD
| |
Collapse
|
19
|
Chen H, Zhang Y, Aikebaier M, Du Y, Liu Y, Zha Q, Zheng L, Shan S, Wang Y, Chen J, Li Y, Yang K, Yang Y, Cui W. Decursin-Loaded Nanovesicles Target Macrophages Driven by the Pathological Process of Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417489. [PMID: 40285666 DOI: 10.1002/advs.202417489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Atherosclerosis (AS) is a major pathological factor contributing to the mortality associated with ischemic heart disease and is driven primarily by macrophage-mediated lipid accumulation and inflammatory processes. Conventional cardiovascular pharmacotherapies address these pathological mechanisms but often show limited efficacy, highlighting the need for innovative agents capable of effectively reducing lipid accumulation and inflammation with minimal toxicity. In this study, decursin, a monomer derived from traditional Chinese medicine, is shown to inhibit both lipid accumulation and inflammatory responses in macrophages through direct interaction with protein kinase Cδ (PKCδ), resulting in low cytotoxicity in vitro and negligible toxicity in vivo. To address the short half-life of decursin, a targeted cascade drug delivery system (ALD@EM), which is specifically designed to target AS pathophysiology, is developed. This system employs ICAM-1 and VCAM-1 antibodies for plaque localization and incorporates low-density lipoproteins (LDLs) to facilitate chemotaxis to lesion sites, with an inner layer of apoptotic endothelial cell membranes to increase macrophage internalization and drug release. As a result, ALD@EM nanovesicles significantly increased the accumulation and therapeutic efficacy of decursin within plaques, substantially reducing lipid deposition and plaque inflammation, thereby offering a novel strategy for targeted AS treatment.
Collapse
Affiliation(s)
- Hui Chen
- Department of Endocrinology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yifeng Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mirenuer Aikebaier
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yawei Du
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200002, China
| | - Yan Liu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Qing Zha
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shuyao Shan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanping Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jiawei Chen
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yiping Li
- Department of Endocrinology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
| | - Ke Yang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Yang
- Department of Endocrinology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
| | - Wenguo Cui
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
20
|
Zaman S, Wasfy JH, Kapil V, Ziaeian B, Parsonage WA, Sriswasdi S, Chico TJA, Capodanno D, Colleran R, Sutton NR, Song L, Karam N, Sofat R, Fraccaro C, Chamié D, Alasnag M, Warisawa T, Gonzalo N, Jomaa W, Mehta SR, Cook EES, Sundström J, Nicholls SJ, Shaw LJ, Patel MR, Al-Lamee RK. The Lancet Commission on rethinking coronary artery disease: moving from ischaemia to atheroma. Lancet 2025; 405:1264-1312. [PMID: 40179933 DOI: 10.1016/s0140-6736(25)00055-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/01/2025] [Accepted: 01/09/2025] [Indexed: 04/05/2025]
Affiliation(s)
- Sarah Zaman
- Westmead Applied Research Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
| | - Jason H Wasfy
- Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Vikas Kapil
- William Harvey Research Institute, Centre for Cardiovascular Medicine and Devices, NIHR Barts Biomedical Research Centre, Queen Mary University of London, St Bartholomew's Hospital, London, UK
| | - Boback Ziaeian
- Division of Cardiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA
| | - William A Parsonage
- Australian Centre for Health Services Innovation, Queensland University of Technology, Brisbane, QLD, Australia; Department of Cardiology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Sira Sriswasdi
- Center of Excellence in Computational Molecular Biology, Chulalongkorn University, Pathum Wan, Bangkok, Thailand; Faculty of Medicine, Chulalongkorn University, Pathum Wan, Bangkok, Thailand
| | - Timothy J A Chico
- School of Medicine and Population Health, University of Sheffield, Sheffield, UK; British Heart Foundation Data Science Centre, Health Data Research UK, London, UK
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero Universitaria Policlinico, University of Catania, Catania, Italy
| | - Róisín Colleran
- Department of Cardiology and Cardiovascular Research Institute, Mater Private Network, Dublin, Ireland; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Nadia R Sutton
- Department of Internal Medicine, and Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Lei Song
- Department of Cardiology, National Clinical Research Centre for Cardiovascular Diseases, Fuwai Hospital, Beijing, China; Peking Union Medical College (Chinese Academy of Medical Sciences), Beijing, China
| | - Nicole Karam
- Cardiology Department, European Hospital Georges Pompidou, Paris City University, Paris, France
| | - Reecha Sofat
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Chiara Fraccaro
- Division of Cardiology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Daniel Chamié
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Mirvat Alasnag
- Cardiac Center, King Fahd Armed Forces Hospital, Jeddah, Saudi Arabia
| | | | - Nieves Gonzalo
- Cardiology Department, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Walid Jomaa
- Cardiology B Department, Fattouma Bourguiba University Hospital, University of Monastir, Monastir, Tunisia
| | - Shamir R Mehta
- Population Health Research Institute, Hamilton Health Sciences, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Elizabeth E S Cook
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Johan Sundström
- Uppsala University, Uppsala, Sweden; The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | | | - Leslee J Shaw
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manesh R Patel
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA; Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Rasha K Al-Lamee
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
21
|
Bittner V, Linnebur SA, Dixon DL, Forman DE, Green AR, Jacobson TA, Orkaby AR, Saseen JJ, Virani SS. Managing Hypercholesterolemia in Adults Older Than 75 years Without a History of Atherosclerotic Cardiovascular Disease: An Expert Clinical Consensus From the National Lipid Association and the American Geriatrics Society. J Am Geriatr Soc 2025. [PMID: 40207842 DOI: 10.1111/jgs.19398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 09/07/2025] [Indexed: 04/11/2025]
Abstract
The risk of atherosclerotic cardiovascular disease increases with advancing age. Elevated LDL-cholesterol and non-HDL-cholesterol levels remain predictive of incident atherosclerotic cardiovascular events among individuals older than 75 years. Risk prediction among older individuals is less certain because most current risk calculators lack specificity in those older than 75 years and do not adjust for co-morbidities, functional status, frailty, and cognition which significantly impact prognosis in this age group. Data on the benefits and risks of lowering LDL-cholesterol with statins in older patients without atherosclerotic cardiovascular disease are also limited since most primary prevention trials have included mostly younger patients. Available data suggest that statin therapy in older primary prevention patients may reduce atherosclerotic cardiovascular events and that benefits from lipid-lowering with statins outweigh potential risks such as statin-associated muscle symptoms and incident Type 2 diabetes mellitus. While some evidence suggests the possibility that statins may be associated with incident cognitive impairment in older adults, a preponderance of literature indicates neutral or even protective statin-related cognitive effects. Shared decision-making which is recommended for all patients when considering statin therapy is particularly important in older patients. Randomized clinical trial data evaluating the use of non-statin lipid-lowering therapy in older patients are sparse. Deprescribing of lipid-lowering agents may be appropriate for select patients older than 75 years with life-limiting diseases. Finally, a patient-centered approach should be taken when considering primary prevention strategies for older adults.
Collapse
Affiliation(s)
- Vera Bittner
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sunny A Linnebur
- University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA
| | - Dave L Dixon
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, USA
| | - Daniel E Forman
- Department of Medicine (Divisions of Geriatrics and Cardiology), University of Pittsburgh and Pittsburgh Geriatrics, Research, Education, and Clinical Center (GRECC), VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - Ariel R Green
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Terry A Jacobson
- Lipid Clinic and Cardiovascular Risk Reduction Program, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Ariela R Orkaby
- New England Geriatric Education, Research and Clinical Center (GRECC), VA Boston Health Care System, Division of Aging, Brigham & Women's Hospital, Harvard Medical School, USA
| | - Joseph J Saseen
- Department of Clinical Pharmacy and Department of Family Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado, USA
| | - Salim S Virani
- Section of Cardiology, Department of Medicine, The Aga Khan University, Karachi, Pakistan
- Texas Heart Institute and Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
22
|
Iakoubova OA, Haji-Sheikhi F, Louie JZ, Rowland CM, Arellano AR, Bare LA, Birse CE, Penn MS. Association of MPO levels with cardiometabolic disease stratified by atherosclerotic cardiovascular risk and intensity of therapy in a workforce population. Sci Rep 2025; 15:12244. [PMID: 40210927 PMCID: PMC11986125 DOI: 10.1038/s41598-025-89373-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 02/05/2025] [Indexed: 04/12/2025] Open
Abstract
Cardiometabolic risk increases cardiovascular (CVD), chronic kidney (CKD) and non-alcoholic fatty liver (NAFLD) disease risk. High myeloperoxidase (MPO) levels identify individuals at risk for CVD. We whether elevation of MPO associated with kidney and liver disease risk in subgroups stratified by ASCVD risk and intensity of therapy. Adjusted logistic models assessed the associations of MPO with markers of kidney disease (estimated glomerular filtration rate) and liver fibrosis (NAFLD score > 0.676 or Fibrosis-4 [FIB-4] score > 2.67) across ASCVD risk (low < 7.5%; intermediate 7.5% to < 20%; high ≥ 20%). This retrospective study comprised 20,772 participants in an employer-sponsored health assessment. High MPO associated with impaired kidney function with low (OR 2.2, 95% CI 1.6-3.7) and intermediate (OR 2.0, 95% CI 1.3-3.5) ASCVD risk, and with high FIB-4 or NAFLD scores in low (OR 2.4, 95% CI 1.2-4.7), intermediate (OR 3.1, 95% CI 2.0-6.0), and high (OR 3.8, 95% CI 2.9-7.4) ASCVD risk groups. High MPO was associated with markers of CKD and liver fibrosis in low to intermediate ASCVD risk and treated groups. These findings demonstrate the commonality of cardiometabolic biomarkers across multiple organs. Prospective studies are warranted to assess whether high MPO levels identify persons at risk for CKD and liver fibrosis who may benefit from preventive strategies.
Collapse
Affiliation(s)
- Olga A Iakoubova
- Quest Diagnostics, 33608 Ortega Highway, San Juan Capistrano, CA, 92675, USA
| | | | - Judy Z Louie
- Quest Diagnostics, 33608 Ortega Highway, San Juan Capistrano, CA, 92675, USA
| | - Charles M Rowland
- Quest Diagnostics, 33608 Ortega Highway, San Juan Capistrano, CA, 92675, USA
| | - Andre R Arellano
- Quest Diagnostics, 33608 Ortega Highway, San Juan Capistrano, CA, 92675, USA
| | - Lance A Bare
- Quest Diagnostics, 33608 Ortega Highway, San Juan Capistrano, CA, 92675, USA
| | - Charles E Birse
- Quest Diagnostics, 33608 Ortega Highway, San Juan Capistrano, CA, 92675, USA
| | - Marc S Penn
- Summa Health Heart and Vascular Institute, Summa Health, 525 E. Market St, Akron, OH, 44304, USA.
| |
Collapse
|
23
|
Bay B, Tanner R, Gao M, Oliva A, Sartori S, Vogel B, Gitto M, Smith KF, Di Muro FM, Hooda A, Sweeny J, Krishnamoorthy P, Moreno P, Krishnan P, Dangas G, Kini A, Sharma SK, Mehran R. Residual cholesterol and inflammatory risk in statin-treated patients undergoing percutaneous coronary intervention†. Eur Heart J 2025:ehaf196. [PMID: 40208236 DOI: 10.1093/eurheartj/ehaf196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/03/2024] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND AND AIMS Elevated LDL-cholesterol levels and inflammation, as assessed by high-sensitivity C-reactive protein, correlate with cardiovascular risk. However, data on the relative impact of residual LDL-cholesterol and inflammatory risk among statin-treated patients undergoing percutaneous coronary intervention (PCI) is lacking. Hence, this study aimed to investigate the impact of residual cholesterol/inflammatory risk in patients on statin therapy undergoing PCI. METHODS From 2012 to 2022, patients at a tertiary centre undergoing PCI were analysed. Patients were stratified according to LDL-cholesterol (≥70 vs <70 mg/dL) and high-sensitivity C-reactive protein (≥2 vs <2 mg/L) levels: no residual cholesterol or inflammatory risk, residual cholesterol risk, residual inflammatory risk, and combined residual cholesterol and inflammatory risk. Patients presenting with acute myocardial infarction, cancer, no statin treatment at admission, or high-sensitivity C-reactive protein levels >10 mg/L were excluded. The primary endpoint was major adverse cardiovascular events (MACEs), defined as the composite of all-cause mortality, spontaneous myocardial infarction, and stroke 1 year after the index PCI. RESULTS A total of 15 494 patients were included. After 1-year follow-up, individuals with isolated residual inflammatory risk had the highest MACE rate (5.1%), followed by patients with combined cholesterol and inflammatory risk, no residual risk, and isolated residual cholesterol risk. After multivariable Cox regression analysis, patients with residual inflammatory risk had a 1.8-fold higher risk for MACE (adjusted hazard ratio: 1.78, 95% confidence interval 1.36-2.33, P < .001) compared with those with no residual cholesterol or inflammatory risk. This was similar in patients with combined residual cholesterol and inflammatory risk (adjusted hazard ratio: 1.56, 95% confidence interval 1.19-2.04, P = 0.001). Of note, no independent association of isolated residual cholesterol risk (adjusted hazard ratio: 1.01, 95% confidence interval .76-1.35, P-value = .920) with MACE was noted (P-trend across all groups <.001). CONCLUSIONS Among statin-treated patients undergoing PCI, residual inflammation but not cholesterol risk was associated with an increased risk of MACE during follow-up.
Collapse
Affiliation(s)
- Benjamin Bay
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Richard Tanner
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
- Department of Cardiology, Cork University Hospital, Cork, Ireland
| | - Michael Gao
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Angelo Oliva
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Samantha Sartori
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Birgit Vogel
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Mauro Gitto
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Kenneth F Smith
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Francesca Maria Di Muro
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
- Structural Interventional Cardiology Division, Department of Cardiac Thoracic and Vascular Medicine, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Amit Hooda
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Joseph Sweeny
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Parasuram Krishnamoorthy
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Pedro Moreno
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Prakash Krishnan
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - George Dangas
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Annapoorna Kini
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Samin K Sharma
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Roxana Mehran
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| |
Collapse
|
24
|
Alcover S, Ramos-Regalado L, Girón G, Muñoz-García N, Vilahur G. HDL-Cholesterol and Triglycerides Dynamics: Essential Players in Metabolic Syndrome. Antioxidants (Basel) 2025; 14:434. [PMID: 40298782 PMCID: PMC12024175 DOI: 10.3390/antiox14040434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Metabolic syndrome (MetS) is a complex cluster of interrelated metabolic disorders that significantly elevate the risk of cardiovascular disease, making it a pressing public health concern worldwide. Among the key features of MetS, dyslipidemia-characterized by altered levels of high-density lipoprotein cholesterol (HDL-C) and triglycerides (TG)-plays a crucial role in the disorder's progression. This review aims to elucidate the intricate interplay between HDL-C and TG within the context of lipid metabolism and cardiovascular health, while also addressing the detrimental impact of various cardiovascular risk factors and associated comorbidities. The dynamics of HDL-C and TG are explored, highlighting their reciprocal relationship and respective contributions to the pathophysiology of MetS. Elevated levels of TGs are consistently associated with reduced concentrations of HDL-C, resulting in a lipid profile that promotes the development of vascular disease. Specifically, as TG levels rise, the protective cardiovascular effects of HDL-C are diminished, leading to the increased accumulation of pro-atherogenic TG-rich lipoproteins and low-density lipoprotein particles within the vascular wall, contributing to the progression of atheromas, which can ultimately result in significant ischemic cardiovascular events. Ultimately, this paper underscores the significance of HDL and TG as essential targets for therapeutic intervention, emphasizing their potential in effectively managing MetS and reducing cardiovascular risk.
Collapse
Affiliation(s)
- Sebastià Alcover
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Lisaidy Ramos-Regalado
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Gabriela Girón
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Natàlia Muñoz-García
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Gemma Vilahur
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
25
|
Song B, Sun L, Qin X, Fei J, Yu Q, Chang X, He Y, Liu Y, Shi M, Guo D, Shen O, Zhu Z. Associations of Lipid-Lowering Drugs With Blood Pressure and Fasting Glucose: A Mendelian Randomization Study. Hypertension 2025; 82:743-751. [PMID: 39902581 DOI: 10.1161/hypertensionaha.124.23829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Observational studies have linked LDL-C (low-density lipoprotein-cholesterol)-lowering drugs with lower blood pressure (BP) and higher fasting glucose, but the causality remains unclear. We conducted a drug target Mendelian randomization study to assess the causal associations of genetically proxied inhibition of HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase), PCSK9 (proprotein convertase subtilisin/kexin type 9), and NPC1L1 (Niemann-Pick C1-Like 1) with BP and fasting glucose. METHODS Single-nucleotide polymorphisms in HMGCR, NPC1L1, and PCSK9 associated with LDL-C in a genome-wide association study meta-analysis from the Global Lipid Genetics Consortium (173 082 European individuals) were used to proxy LDL-C-lowering drug targets. BP and fasting glucose data were obtained from genome-wide association studies conducted by the International Consortium of Blood Pressure (757 601 European participants) and the Glucose and Insulin-related Traits Consortium (58 074 European participants). We used the inverse-variance weighted method and a series of sensitivity analyses for assessment. RESULTS Genetically proxied inhibition of HMGCR was negatively associated with systolic BP (β, -0.81 [95% CI, -1.26 to -0.37 mm Hg]; P=3.72×10-4) and diastolic BP (β, -1.58 [95% CI, -2.24 to -0.91 mm Hg]; P=3.23×10-6). Conversely, we observed a positive association between genetically proxied inhibition of HMGCR and high fasting glucose (β, 0.13 [95% CI, 0.08-0.17 mmol/L]; P=4.25×10-8). However, there was no association of PCSK9 and NPC1L1 inhibition with BP or fasting glucose. CONCLUSIONS Genetically proxied inhibition of HMGCR was significantly associated with low BP and high fasting glucose, while there was no effect of PCSK9 and NPC1L1 inhibition on BP or fasting glucose.
Collapse
Affiliation(s)
- Beiping Song
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Lulu Sun
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Xiaoli Qin
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Jiawen Fei
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Quan Yu
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Xinyue Chang
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Yu He
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Yi Liu
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Mengyao Shi
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (M.S., Z.Z.)
| | - Daoxia Guo
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Ouxi Shen
- Department of Occupational Health, Suzhou Industrial Park Center for Disease Control and Prevention, China (O.S.)
| | - Zhengbao Zhu
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (M.S., Z.Z.)
| |
Collapse
|
26
|
Ling Y, Yuan S, Cheng H, Tan S, Huang X, Tang Y, Bai Z, Li R, Li L, Li S, Huang L, Xu A, Lyu J. Exploring the Link Between C-Reactive Protein Change and Stroke Risk: Insights From a Prospective Cohort Study and Genetic Evidence. J Am Heart Assoc 2025; 14:e038086. [PMID: 40135578 DOI: 10.1161/jaha.124.038086] [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: 08/03/2024] [Accepted: 12/17/2024] [Indexed: 03/27/2025]
Abstract
BACKGROUND Previous research on how changes in CRP (C-reactive protein) levels predict stroke risk is limited. This study aimed to examine the association between CRP change and the risk of stroke and its subtypes. METHODS AND RESULTS Based on the UK Biobank data, we investigated the association between CRP change and the risk of stroke and its subtypes with Cox proportional hazards regression analysis. We further performed genetic analyses including genetic correlation, pairwise genome-wide association study, and polygenic risk score. Our study involved 14 754 participants with a median follow-up time of 10.4 years. After categorizing participants by CRP percentage change and making adjustments for potential confounders, it was observed that those with an elevated percentage of CRP change had a higher risk of any stroke (hazard ratio [HR], 1.44 [95% CI, 1.12-1.85]) and ischemic stroke (HR, 1.65 [95% CI, 1.24-2.18]). After categorization by CRP change types and adjustment for confounders, the group that became high level had a higher any-stroke risk (HR, 1.45 [95% CI, 1.04-2.02]), with the group that remained at a high level facing the greatest risk (HR, 1.74 [95% CI, 1.30-2.33]). Similar trends were observed for ischemic stroke. The group that remained at a high level also had a heightened hemorrhagic stroke risk (HR, 1.91 [95% CI, 1.07-3.44]). Genetic analysis showed a significant genetic correlation between CRP and stroke (rg, 0.257; rg_P=2.39E-07). Pairwise genome-wide association study analysis identified 5 shared genomic regions between CRP and stroke. Polygenic risk score analysis showed that participants with high stroke polygenic risk score and elevated or remaining high CRP levels have the highest risk of stroke. CONCLUSIONS Both any stroke and ischemic stroke are related to elevated and remaining high CRP levels, while hemorrhagic stroke is only related to remaining high CRP levels.
Collapse
Affiliation(s)
- Yitong Ling
- Department of Neurology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Shiqi Yuan
- Department of Neurology The Second People's Hospital of Guiyang City Guiyang Guizhou China
| | - Hongtao Cheng
- School of Nursing Jinan University Guangzhou Guangdong China
- School of Nursing Sun Yat-sen University Guangzhou China
| | - Shanyuan Tan
- Department of Neurology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Xiaxuan Huang
- Department of Anesthesiology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Yonglan Tang
- School of Nursing Jinan University Guangzhou Guangdong China
| | - Zihong Bai
- Department of Neurology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Rui Li
- Department of Neurology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Li Li
- Department of Clinical Research Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Shuna Li
- Department of Clinical Research Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Liying Huang
- Department of Clinical Research Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Anding Xu
- Department of Neurology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Jun Lyu
- Department of Clinical Research Jinan University First Affiliated Hospital Guangzhou Guangdong China
- Key Laboratory of Regenerative Medicine of Ministry of Education Guangzhou Guangdong China
| |
Collapse
|
27
|
Ravi A, Koyama S, Cho SMJ, Haidermota S, Hornsby W, Ellinor PT, Natarajan P. Genetic Predisposition to Low-Density Lipoprotein Cholesterol and Incident Type 2 Diabetes. JAMA Cardiol 2025; 10:379-383. [PMID: 39813027 PMCID: PMC11983226 DOI: 10.1001/jamacardio.2024.5072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/07/2024] [Indexed: 01/16/2025]
Abstract
Importance Treatment to lower high levels of low-density lipoprotein cholesterol (LDL-C) reduces incident coronary artery disease (CAD) risk but modestly increases the risk for incident type 2 diabetes (T2D). The extent to which genetic factors across the cholesterol spectrum are associated with incident T2D is not well understood. Objective To investigate the association of genetic predisposition to increased LDL-C levels with incident T2D risk. Design, Setting, and Participants In this large prospective, population-based cohort study, UK Biobank participants who underwent whole-exome sequencing and genome-wide genotyping were included. Participants were separated into 7 groups with familial hypercholesterolemia (FH), predicted loss of function (pLOF) in APOB or PCSK9 variants, and LDL-C polygenic risk score (PRS) quintiles. Data were collected between 2006 and 2010, with a median follow-up of 13.7 (IQR, 12.9-14.5) years. Data were analyzed from March 1 to November 1, 2024. Exposures LDL-C level, LDL-C PRS, FH, or pLOF variant status. Main Outcomes and Measures Cox proportional hazards regression models adjusted for age, sex, genotyping array, lipid-lowering medication use, and the first 10 genetic principal components were fitted to assess the association between LDL-C genetic factors and incident T2D and CAD risks. Results Among the 361 082 participants, mean (SD) age was 56.8 (8.0) years, 194 751 (53.9%) were female, and mean (SD) baseline LDL-C level was 138.0 (33.6) mg/dL. During the follow-up period, 22 619 (6.3%) participants developed incident T2D and 17 966 (5.0%) developed incident CAD. The hazard ratio for incident T2D was lowest in the FH group (0.65; 95% CI, 0.54-0.77), while the highest risk was in the pLOF group (1.48; 95% CI, 1.18-1.86). The association between LDL-C PRS and incident T2D was 0.72 (95% CI, 0.66-0.79) for very high LDL-C PRS, 0.87 (95% CI, 0.84-0.90) for high LDL-C PRS, 1.13 (95% CI, 1.09-1.17) for low LDL-C PRS, and 1.26 (95% CI, 1.15-1.38) for very low LDL-C PRS. CAD risk increased directly with the LDL-C PRS. Conclusions and Relevance In this cohort study, LDL-C and T2D risks were inversely associated across genetic mechanisms for LDL-C variation. Further elucidation of the mechanisms associating low LDL-C risk with increased risk of T2D is warranted.
Collapse
Affiliation(s)
- Akshaya Ravi
- Program of Medical and Population Genetics, Broad Institute of MIT (Massachusetts Institute of Technology) and Harvard, Cambridge, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Satoshi Koyama
- Program of Medical and Population Genetics, Broad Institute of MIT (Massachusetts Institute of Technology) and Harvard, Cambridge, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - So Mi Jemma Cho
- Program of Medical and Population Genetics, Broad Institute of MIT (Massachusetts Institute of Technology) and Harvard, Cambridge, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sara Haidermota
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Whitney Hornsby
- Program of Medical and Population Genetics, Broad Institute of MIT (Massachusetts Institute of Technology) and Harvard, Cambridge, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Patrick T. Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Pradeep Natarajan
- Program of Medical and Population Genetics, Broad Institute of MIT (Massachusetts Institute of Technology) and Harvard, Cambridge, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
28
|
Wolska M, Peruzzi M, Kaziród-Wolski K, Wróbel P, Oleś I, Sielski J, Jankowski P. Risk factors for cardiovascular diseases: the focus on primary prevention. Minerva Cardiol Angiol 2025; 73:245-253. [PMID: 37971709 DOI: 10.23736/s2724-5683.23.06360-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Views on the etiopathogenesis of atherosclerosis are subject to evolution. In addition to the classic well-known risk factors, new ones related to mental state, social life and environment are being discovered. Both acute and chronic stress stimulate inflammatory processes. Due to the change in lifestyle and eating habits, the accumulation of risk factors in childhood is an increasing problem. Knowledge of risk factors allows for effective primary prevention of cardiovascular diseases. The effectiveness of prevention increases when the activities cover the largest possible part of the society, and access to a doctor is easy. Therefore, government programs are being implemented offering patients easier access to diagnostics of cardiovascular diseases at the level of primary health care, which enables faster identification of people at the greatest cardiovascular risk. Easier access to primary care and a good doctor-patient relationship improve patient compliance. In this situation, the importance of the family doctor as a key link in the diagnosis, prevention and treatment of cardiovascular diseases is increasing.
Collapse
Affiliation(s)
| | - Mariangela Peruzzi
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University, Rome, Italy -
- Mediterranea Cardiocentro, Naples, Italy
| | - Karol Kaziród-Wolski
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Paweł Wróbel
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Izabela Oleś
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Janusz Sielski
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Piotr Jankowski
- Department of Epidemiology and Health Promotion, School of Public Health, Center of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
29
|
Manning ES, Shroff GR, Jacobs DR, Duprez DA. Chronic Inflammatory-Related Disease and Cardiovascular Disease in MESA. JACC. ADVANCES 2025; 4:101640. [PMID: 40037010 PMCID: PMC11925088 DOI: 10.1016/j.jacadv.2025.101640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 01/20/2025] [Accepted: 01/30/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Inflammation plays a role in cardiovascular disease (CVD). We defined various noncardiovascular and noncancer conditions, both infectious and noninfectious, with a common basis of inflammation, collectively termed chronic inflammatory-related disease (ChrIRD). We describe ChrIRD and its interplay with CVD during follow-up in the Multi-Ethnic Study of Atherosclerosis. OBJECTIVES The aim of the study was to describe ChrIRD, its associations with CVD, and its association with mortality. METHODS Participants were free of overt CVD at baseline with median 17.9 (Q1-Q3: 14.9-18.6) years of follow-up. ChrIRD was determined by review of hospitalization and death records of International Classification of Diseases codes. CVD diagnosis was adjudicated based on medical records. We performed time-dependent proportional hazard regressions to identify risks related to ChrIRD or CVD events. RESULTS MESA (Multi-Ethnic Study of Atherosclerosis) participants (n = 6,791) had a mean age of 62 ± 10 years, with 47% (3,201/6,791) men, 39% (2,617/6,791) White, 28% (1,882/6,791) Black, 22% (1,489/6,791) Hispanic, and 12% (803/6,791) Chinese race/ethnicity. ChrIRD was observed in 29% (1,965/6,791) and CVD in 21% (1,420/6,791); including 11% (761/6,791) with both conditions. Mortality after ChrIRD only was 47% (567/1,204; 95% CI: 44%-49%); after CVD only was 45% (300/659; 95% CI: 41%-49%); and after both conditions was 67% (510/761; 95% CI: 63%-70%). CVD was associated with increased risk of ChrIRD (HR: 1.48; 95% CI: 1.23-1.77) and ChrIRD was associated with increased risk of CVD (HR: 2.23; 95% CI: 1.97-2.52). Baseline inflammatory markers predicted both conditions. CONCLUSIONS ChrIRD is common, present in all organ systems, and is associated with significant mortality, particularly in combination with CVD. The association between CVD and ChrIRD is bidirectional, and baseline inflammatory markers are associated with ChrIRD and CVD.
Collapse
Affiliation(s)
- Evan S Manning
- Cardiovascular Division, Department of Medicine, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Gautam R Shroff
- Division of Cardiology, Department of Medicine, Hennepin Healthcare, Minneapolis, Minnesota, USA; University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - David R Jacobs
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel A Duprez
- Cardiovascular Division, Department of Medicine, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
30
|
Cha JJ, Kim JH, Hong SJ, Lim S, Joo HJ, Park JH, Yu CW, Lee PH, Lee SW, Lee CW, Moon JY, Lee JY, Kim JS, Park JS, Lim DS. Safety and efficacy of moderate-intensity statin with ezetimibe in elderly patients with atherosclerotic cardiovascular disease. J Intern Med 2025; 297:400-408. [PMID: 39709592 DOI: 10.1111/joim.20029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2024]
Abstract
BACKGROUND High-intensity statin therapy significantly reduces mortality and cardiovascular events in patients with atherosclerotic cardiovascular disease (ASCVD). However, moderate-intensity statins are often preferred for elderly patients due to their higher risk of intolerance to high-intensity statins. OBJECTIVE To compare the incidence of statin-associated muscle symptoms (SAMS) and the effect on low-density lipoprotein cholesterol (LDL-C) levels between elderly ASCVD patients receiving high-intensity statin monotherapy and those receiving moderate-intensity statin with ezetimibe in a combination therapy. METHOD In a prospective, multicenter, open-label trial conducted in South Korea, 561 patients aged 70 years or above with ASCVD were randomly assigned to receive either moderate-intensity statin with ezetimibe combination therapy (rosuvastatin 5 mg with ezetimibe 10 mg) or high-intensity statin monotherapy (rosuvastatin 20 mg) over 6 months. The primary endpoint was the incidence of SAMS, and the key secondary endpoint was the achievement of target LDL-C levels (<70 mg/dL) within 6 months. RESULTS The primary endpoint showed a lower incidence of SAMS in the combination therapy group (0.7%) compared to the high-intensity statin monotherapy group (5.7%, p = 0.005). Both groups achieved similar LDL-C levels, with 75.4% in the combination therapy group and 68.7% in the monotherapy group reaching target levels. CONCLUSION Moderate-intensity statin with ezetimibe combination therapy offers a lower risk of SAMS and similar LDL-C reduction in elderly patients with ASCVD, compared to high-intensity statin monotherapy.
Collapse
Affiliation(s)
- Jung-Joon Cha
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Ju Hyeon Kim
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Soon Jun Hong
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Subin Lim
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Hyung Joon Joo
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Jae Hyoung Park
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Cheol Woong Yu
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Pil Hyung Lee
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seung Whan Lee
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Cheol Whan Lee
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae Youn Moon
- Department of Cardiology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jong-Young Lee
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jung-Sun Kim
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Suk Park
- Division of Cardiology, Incheon Sejong Hospital, Incheon, South Korea
| | - Do-Sun Lim
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
31
|
Barnett MJ, Casipit C, Lam J, Rivadeneira A. Statins and Thyroid Eye Disease: A Propensity Score-Matched Retrospective Cohort Analysis. Cureus 2025; 17:e83243. [PMID: 40309510 PMCID: PMC12042245 DOI: 10.7759/cureus.83243] [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] [Accepted: 04/30/2025] [Indexed: 05/02/2025] Open
Abstract
Background Thyroid eye disease is a sight-threatening disorder associated with Graves's disease. While treatment is aimed at stabilizing established thyroid eye disease, there are limited investigations regarding prevention. Statins, known for their anti-inflammatory properties, are one such treatment that has been investigated for thyroid eye disease. This study retrospectively analyzes the impact of statin medications on thyroid eye disease and related outcomes from real-world data using TriNetX Global Collaborative Network (TriNetX, Cambridge, MA, USA). Objective The primary objective of this study is to analyze the risk of developing thyroid eye disease in patients with Graves' disease treated with statin medications relative to those without exposure up to five years. Secondary objectives of this study include analyzing thyroid eye disease-related outcomes, including diplopia, optic nerve surgery, optic nerve injury, strabismus, blindness, requirement for teprotumumab, color vision deficiencies, and referral to ophthalmology. Methods Data were collated utilizing TriNetX Global Collaborative Network, a platform of globally de-identified patient information from over 140 healthcare organizations. Patients more than 18 years of age with a diagnosis of Graves' disease (ICD-10: E05.0 Thyrotoxicosis with Diffuse Goiter) were divided into Cohorts A (administration of statin medication following diagnosis of Graves' disease, n = 66,444) and B (no statin medication administration, n = 225,905). Via implementation of propensity score matching to control for confounding factors, a total of n = 43,025 matched patients per cohort was obtained. Hazard ratios (HRs) and 95% confidence intervals (CIs) were used to assess the primary and secondary objectives over a five-year follow-up period. The t-test was used for the calculation of mean averages of ophthalmology referrals and to determine statistical significance. Results Patients treated with statin medications (Cohort A) demonstrated a significantly lower HR for the development of exophthalmos (HR = 0.559, 95% CI 0.521-0.601, p < 0.001). Statin medications were furthermore associated with a reduced HR for diplopia (p < 0.001), optic nerve surgery (p = 0.018), and requirement for teprotumumab (p = 0.047). No significant reduction in hazards for color vision deficits, strabismus, optic nerve injury, or blindness was noted. Additionally, there was no significant difference in referrals to ophthalmology. Conclusion The findings from this study suggest that statin therapy may be protective against the development of thyroid eye disease and associated complications. Further randomized controlled trials are required to validate these findings and establish a therapeutic role for statins in thyroid eye disease.
Collapse
Affiliation(s)
- Maxim J Barnett
- Internal Medicine, Jefferson Einstein Hospital, Philadelphia, USA
| | - Carlo Casipit
- Internal Medicine, Jefferson Einstein Hospital, Philadelphia, USA
| | - Justin Lam
- Internal Medicine, Jefferson Einstein Hospital, Philadelphia, USA
| | - Ana Rivadeneira
- Endocrinology and Diabetes, Jefferson Einstein Hospital, Philadelphia, USA
| |
Collapse
|
32
|
Treihaft AM, Parikh MA, Jackson KA, Frishman WH, Peterson SJ. New Therapies for the Management of Chronic Kidney Disease. Cureus 2025; 17:e81824. [PMID: 40337581 PMCID: PMC12057290 DOI: 10.7759/cureus.81824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2025] [Indexed: 05/09/2025] Open
Abstract
A major public health concern gripping the nation is chronic kidney disease (CKD), and for individuals concomitantly diagnosed with type 2 diabetes mellitus (T2DM), the coexistence significantly increases the cardiovascular morbidity and mortality by two to three times higher than patients diagnosed without CKD. CKD management encompasses both non-pharmacological approaches, such as dietary sodium restriction and lifestyle modification for blood pressure control, and pharmacological approaches. Current pharmacological management focuses on four key pillars: renin-angiotensin system inhibitors (RASi), sodium-glucose cotransporter-2 inhibitors (SGLT2i), glucagon-like peptide-1 receptor agonists (GLP-1RA), and mineralocorticoid receptor antagonists (MRAs), all of which have shown renoprotective and cardiovascular benefits. An incomplete block of aldosterone activity remains a challenge and is one of the factors contributing to the progression of kidney damage. Aldosterone synthase inhibitors (ASIs), such as vicadrostat, may represent a new horizon in selectively inhibiting aldosterone synthesis while preserving cortisol production. Early-phase trials have shown reductions in albuminuria and a potential for renal protection. The question is, could ASIs emerge as a fifth pillar in CKD management and help curb the progression?
Collapse
Affiliation(s)
- Andrew M Treihaft
- Department of Medicine, New York-Presbyterian Brooklyn Methodist Hospital, Brooklyn, USA
| | - Manish A Parikh
- Department of Medicine, New York-Presbyterian Brooklyn Methodist Hospital, Brooklyn, USA
- Department of Medicine, Weill Cornell Medicine, New York, USA
| | - Kaedrea A Jackson
- Department of Emergency Medicine, New York-Presbyterian Brooklyn Methodist Hospital, Brooklyn, USA
- Department of Emergency Medicine, Weill Cornell Medicine, New York, USA
| | | | - Stephen J Peterson
- Department of Medicine, New York-Presbyterian Brooklyn Methodist Hospital, Brooklyn, USA
- Department of Medicine, Weill Cornell Medicine, New York, USA
| |
Collapse
|
33
|
Perdomo-Celis F, Passaes C, Monceaux V, Lambotte O, Costagliola D, Chevalier MF, Weiss L, Sáez-Cirión A. Impact of rosuvastatin on the memory potential and functionality of CD8 + T cells from people with HIV. EBioMedicine 2025; 114:105672. [PMID: 40158388 PMCID: PMC11995788 DOI: 10.1016/j.ebiom.2025.105672] [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: 12/02/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Virus-specific CD8+ T cells play a major role in the natural control of HIV infection, linked to memory-like features such as high survival capacity and polyfunctionality. However, virus-specific CD8+ T cells from HIV non-controllers exhibit an effector-like and exhausted profile, with limited antiviral potential. Metabolic reprogramming of cells from non-controllers could reinvigorate their functional capacities. Considering the implication of the cholesterol pathway in the induction of T cell exhaustion, here we evaluated the impact of rosuvastatin, an inhibitor of cholesterol synthesis, on the functionality and memory profile of HIV-specific CD8+ T cells from people on antiretroviral treatment. METHODS We analysed samples from 10 individuals with HIV-1 on ART who participated in the IMEA 043-CESAR trial and received rosuvastatin for 12 weeks. We explored whether rosuvastatin treatment was accompanied by changes in the memory potential of CD8+ T cells. We evaluated the phenotype and functionality of total and HIV-specific CD8+ T cells before, during, and after treatment with rosuvastatin. A mixed effects model was used for repeated measures and corrected for multiple comparisons. FINDINGS Total and HIV-specific CD8+ T cell survival and functionality were enhanced in individuals who received a 12-week course of rosuvastatin, with a consistent increase in polyfunctional IFN-γ+ TNF-α+ cells. The superior CD8+ T cell functionality after rosuvastatin treatment was associated with intrinsic metabolic changes, including the decrease of fatty acid uptake, as well as a reduction in effector/exhaustion markers. Changes in the characteristics of CD8+ T cells coincided with the duration of rosuvastatin administration, and most effects waned after the cessation of the treatment. INTERPRETATION CD8+ T cell metabolic reprogramming by targeting the cholesterol pathway, combined with other available immunotherapies, might represent a promising strategy in the search for the cure of HIV or other chronic viral infections. FUNDING The CESAR trial was sponsored by IMEA. This work was supported by the NIH (grants UM1AI164562 and R01DK131476).
Collapse
Affiliation(s)
- Federico Perdomo-Celis
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, 75015, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistance Unit, Paris, 75015, France; Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Caroline Passaes
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, 75015, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistance Unit, Paris, 75015, France
| | - Valérie Monceaux
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, 75015, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistance Unit, Paris, 75015, France
| | - Olivier Lambotte
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological, Bacterial Diseases (IMVA-HB/IDMIT/UMRS1184), Le Kremlin Bicêtre, Fontenay aux Roses, France; Assistance Publique Hôpitaux de Paris, Groupe Hospitalier Universitaire Paris Saclay, Service de Médecine interne immunologie clinique, Le Kremlin Bicêtre, France
| | - Dominique Costagliola
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (IPLESP), Paris, France
| | - Mathieu F Chevalier
- INSERM UMR 1342, Institut de Recherche Saint-Louis (IRSL), Université Paris Cité, Paris, France
| | - Laurence Weiss
- Université de Paris Cité, AP-HP, Paris Centre, Paris, France
| | - Asier Sáez-Cirión
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, 75015, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistance Unit, Paris, 75015, France.
| |
Collapse
|
34
|
Bradley NA, Walter A, Sankey C, Wilson A, Siddiqui T, Roxburgh CS, McMillan DC, Guthrie GJ. Longitudinal association between statins and changes in CT-derived body composition in patients with abdominal aortic aneurysm. J Cachexia Sarcopenia Muscle 2025; 16:10.1002/jcsm.13565. [PMID: 40204359 PMCID: PMC11981686 DOI: 10.1002/jcsm.13565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Loss of skeletal muscle mass and systemic inflammation may offer prognostic value in patients with abdominal aortic aneurysm (AAA). The longitudinal progression of abnormal body composition parameters and their determinants is poorly reported. Statins are widely used medications that improve the prognosis of cardiovascular disease and interact with both muscle tissue and systemic inflammation. The present study aimed to describe the association between statin therapy and both pre-operative and longitudinal CT-derived body composition in patients undergoing elective intervention for AAA. METHODS A total of 756 consecutive patients undergoing elective intervention for AAA at three centres were retrospectively recruited. Body composition analysis was performed on pre-operative and follow-up CTs at L3 to generate subcutaneous adipose tissue index, visceral adipose tissue index and skeletal muscle index and density (SMI and SMD). Systemic inflammation was assessed using the systemic inflammatory grade. RESULTS A total of 756 patients (702 [93%] males, median [interquartile range, IQR] age 73.0 [11.0] years) were included, with a median (IQR) follow-up of 67.0 (32) months and 235 deaths during the follow-up period. There were 582 patients (77%) receiving statin therapy and 174 patients (23%) not receiving statin therapy. Follow-up CTs were available for 273 patients. From pre-operative to follow-up CTs, there was a decrease in median SMI (P < 0.001) and SMD (P < 0.001) and an increase in the comparative prevalences of low SMI (43% vs. 50%, P < 0.01) and low SMD (64% vs. 88%, P < 0.001). There were no differences in baseline clinicopathological characteristics, systemic inflammation or pre-operative CT-derived body composition parameters between patients with and without >10% loss of skeletal muscle mass. In patients with ≤10% loss of SMI, mean (95% confidence interval) survival was 91.6 (87.2-95.9) months versus 89.3 (80.4-98.2) months in patients with >10% loss of SMI (P = 0.58). Patients receiving statin therapy had a higher American Society of Anesthesiologists grade (P < 0.001), a higher body mass index (BMI) (P < 0.05) and a greater prevalence of normal pre-operative SMI (P < 0.001). CONCLUSIONS In patients with AAA, skeletal muscle mass and density appear to progressively decline despite treatment of AAA, though specific determinants of this are uncertain, and statin use does not appear to predispose to either muscle loss or preservation. Statin therapy appears to be associated with a lower rate of pre-operative low skeletal muscle mass, despite greater comorbidity and BMI. Further investigation of the progressive changes in muscle mass and quality, statin therapy and systemic inflammation is warranted.
Collapse
|
35
|
Kholaif N, Batha L, Elmahi I, Alnaser S, Alzaher S, Almallohi N, Alhussein M, Alhalees D, Alshehri A. Transplantation of heterozygous familial hypercholesterolemia living donor liver resulting in early myocardial infarction: a possible dangerous link. Monaldi Arch Chest Dis 2025; 95. [PMID: 38226645 DOI: 10.4081/monaldi.2024.2907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 01/14/2024] [Indexed: 01/17/2024] Open
Abstract
Living donor liver transplantation (LDLT) is a lifesaving procedure that is often curative for several liver diseases. Familial hypercholesterolemia (FH) is a metabolic disease that results from an autosomal dominant mutation in the low-density lipoprotein receptor; yet, young patients with FH can live years without detection. Herein, we report a case of a patient who developed early myocardial infarction (MI) after having a transplant from a donor with undetected heterozygous FH. The patient was a 67-year-old female with non-alcoholic steatohepatitis-related liver cirrhosis, free from coronary artery disease, who underwent LDLT from her daughter, a 45-year-old female with no past medical history. One year post-transplantation, she presented with an acute MI with a large atherosclerotic burden. Genetic analysis confirmed heterozygous FH in the donor but not in the recipient. This case emphasizes the importance of incorporating a thorough clinical history and lipid profile into pre-transplant testing for both the recipient and donor, as well as aggressive lipid-lowering therapy post-transplantation to avoid cardiovascular complications.
Collapse
Affiliation(s)
- Naji Kholaif
- Heart Center, King Faisal Specialist Hospital and Research Center, Riyadh; College of Medicine, Alfaisal University, Riyadh
| | - Lin Batha
- College of Medicine, Alfaisal University, Riyadh
| | - Isra Elmahi
- College of Medicine, Alfaisal University, Riyadh
| | - Sulaiman Alnaser
- Heart Center, King Faisal Specialist Hospital and Research Center, Riyadh
| | - Sultan Alzaher
- Heart Center, King Faisal Specialist Hospital and Research Center, Riyadh
| | - Norah Almallohi
- Department of Anesthesiology, King Faisal Specialist Hospital and Research Center, Riyadh
| | - Mosaad Alhussein
- Heart Center, King Faisal Specialist Hospital and Research Center, Riyadh; College of Medicine, King Saud University, Riyadh
| | | | - Ahmed Alshehri
- Heart Center, King Faisal Specialist Hospital and Research Center, Riyadh
| |
Collapse
|
36
|
Chu Z, Yue W, Mu Q, Xu D, Chang Z, Liang M, Geng Y, Ding P. Effects of statin use on blood pressure and other hypertension-related outcome indicators in hypertensive patients: A systematic review and meta-analysis. Prostaglandins Other Lipid Mediat 2025; 178:106991. [PMID: 40174858 DOI: 10.1016/j.prostaglandins.2025.106991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/19/2024] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
AIMS This meta-analysis delved into the impact of statin therapy, both as a monotherapy and in conjunction with antihypertensive medications, on blood pressure levels and outcomes pertinent to hypertension. METHODS We searched the PubMed, EMBASE, and Cochrane databases for studies published before October 1 2023. Studies designed as cohort studies or randomized controlled trials and investigating the effects of single use of statin or its combined use with other antihypertensive therapy were included. Authors extracted the data independently and differences were decided through discussion. Random-effects model was used to evaluate the merged outcomes. Due to the high heterogeneity of the HDL-C group, we performed subgroup analysis according to the type of statin. To assess the robustness and potential publication bias of our findings, we utilized sensitivity analysis, Egger's test, and funnel plots. RESULTS 23 trials were included in this meta-analysis. The primary outcomes revealed that administering statins did not significantly impact the systolic pressure (SBP) of hypertensive patients (MD, -1.77; 95 % CI, -4.82-1.27). -The promoted effect of statin treatment on diastolic pressure (DBP) in hypertensive patients was found (MD, -1.87; 95 % CI, -3.72 --0.01). The secondary outcomes revealed that the use of statins resulted in a significant reduction in low-density lipoprotein (LDL-C), while significantly increasing high-density lipoprotein (HDL-C) in hypertensive patients. CONCLUSION Statin use did not modulate SBP and DBP of patients with hypertension, but SBP was decreased in the rosuvastatin or pravastatin subgroup, while DBP was decreased in the simvastatin or pravastatin subgroup. Statin treatment reduced LDL-C, increased HDL-C, and reduced the incidence of cardiovascular events and mortality compared to control groups.
Collapse
Affiliation(s)
- Zhaohan Chu
- Department of Cardiology Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| | - Wei Yue
- Department of Critical Care Medicine, 988 hospital of the PLA Joint Logistic Support Force (PLAJLSF), Zhengzhou 450000, China.
| | - Qingqing Mu
- School of Clinical Medicine, Zhengzhou University, Zhengzhou 450000, China.
| | - Dong Xu
- School of Clinical Medicine, Zhengzhou University, Zhengzhou 450000, China.
| | - Zexu Chang
- School of Clinical Medicine, Zhengzhou University, Zhengzhou 450000, China.
| | - Mengke Liang
- School of Clinical Medicine, Zhengzhou University, Zhengzhou 450000, China.
| | - Yixiao Geng
- School of Clinical Medicine, Zhengzhou University, Zhengzhou 450000, China.
| | - Ping Ding
- Department of Critical Care Medicine, 988 hospital of the PLA Joint Logistic Support Force (PLAJLSF), Zhengzhou 450000, China.
| |
Collapse
|
37
|
Rikken SAOF, Gibson CM, Bahit MC, Duffy D, Chi G, Korjian S, Mohammadnia N, White H, Anschuetz G, Kingwell BA, Ophuis TO, Nicolau JC, Lopes RD, Lewis BS, El Messaoudi S, Vinereanu D, Ten Berg JM, Goodman SG, Bode C, Steg PG, Libby P, Bainey KR, van 't Hof AWJ, Ridker PM, Mahaffey KW, Nicholls SJ, Mehran R, Harrington RA, Cornel JH. Impact of Apolipoprotein A-I Infusions on Cardiovascular Events Post-MI by Neutrophil-Lymphocyte Ratio and LDL-Cholesterol Levels. JACC. ADVANCES 2025; 4:101727. [PMID: 40288083 PMCID: PMC12059331 DOI: 10.1016/j.jacadv.2025.101727] [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: 12/16/2024] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The AEGIS-II (ApoA-I Event Reducing in Ischemic Syndromes-II; NCT03473223) trial evaluated CSL112, a human plasma-derived apolipoprotein A-I therapy, for reducing cardiovascular events after acute myocardial infarction (AMI). Given CSL112's potential anti-inflammatory properties, we conducted an exploratory post hoc analysis to determine if its efficacy is influenced by baseline neutrophil-lymphocyte ratio (NLR), a marker of systemic inflammation, and low-density lipoprotein cholesterol (LDL-C). OBJECTIVES The purpose of this study was to investigate the association of baseline NLR and cardiovascular events and explore whether NLR and LDL-C modify CSL112's efficacy in post-AMI patients. METHODS A total of 18,219 participants with AMI, multivessel coronary artery disease, and additional cardiovascular risk factors were randomized to 4 weekly infusions of 6 g CSL112 or placebo. The primary endpoint was a composite of cardiovascular death, myocardial infarction, or stroke (major adverse cardiovascular events [MACE]). Cox proportional hazards models evaluated risk by dichotomized baseline NLR (>median vs ≤median). Treatment interactions with NLR and LDL-C (≥100 vs <100 mg/dL) were assessed. RESULTS Among 15,966 participants, those with baseline NLR >median (>3.3) had a significantly greater risk of MACE at 90 days (HR: 1.40; 95% CI: 1.21-1.63), persisting at 180 and 365 days. CSL112 reduced MACE at 90 days among participants with elevated NLR and LDL-C ≥100 mg/dL (HR: 0.63; 95% CI: 0.42-0.93), with sustained benefits at 180 and 365 days. Significant interactions were observed between treatment and NLR (Pinteraction = 0.010) and among treatment, NLR, and LDL-C at 180 days (Pinteraction = 0.029). CONCLUSIONS Baseline elevated NLR predicts MACE in post-AMI patients, and CSL112 showed an associated reduction in MACE in patients with elevated NLR and LDL-C ≥100 mg/dL.
Collapse
Affiliation(s)
- Sem A O F Rikken
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | - C Michael Gibson
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| | - M Cecilia Bahit
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; INECO Neurociencias Rosario, Rosario, Argentina
| | | | - Gerald Chi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Serge Korjian
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Harvey White
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Health New Zealand Te Toka Tumai Auckland City Hospital, Auckland, New Zealand
| | | | | | | | - Jose C Nicolau
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Renato D Lopes
- Duke Clinical Research Institute, Duke Health, Durham, North Carolina, USA; Brazilian Clinical Research Institute, Sao Paulo, SP, Brazil
| | - Basil S Lewis
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Lady Davis Carmel Medical Center, Haifa, Israel; The Technion-Israel Institute of Technology, Haifa, Israel
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dragos Vinereanu
- University of Medicine and Pharmacy Carol Davila, University and Emergency Hospital, Bucharest, Romania
| | - Jurriën M Ten Berg
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; Department of Cardiology, University Medical Center Maastricht, Maastricht, the Netherlands
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Christoph Bode
- Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - P Gabriel Steg
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Université Paris-Cité, INSERM U_1148, FACT and AP-HP, Hôpital Bichat, Paris, France
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kevin R Bainey
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Walter Mackenzie Health Sciences Centre, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Arnoud W J van 't Hof
- Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; University of Medicine and Pharmacy Carol Davila, University and Emergency Hospital, Bucharest, Romania; Department of Cardiology, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kenneth W Mahaffey
- Stanford Center for Clinical Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | - Stephen J Nicholls
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| | - Roxana Mehran
- Icahn School of Medicine at Mount Sinai, Zena and Michael A. Wiener Cardiovascular Institute, New York, New York, USA
| | | | - Jan H Cornel
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands; Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| |
Collapse
|
38
|
Komnianou A, Kyriakoulis KG, Menti A, Dimakakos E, Stergiou GS, Kollias A. Cardiovascular Risk Assessment and Lipid-Lowering Therapy Recommendations in Primary Prevention. J Clin Med 2025; 14:2220. [PMID: 40217673 PMCID: PMC11989271 DOI: 10.3390/jcm14072220] [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: 01/30/2025] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Cardiovascular disease remains the leading cause of morbidity and mortality worldwide, underscoring the importance of effective primary prevention strategies. Current total cardiovascular disease (CVD) risk assessment tools, such as the Systematic Coronary Risk Evaluation 2 (SCORE2) in Europe and the Pooled cohort equations (PCEs) and Predicting Risk of CVD EVENTs (PREVENT) in the USA, aim to identify individuals at high CVD risk and guide clinical decision-making in the primary prevention setting. Statin therapy reduces cardiovascular events and is recommended as the first step for individuals with estimated CVD risk above specific thresholds. Moreover, the presence of risk modifiers, as well as the detection of asymptomatic atherosclerosis, reclassifies low-moderate CVD risk individuals into higher risk categories, contributing to tailored therapeutic decisions in primary prevention. However, differences in the performance of the available CVD risk assessment tools, the recommended thresholds for intervention, and the treatment targets by scientific societies introduce considerable inconsistency to the statin therapy practices. In addition, physicians' inertia and poor patients' adherence contribute to inadequate dyslipidemia control rates. This narrative review examines the available evidence on the current most used CVD risk assessment tools and the respective lipid-lowering recommendations, and highlights the role of targeted screening for asymptomatic atherosclerosis in terms of individualized therapy for primary prevention.
Collapse
Affiliation(s)
| | | | | | | | | | - Anastasios Kollias
- Hypertension Center STRIDE-7, School of Medicine, National and Kapodistrian University of Athens, Third Department of Medicine, Sotiria Hospital, 152 Mesogion Avenue, Athens 11527, Greece; (A.K.); (K.G.K.); (A.M.); (E.D.); (G.S.S.)
| |
Collapse
|
39
|
Cinza-Sanjurjo S, Pallarés-Carratalá V, Díaz Rodríguez A, Fierro-González D, Turégano-Yedro M, Polo-García J. [Practical approach to the patient with hypercholesterolemia in Spain. SEMERGEN position statement]. Semergen 2025; 51:102460. [PMID: 40139108 DOI: 10.1016/j.semerg.2025.102460] [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: 11/11/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 03/29/2025]
Abstract
Hypercholesterolemia, specifically the increase in the set of lipoproteins containing apolipoproteinB and, in particular, low-density lipoprotein cholesterol (LDL-C), together with the decrease in high-density lipoprotein cholesterol (HDL-C) constitute the etiopathogenic basis of atherosclerotic vascular disease. Multiple clinical trials have shown that lowering LDL-C by lipid-lowering therapy is associated with a significant decrease in the risk of vascular complications. Thus, LDL-C is the main therapeutic target in patients with dyslipidemia. Unfortunately, current LDL-C control figures are still very low, partly due to insufficient intensification of lipid-lowering therapy, but also due to the need for new tools to achieve these goals. This paper reviews the different lipid-lowering treatment options, including the latest available therapies, and provides a practical approach to achieving LDL-C control goals in patients with hypercholesterolemia, as well as in different patient subgroups.
Collapse
Affiliation(s)
- S Cinza-Sanjurjo
- Centro de Salud Milladoiro, Área Sanitaria de Santiago de Compostela, Santiago de Compostela, A Coruña, España; Instituto de Investigación de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, España; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, España; Grupo de Trabajo de Hipertensión Arterial y Enfermedad Cardiovascular de SEMERGEN; Agencia de Investigación de SEMERGEN; Junta directiva de SEMERGEN.
| | - V Pallarés-Carratalá
- Grupo de Trabajo de Hipertensión Arterial y Enfermedad Cardiovascular de SEMERGEN; Junta directiva de SEMERGEN; Departamento de Medicina, Universitat Jaume I, Castellón, España
| | - A Díaz Rodríguez
- Centro de Salud Bembibre, Bembibre, León, España; Grupo de Trabajo de Lípidos de SEMERGEN
| | - D Fierro-González
- Grupo de Trabajo de Lípidos de SEMERGEN; Centro de Salud de Armunia, León, España
| | - M Turégano-Yedro
- Grupo de Trabajo de Hipertensión Arterial y Enfermedad Cardiovascular de SEMERGEN; Centro de Salud Casar de Cáceres, Casar de Cáceres, Cáceres, España
| | - J Polo-García
- Grupo de Trabajo de Hipertensión Arterial y Enfermedad Cardiovascular de SEMERGEN; Junta directiva de SEMERGEN; Centro de Salud Casar de Cáceres, Casar de Cáceres, Cáceres, España
| |
Collapse
|
40
|
Balmforth C, McDermott M, Khaing P, Dweck MR, Newby DE. Identification and management of non-obstructive high-risk coronary artery plaque. Heart 2025:heartjnl-2024-324858. [PMID: 40122588 DOI: 10.1136/heartjnl-2024-324858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Despite advances in its understanding and treatment, coronary heart disease remains the leading cause of death worldwide. Acute coronary syndromes most commonly result as a consequence of the rupture of non-flow-limiting, high-risk coronary artery plaques. Advances in multimodality imaging have allowed the detailed assessment of the high-risk plaque including the assessment of plaque burden, high-risk plaque features, plaque activity and thrombosis. This in turn may help identify those patients at greatest risk, and thus benefit from intensification of pharmacotherapies or from local preventative strategies including coronary revascularisation. However, the optimal management of high-risk plaques and the prevention of consequent adverse coronary events have yet to be established. Further development and observational studies are required to determine how best to apply high-risk plaque metrics into clinical practice. Nevertheless, a new era of precision medicine is upon us, with advanced plaque imaging allowing enhanced risk stratification and targeting of the growing armamentarium of atherosclerotic therapies to the highest risk patients.
Collapse
Affiliation(s)
- Craig Balmforth
- The University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| | - Michael McDermott
- The University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| | - Phyo Khaing
- The University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| | - Marc Richard Dweck
- The University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| | - David E Newby
- The University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| |
Collapse
|
41
|
Mahmoud A, Abdelsayed K, Mohamed AA, Najah Q, Abdulkader A, Ali K, Tabassum S, Abouzid MR, Abdelazeem B, Mills JD. Safety and efficacy of antisense oligonucleotides on triglyceride, apolipoprotein C-III, and other lipid parameters levels in hypertriglyceridemia; a network meta-analysis of randomized controlled trials. Lipids Health Dis 2025; 24:109. [PMID: 40119340 PMCID: PMC11929262 DOI: 10.1186/s12944-024-02389-2] [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: 08/26/2024] [Accepted: 11/27/2024] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Hypertriglyceridemia is an independent risk factor for cardiovascular diseases. In previous trials, apolipoprotein C-III (APOC3) inhibition through the antisense oligonucleotides volanesorsen, olezarsen, and plozasiran reduced triglyceride levels. However, the three medications' safety and efficacy have yet to be compared. METHODS A network meta-analysis was performed to compare multiple doses of the three medications to each other through the placebo. Randomized controlled trials (RCTs) were retrieved by searching PubMed, EMBASE, Web of Science, SCOPUS, and Cochrane until November 22nd, 2024. The mean difference (MD) and 95% confidence interval (CI) were used for continuous outcomes. The risk ratio (RR) and 95% CI were used for dichotomous outcomes. RESULTS Ten RCTs with a total of 1,129 patients were included. volanesorsen 300 mg once weekly showed the most significant percent reduction in triglyceride levels (MD = -91.0%, 95% CI: (-109.2%; -72.8%); P < 0.01). Only plozasiran once monthly, regardless of the dose, showed a non-significant percent reduction in triglycerides. This finding should be taken cautiously as the data were derived from a phase 1 trial with a small sample size. All the regimens significantly reduced APOC3 levels compared to placebo, with plozasiran 100 mg monthly and volanesorsen 300 mg once weekly showing the most significant reduction (MD range: -92.8% to -88.5%; P < 0.01). None of the treatments showed a statistically significant difference in overall adverse events rate compared to the placebo. CONCLUSION APOC3 antisense oligonucleotide inhibitors effectively reduced triglyceride and APOC3 levels in hypertriglyceridemia with an acceptable safety profile. However, the results should be interpreted cautiously due to the small sample size. Further research is needed to confirm the beneficial effects of APOC3 inhibitors and show strong evidence of the impact of each regimen.
Collapse
Affiliation(s)
| | - Kerollos Abdelsayed
- Clinical Research Department, Aswan Heart Centre, Magdi Yacoub Foundation, Aswan, Egypt.
| | - Ahmed Almahdy Mohamed
- Department of Medicine, Division of Cardiology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Qasi Najah
- Faculty of Medicine, Elmergib University, Alkhums, Libya
| | - Anas Abdulkader
- College of Medicine, AlMaarefa University, Riyadh, KSA, Saudi Arabia
| | - Karim Ali
- Department of Internal Medicine, Hennepin County Medical Center, Minneapolis, MN, USA
| | - Shehroze Tabassum
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Mohamed Riad Abouzid
- Department of Internal Medicine, Baptist Hospitals of Southeast Texas, Beaumont, TX, USA
| | - Basel Abdelazeem
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| | - James D Mills
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
42
|
Odeberg J, Halling A, Ringborn M, Freitag M, Persson ML, Vaara I, Råstam L, Odeberg H, Lindblad U. Markers of inflammation predicts long-term mortality in patients with acute coronary syndrome - a cohort study. BMC Cardiovasc Disord 2025; 25:190. [PMID: 40089663 PMCID: PMC11909928 DOI: 10.1186/s12872-025-04608-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/25/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Chronic low-grade inflammation is a well-known risk factor for coronary heart disease (CHD) and future cardiovascular events. Anti-inflammatory therapy can reduce the risk of ischemic cardiovascular disease (CVD) events following myocardial infarction (MI). However, it remains unknown to what extent inflammation at the time of an acute event predicts long-term outcomes. We explored whether routine blood measurements of inflammatory markers during an acute coronary syndrome (ACS) are predictive of long-term mortality. METHODS In a cohort of 5292 consecutive patients admitted to a coronary intensive care unit with suspected ACS over a four-year period in the Carlscrona Heart Attack Prognosis Study (CHAPS), 908 patients aged 30-74 years (644 men, 264 women) were diagnosed with MI (527) or unstable angina (UA) (381). A 10-year follow-up study was conducted using Swedish national registries, with total mortality and cardiac mortality as primary outcomes. RESULTS Long-term total and cardiac mortality were significantly associated with higher leukocyte counts (e.g., neutrophils, monocytes, p ≤ 0.001), higher levels of inflammatory biomarkers (e.g., C-reactive protein, Serum Amyloid A, fibrinogen, p ≤ 0.001), and elevated neutrophil-lymphocyte ratio (NLR) (p < 0.001) and monocyte-lymphocyte ratio (MLR) (p = 0.002), all measured at ACS admission. These associations were independent of ACS diagnosis. CONCLUSION Our results suggest that level of inflammation at ACS presentation-beyond its established role as a major CHD risk factor-also predicts long-term mortality following ACS. Notably, inflammation at the time of the event was a stronger predictor of long-term mortality than the acute event outcome itself. However, limitations include the observational study design, moderate sample size, and absence of modern high-sensitivity cardiac biomarkers and contemporary ACS management strategies in this cohort. The results should therefore be interpreted in the context of historical clinical practice. While our model-wise complete-case approach ensured consistency, missing data remains a potential source of bias. Future studies in larger, more contemporary cohorts are needed to validate these findings and refine risk stratification strategies.
Collapse
Affiliation(s)
- Jacob Odeberg
- Department of Protein Science, Science for Life Laboratory Stockholm, CBH, KTH Royal Institute of Technology, Stockholm, 100 44, Sweden.
- Department of Clinical Medicine, Faculty of Health Science, Arctic University of Tromsö (UiT), Tromsö, N 9037, Norway.
- Division of Internal Medicine, University Hospital North Norway (UNN), Tromsö, Norway.
- Department of Hematology, Coagulation Unit, Karolinska University Hospital, Stockholm, Sweden.
- Department of Medicine, Solna, Karolinska Institute, Stockholm, SE, 171 77, Sweden.
| | - Anders Halling
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Michael Ringborn
- Thoracic Center, Blekinge County Hospital Karlskrona, Karlskrona, Sweden
| | - Michael Freitag
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Marie Louise Persson
- Department of Laboratory Medicine, Blekinge County Hospital Karlskrona, Karlskrona, Sweden
| | - Ivar Vaara
- Department of Laboratory Medicine, Blekinge County Hospital Karlskrona, Karlskrona, Sweden
| | - Lennart Råstam
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Håkan Odeberg
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Ulf Lindblad
- School of Public Health and Community Medicine/Primary Health Care, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
43
|
Miao M, Liu X, Zhang H, Dai H. Immuno-inflammatory mechanisms in cardio-oncology: new hopes for immunotargeted therapies. Front Oncol 2025; 15:1516977. [PMID: 40182041 PMCID: PMC11966441 DOI: 10.3389/fonc.2025.1516977] [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: 10/25/2024] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
Cardio-oncology is an emerging interdisciplinary field concerned with cancer treatment-related cardiovascular toxicities (CTR-CVT) and concomitant cardiovascular diseases (CVD) in patients with cancer. Inflammation and immune system dysregulation are common features of tumors and cardiovascular disease (CVD). In addition to the mutual exacerbating effect through inflammation, tumor treatments, including immunotherapy, chemotherapy, radiation therapy, and targeted therapy, may induce immune inflammatory reactions leading to cardiovascular damage. Cancer immunotherapy is currently a new method of cancer treatment. Immunotherapeutic agents, such as immune checkpoint inhibitors (ICIs), chimeric antigen receptor T cell immunotherapy (CAR-T), mRNA vaccines, etc., can induce anti-tumor effects by enhancing the host immune response to eliminate tumor cells. They have achieved remarkable therapeutic efficacy in clinical settings but lead to many immune-related adverse events (irAEs), especially CTR-CVT. Establishing specific evaluation, diagnostic, and monitoring criteria (e.g., inflammatory biomarkers) for both immunotherapy and anti-inflammatory therapy-related cardiovascular toxicity is vital to guide clinical practice. This article explores the role of immune response and inflammation in tumor cardiology, unravels the underlying mechanisms, and provides improved methods for monitoring and treating in CTR-CVT in the field of cardio-oncology.
Collapse
Affiliation(s)
- Meiqi Miao
- Department of Cardiology, Kunshan Hospital of Chinese Medicine, Kunshan, China
| | - Xinxin Liu
- Postdoctoral Mobile Station, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- The Innovation Base, Mudanjiang Collaborative Innovation Center for the Development and Application of Northern Medicinal Resources, Mudanjiang, China
| | - Han Zhang
- Department of Cardiology, Yan’an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hailong Dai
- Department of Cardiology, Yan’an Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
44
|
Sgarra L, Desantis V, Matteucci A, Caccavo VP, Troisi F, Di Monaco A, Mangini F, Katsouras G, Guaricci AI, Dadamo ML, Fortunato F, Nacci C, Potenza MA, Montagnani M, Grimaldi M. Non-Anticoagulation Strategies Aimed at Primary Stroke Prevention in Nascent Atrial Fibrillation. Biomedicines 2025; 13:660. [PMID: 40149635 PMCID: PMC11939867 DOI: 10.3390/biomedicines13030660] [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: 01/16/2025] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
At its earliest appearance, atrial fibrillation (AF) is often unnoticed, asymptomatic, and/or merely device-detected. Widespread use of heart-rate monitoring technologies has facilitated such "nascent atrial fibrillation (nAF)" recognition. Consequently, clinicians face a growing number of patients affected by new-onset AF in the absence of a definite indication for anticoagulation due to several counterarguments: (1) a CHA2DS2-VA score ≤ 1 in otherwise apparently healthy subjects; (2) an uncertain embolic/hemorrhagic benefit/risk ratio with anticoagulation; (3) EKG demonstration and confirmation of AF; and (4) existence of a pathogenic mechanism other than atrial hypercoagulability. In this frustrating limitation of pharmacological options, cardiologists may miss a complete comprehension of drugs with proven anti-ictal potential, whose administration may serve both as a bridge strategy toward future anticoagulation and as a consolidative strategy paralleling anticoagulation. This review aims to summarize and elucidate such therapeutic strategies and their preventative mechanisms.
Collapse
Affiliation(s)
- Luca Sgarra
- Cardiology Unit, Medicine Department, General Hospital “F. Miulli” Acquaviva delle Fonti, 70021 Bari, Italy
| | - Vanessa Desantis
- Pharmacology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, 70124 Bari, Italy (M.M.)
| | - Andrea Matteucci
- Clinical and Rehabilitation Cardiology Unit, Emergency Department, San Filippo Neri Hospital, ASL Rome 1, 00135 Rome, Italy
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Vincenzo Paolo Caccavo
- Cardiology Unit, Medicine Department, General Hospital “F. Miulli” Acquaviva delle Fonti, 70021 Bari, Italy
| | - Federica Troisi
- Cardiology Unit, Medicine Department, General Hospital “F. Miulli” Acquaviva delle Fonti, 70021 Bari, Italy
| | - Antonio Di Monaco
- Cardiology Unit, Medicine Department, General Hospital “F. Miulli” Acquaviva delle Fonti, 70021 Bari, Italy
| | - Francesco Mangini
- Cardiology Unit, Medicine Department, General Hospital “F. Miulli” Acquaviva delle Fonti, 70021 Bari, Italy
| | - Grigorios Katsouras
- Cardiology Unit, Medicine Department, General Hospital “F. Miulli” Acquaviva delle Fonti, 70021 Bari, Italy
| | - Andrea Igoren Guaricci
- Cardiology Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Michele Luca Dadamo
- Cardiology Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Fabrizio Fortunato
- Department of Cardiology, Azienda Ospedaliera Universitaria Policlinico Paolo Giaccone, 90127 Palermo, Italy
| | - Carmela Nacci
- Pharmacology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, 70124 Bari, Italy (M.M.)
| | - Maria Assunta Potenza
- Pharmacology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, 70124 Bari, Italy (M.M.)
| | - Monica Montagnani
- Pharmacology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, 70124 Bari, Italy (M.M.)
| | - Massimo Grimaldi
- Cardiology Unit, Medicine Department, General Hospital “F. Miulli” Acquaviva delle Fonti, 70021 Bari, Italy
| |
Collapse
|
45
|
Diau JL, Lange RA. Coronary Inflammation and Cardiovascular Events in Patients Without Obstructive Coronary Artery Disease. Curr Cardiol Rep 2025; 27:68. [PMID: 40053166 PMCID: PMC11889004 DOI: 10.1007/s11886-025-02221-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 03/10/2025]
Abstract
PURPOSE OF REVIEW This review evaluates the role of vascular inflammation in patients who develop myocardial infarction with non-obstructive coronary arteries (MINOCA). It also introduces pericoronary adipose tissue (PCAT) and epicardial adipose tissue (EAT) as possible biomarkers for risk prediction in patients with non-obstructive coronary artery disease (CAD). RECENT FINDINGS PCAT and EAT contribute to the development and progression of coronary artery inflammation and plaque vulnerability. Coronary computed tomography angiography (CCTA) can detect localized areas of inflammation through changes in the attenuation values of PCAT and EAT. Attenuation values can be further integrated with traditional risk factors using artificial intelligence to generate risk scores that significantly enhance prognostic accuracy in patients with and without obstructive coronary artery disease. Assessing PCAT and EAT inflammation via CCTA and AI-driven risk algorithms enable precise risk prediction of MINOCA and major adverse coronary events (MACE) in patients with non-obstructive CAD.
Collapse
Affiliation(s)
- Jia Ling Diau
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| | - Richard A Lange
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA.
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA.
- TTUHSC-El Paso, 130 Rick Francis St. MC 11001, El Paso, TX, 79905, USA.
| |
Collapse
|
46
|
Yazdani A, Hoshi R, Ammar M, Li C, Cummings RD, Akmačić-Trbojević I, Cindrić A, Šimunić-Briški N, Gudelj I, Glynn R, Ridker P, Chasman DI, Lauc G, Demler O, Mora S. Statin effects on immunoglobulin-G glycomic architecture and the link to cardiovascular disease. RESEARCH SQUARE 2025:rs.3.rs-6112380. [PMID: 40092445 PMCID: PMC11908345 DOI: 10.21203/rs.3.rs-6112380/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Background Immunoglobulin G (IgG) plays a critical role in immune defense yet our understanding of its role in cardiovascular disease (CVD) is evolving. Observational studies have correlated statin use with changes in IgG N-glycan structures. However, statin effects on IgG N-glycan changes have not been tested in randomized controlled trials, and their direct association with CVD remains unclear. Methods IgG N-glycans were measured at baseline and after one year of randomized high-intensity statin interventions in 2 sub-studies of randomized trials: JUPITER (Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin; NCT00239681; primary prevention; discovery, n = 239 participants); and TNT (Treating to New Targets; NCT00327691; secondary prevention; validation, n = 711). Using linear regression adjusted for baseline levels of IgG N-glycans and clinical risk factors (e.g., age, sex) as well as the occurrence of CVD during the year of follow-up, we investigated the one-year randomized effects of high-intensity rosuvastatin v. placebo on IgG N-glycans in JUPITER. Significant statin-IgG N-glycan associations were then validated in TNT with one-year randomized effects of high- v. low-intensity atorvastatin intervention. We examined the architecture of IgG N-glycan connectivity at baseline using a data-driven Bayesian network and compared it with the architecture after one year of randomized statin intervention. We then investigated whether the changes in IgG N-glycans triggered by statins were associated with incident CVD events. Results We identified 5 IgG N-glycans (corresponding to core fucosylated, monosialylated, and disialylated IgG N-glycans) in JUPITER whose levels decreased significantly with statin versus placebo (false discovery rate < 0.05), with an approximate 11.3-25.9% reduction in the individual IgG N-glycan levels. Four out of the five IgG N-glycans altered by statin were validated in TNT. Furthermore, monosialylation and core fucosylation (glycan peaks, GP 16 and 18) were inversely associated with CVD in JUPITER (OR = 0.87 and 0.73 per standard deviation increase, 95% CI: (0.57, 0.98) and (0.55, 0.96) respectively), and validated in TNT. Despite the effect of statin therapy on certain IgG N-glycans, the overall architecture of the IgG N-glycan network remained unchanged after one year of statin intervention. Conclusion High-intensity statin interventions decreased several specific IgG N-glycan levels without changing the overall architecture of IgG N-glycan connectivity. Two IgG N-glycans that were decreased by statins were inversely associated with CVD outcomes, suggesting that statins have effects on monosialylated and core fucosylated IgG N-glycans, which may affect their cardioprotective properties. These findings highlight a potential immunomodulatory role of statins through IgG N-glycan alterations that should be further investigated in relation to CVD.
Collapse
|
47
|
Nomura SO, Bhatia HS, Garg PK, Karger AB, Guan W, Cao J, Shapiro MD, Tsai MY. Lipoprotein(a), high-sensitivity c-reactive protein, homocysteine and cardiovascular disease in the Multi-Ethnic Study of Atherosclerosis. Am J Prev Cardiol 2025; 21:100903. [PMID: 39802678 PMCID: PMC11722194 DOI: 10.1016/j.ajpc.2024.100903] [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: 08/14/2024] [Revised: 11/26/2024] [Accepted: 12/01/2024] [Indexed: 01/16/2025] Open
Abstract
Background and aims Elevated lipoprotein(a) [Lp(a)], high-sensitivity C-Reactive Protein (hs-CRP), and total homocysteine (tHcy) are associated with atherosclerotic cardiovascular disease (ASCVD) risk. This study investigated the individual and joint associations of Lp(a), hs-CRP and tHcy with coronary heart disease (CHD) and stroke. Methods This study was conducted in the Multi-Ethnic Study of Atherosclerosis (MESA) cohort (2000-2017) (CHD analytic N = 6,676; stroke analytic N = 6,674 men and women). Associations between Lp(a) (<50 vs. ≥50 mg/dL), hs-CRP (<2 vs. ≥2 mg/L) and tHcy (<12 vs. ≥12 µmol/L) and CHD and stroke incidence were evaluated individually and jointly using Cox proportional hazards regression. Results Individually, elevated tHcy was associated with CHD and stroke incidence, Lp(a) with CHD only and hs-CRP with stroke only. In combined analyses, CHD risk was higher when multiple biomarkers were elevated [hs-CRP+Lp(a), hazard ratio (HR)=1.39, 95 % confidence interval (CI): 1.06, 1.82; hs-CRP+ tHcy, HR = 1.34, 95 % CI: 1.02, 1.75; Lp(a)+ tHcy HR = 1.58, 95 % CI: 1.08, 2.30; hs-CRP+Lp(a)+ tHcy HR = 2.02, 95 % CI: 1.26, 3.24]. Stroke risk was elevated when hs-CRP and either Lp(a) (HR = 1.51, 95 % CI: 1.02, 2.23) or tHcy (HR = 2.10, 95 % CI: 1.44, 3.06) was also high, when all three biomarkers were elevated (HR = 2.99, 95 % CI: 1.61, 5.58), or when hs-CRP and tHcy (HR = 1.79, 95 % CI: 1.16, 2.76) were both high. Conclusions Risk of ASCVD was highest with concomitant elevation of tHcy, hs-CRP and Lp(a). Inclusion of tHcy and consideration of biomarker combination rather than individual biomarker levels may help better identify individuals at greatest risk for ASCVD events.
Collapse
Affiliation(s)
- Sarah O. Nomura
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN 55455, USA
| | - Harpreet S. Bhatia
- Division of Cardiovascular Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Parveen K. Garg
- Division of Cardiology, University of Southern California Keck School of Medicine, 1975 Zonal Ave., Los Angeles, CA 90033, USA
| | - Amy B. Karger
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN 55455, USA
| | - Weihua Guan
- School of Public Health Biostatistics Division, University of Minnesota, 420 Delaware St SE, MN, 55455, USA
| | - Jing Cao
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Michael D. Shapiro
- Center for the Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Wake Forest University School of Medicine, 475 Vine Street, Winston-Salem, North Carolina 27101, USA
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN 55455, USA
| |
Collapse
|
48
|
Hu Y, Cui X, Lu M, Guan X, Li Y, Zhang L, Lin L, Zhang Z, Zhang M, Hao J, Wang X, Huan J, Li Y, Li C. Body Fat Distribution and Ectopic Fat Accumulation as Mediator of Diabetogenic Action of Lipid-Modifying Drugs: A Mediation Mendelian Randomization Study. Mayo Clin Proc 2025; 100:424-439. [PMID: 39918451 DOI: 10.1016/j.mayocp.2024.10.018] [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: 04/24/2024] [Revised: 10/06/2024] [Accepted: 10/25/2024] [Indexed: 05/08/2025]
Abstract
OBJECTIVE To investigate the causal relationship between various lipid-modifying drugs and new-onset diabetes, as well as the mediators contributing to this relationship. METHODS Mediation Mendelian randomization was performed to investigate the causal effect of lipid-modifying drug targets on type 2 diabetes (T2D) outcomes and the proportion of this association that is mediated through ectopic fat accumulation traits. Specific sets of variants in or near genes that encode 11 lipid-modifying drug targets (LDLR, HMGCR, NPC1L1, PCSK9, APOB, ABCG5/ABCG8, LPL, PPARA, ANGPTL3, APOC3, and CETP; for expansion of gene symbols, use search tool at www.genenames.org) were extracted. Random effects inverse variance weighted were performed to evaluate the causal effects among outcomes. Mediation analyses were performed to identify the mediators of the association between lipid-modifying drugs and T2D. The study was conducted from November 10, 2023, to April 2, 2024 RESULTS: The genetic mimicry of HMGCR and APOB inhibition was associated with an increased T2D risk, whereas the genetic mimicry of LPL enhancement was linked to a lower T2D risk. Gluteofemoral adipose tissue volume was a mediator for explaining 9.52% (P=.002), 16.90% (P=.03), and 10.50% (P=.003) of the total effect of HMGCR, APOB, and LPL on T2D susceptibility, respectively. Liver fat was a mediator for explaining 21.12% (P=.005), 12.28% (P=.03), and 9.84% (P=.005) of the total effect of HMGCR, APOB, and LPL on T2D susceptibility, respectively. CONCLUSION Our findings support the hypothesis that liver fat and gluteofemoral adipose tissue play a mediating role in the prodiabetic effects of HMGCR and APOB inhibition, as well as in the antidiabetic effects of LPL enhancement.
Collapse
Affiliation(s)
- Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinhai Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiuya Guan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuan Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Muxin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jiaqi Hao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaojie Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Jiaming Huan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yunlun Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China; Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
49
|
Laufs U, Lincoff AM, Nicholls SJ, Li N, Bloedon L, Sasiela WJ, Powell HA, Herout PM, Thompson PD, Nissen SE. Characteristics and outcomes of patients with and without statin-associated muscle symptoms treated with bempedoic acid in the CLEAR Outcomes trial. J Clin Lipidol 2025; 19:337-347. [PMID: 39939212 DOI: 10.1016/j.jacl.2024.12.014] [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: 11/05/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 02/14/2025]
Abstract
BACKGROUND Cholesterol Lowering via bEmpedoic Acid Regimen (CLEAR) outcomes, a randomized, double-blind, placebo-controlled cardiovascular outcomes trial, and the largest prospective database of patients with statin intolerance (SI), demonstrated that bempedoic acid reduces low-density lipoprotein cholesterol and cardiovascular risk in patients at high cardiovascular risk. OBJECTIVE Assess baseline differences in SI symptoms and whether these influenced the clinical course during CLEAR outcomes. METHODS Symptoms and impact of SI on daily living were recorded prior to randomization. This posthoc analysis grouped patients as reporting statin-associated muscle symptoms only (SAMS), nonmuscle adverse effects only (nonSAMS), or BOTH. RESULTS Of the 13,970 patients at baseline, 49% reported SAMS, 18% nonSAMS, and 33% BOTH. Moderate/severe impact on daily living was recorded for 62% SAMS, 48% nonSAMS, and 69% BOTH. Baseline lipid modifying treatment (LMT) was used in 43% SAMS, 36% nonSAMS, and 42% BOTH. Drop-in use of moderate/high-intensity statin at any time during the study was higher in the placebo group in all SI groups and higher in SAMS and BOTH vs nonSAMS, but was not generally maintained at study end. SAMS and BOTH groups had more muscle symptoms and higher rates of treatment discontinuation vs. nonSAMS but there was no difference between treatments. CONCLUSION Patients who reported SAMS, regardless of randomization to bempedoic acid or placebo, had higher rates of discontinuation, higher rates of skeletal muscle symptoms, and a greater percentage of patients to attempt statin rechallenge. These findings indicate patients with history of SAMS may have background factors impacting their tolerance to LMT and may require more focused clinical management. CLINICALTRIALS GOV IDENTIFIER NCT02993406.
Collapse
Affiliation(s)
- Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany (Dr Laufs).
| | - A Michael Lincoff
- Department of Cardiovascular Medicine and Cleveland Clinic Coordinating Center for Clinical Research (C5Research), Cleveland Clinic, Cleveland OH, USA (Drs Lincoff and Nissen)
| | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, Melbourne, Australia (Dr Nicholls)
| | - Na Li
- Esperion Therapeutics, Inc, Ann Arbor, MI, USA (Drs Li, Bloedon, Sasiela, Powell, and Herout)
| | - LeAnne Bloedon
- Esperion Therapeutics, Inc, Ann Arbor, MI, USA (Drs Li, Bloedon, Sasiela, Powell, and Herout)
| | - William J Sasiela
- Esperion Therapeutics, Inc, Ann Arbor, MI, USA (Drs Li, Bloedon, Sasiela, Powell, and Herout)
| | - Heather A Powell
- Esperion Therapeutics, Inc, Ann Arbor, MI, USA (Drs Li, Bloedon, Sasiela, Powell, and Herout)
| | - Peter M Herout
- Esperion Therapeutics, Inc, Ann Arbor, MI, USA (Drs Li, Bloedon, Sasiela, Powell, and Herout)
| | | | - Steven E Nissen
- Department of Cardiovascular Medicine and Cleveland Clinic Coordinating Center for Clinical Research (C5Research), Cleveland Clinic, Cleveland OH, USA (Drs Lincoff and Nissen)
| |
Collapse
|
50
|
Sou FM, Hsu CN, Chiu YC, Wu CK, Lu LS, Kuo CM, Chiu SM, Chuah SK, Yang YH, Liang CM. The association between trajectory of serum cholesterol, statin dosage, and the risk of recurrent biliary stone diseases. J Formos Med Assoc 2025; 124:246-252. [PMID: 38589275 DOI: 10.1016/j.jfma.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 02/13/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Statins may reduce the risk of recurrent gallstone disease by decreasing bile cholesterol saturation and pathogenicity. However, limited studies have investigated this issue. This study aimed to assess whether statin doses and serum cholesterol levels were associated with a decreased risk of recurrent biliary stone diseases after the first event index, with a follow-up time of 15 years. METHODS Based on the Chang Gung Research Database (CGRD) between January 1, 2001, and December 31, 2020, we enrolled 68,384 patients with the International Classification of Diseases, Ninth and Tenth Revision codes of choledocholithiasis. After exclusions, 32,696 patients were divided into non-statin (<28 cDDD, cumulative defined daily doses) (n = 27,929) and statin (≥28 cDDD) (n = 4767) user groups for analysis. Serum cholesterol trajectories were estimated using group-based trajectory modeling (n = 8410). RESULTS The statin users had higher Charlson Comorbidity Index (CCI) scores than the non-statin users. Time-dependent Cox regression analysis showed that statin use >365 cDDD was associated with a significantly lower risk of recurrent biliary stones (adjusted hazard ratio [aHR] = 0.28, 95% CI, 0.24-0.34; p < 00.0001), acute pancreatitis (aHR = 0.24, 95% CI, 0.17-0.32, p < 00.0001), and cholangitis (aHR = 0.28, 95% CI, 0.25-0.32, p < 00.0001). Cholecystectomy was also a protective factor for recurrent biliary stones (aHR = 0.41, 95% CI, 0.37-0.46; p < 00.0001). The higher trajectory serum cholesterol group (Group 3) had a lower risk trend for recurrent biliary stones (aHR = 0.79, p = 0.0700) and a lower risk of cholangitis (aHR = 0.79, p = 0.0071). CONCLUSION This study supports the potential benefits of statin use and the role of cholecystectomy in reducing the risk of recurrent biliary stone diseases.
Collapse
Affiliation(s)
- Fai-Meng Sou
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Chun Chiu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Cheng-Kun Wu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Lung-Sheng Lu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chung-Mou Kuo
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shao-Ming Chiu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Seng-Kee Chuah
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yao-Hsu Yang
- Department of Traditional Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan; Health Information and Epidemiology Laboratory of Chang Gung Memorial Hospital, Chiayi, Taiwan; School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Ming Liang
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|