1
|
Tang XL, Hooper AJ, Burnett JR. An evaluation of recaticimab for the treatment of hypercholesterolemia. Expert Opin Biol Ther 2025:1-6. [PMID: 40380894 DOI: 10.1080/14712598.2025.2508837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/13/2025] [Accepted: 05/16/2025] [Indexed: 05/19/2025]
Abstract
INTRODUCTION Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, by preventing the degradation of LDL receptors, either through interference in the binding of PCSK9 to LDL receptors or through silencing of PCSK9 at a molecular level, have revolutionized lipid-lowering treatment and offer the opportunity to further improve clinical outcomes for patients with hypercholesterolemia. AREAS COVERED We discuss the role of PCSK9 as a therapeutic target for hypercholesterolemia, describe the pharmacodynamics, pharmacokinetics, and metabolism of recaticimab, and report the recent clinical trials with this 'humanized' IgG1 monoclonal antibody (mAb) against PCSK9. EXPERT OPINION Recaticimab has a high affinity for PCSK9 that confers a prolonged duration of action. Recaticimab durably decreases LDL-cholesterol, non-HDL-cholesterol and apoB, but can also lower Lp(a). Recaticimab may offer advantages over current mAbs in clinical use in terms of its long half-life, dosing interval of up to 12 weeks, and potentially a lower cost; however, long-term concerns regarding immunogenicity remain. Longer-term studies in a variety of more diverse patient cohorts will be needed to further evaluate the efficacy, safety, and durability of recaticimab and to ascertain the optimal dosing schedule for cardiovascular outcome studies.
Collapse
Affiliation(s)
- Xuan L Tang
- Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth Hospital & Fiona Stanley Hospital Network, Perth, Western Australia
| | - Amanda J Hooper
- Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth Hospital & Fiona Stanley Hospital Network, Perth, Western Australia
- School of Medicine, University of Western Australia, Perth, Western Australia
| | - John R Burnett
- Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth Hospital & Fiona Stanley Hospital Network, Perth, Western Australia
- School of Medicine, University of Western Australia, Perth, Western Australia
| |
Collapse
|
2
|
Gong X, Gang Y, Lu Z, Cai Q, Xue M, Zhu B, Cheng X, Yu C, Lu J. Residual inflammatory risk is associated with leukoaraiosis in patients with ischemic stroke. J Stroke Cerebrovasc Dis 2025; 34:108261. [PMID: 39988002 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/25/2025] Open
Abstract
OBJECTIVE Emerging evidence has highlighted the clinical significance of residual inflammation risk (RIR) in cardiovascular and cerebrovascular diseases, with studies demonstrating its association with disease recurrence and poor prognosis. This study aimed to investigate the relationship between RIR and leukoaraiosis (LA) severity in patients with ischemic stroke. METHODS In this observational cohort study, we classified patients into four groups based on low-density lipoprotein cholesterol (LDL-C) and high-sensitivity C-reactive protein (hsCRP) levels: RIR (LDL-C < 2.6 mmol/L and hsCRP ≥ 2 mg/L), residual cholesterol risk (RCR) (LDL-C ≥ 2.6 mmol/L and hsCRP < 2 mg/L), both risk or residual cholesterol and inflammatory risk (RCIR) (LDL-C ≥ 2.6 mmol/L and hsCRP ≥ 2 mg/L) and neither risk (LDL-C < 2.6 mmol/L and hsCRP < 2 mg/L). LA presence and severity were assessed using magnetic resonance imaging (MRI) and graded according to the Fazekas scale. Ordinal logistic regression analyses were performed to evaluate the association between RIR and LA severity. RESULTS Among 643 enrolled patients, 413 (64.2 %) exhibited LA. The distribution of patients across RIR, RCR, RCIR, and neither risk groups was 28.9 %, 19.8 %, 20.4 %, and 30.9 %, respectively. Comparative analysis revealed that LA patients exhibited significantly higher age (P < 0.001), elevated BMI (P < 0.001), increased hypertension prevalence (P = 0.004), greater RIR proportion (P < 0.001), and higher smoking rates (P = 0.007) compared to non-LA counterparts. Ordinal logistic regression analysis demonstrated that RIR (OR 1.447, 95 % CI 1.044-1.851, P < 0.001) was independently associated with the severity of LA after adjusting for multiple confounding variables. Subgroup analysis stratified by BMI further revealed that RIR (OR 2.994, 95 % CI 2.259-3.730, P < 0.001) was significantly correlated with LA severity in patients with a BMI ≥ 25.0. CONCLUSIONS These findings suggest that RIR may serve as an independent risk factor for LA in patients with ischemic stroke, particularly among those with a BMI ≥25.0.
Collapse
Affiliation(s)
- Xiuqun Gong
- Department of Neurology, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan First People's Hospital, 203 Huaibin Road, Huainan, Anhui Province 232007, China.
| | - Yuwen Gang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan First People's Hospital, 203 Huaibin Road, Huainan, Anhui Province 232007, China; School of Medicine, Anhui University of Science and Technology, Huainan, Anhui Province, China.
| | - Zeyu Lu
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China.
| | - Qiankun Cai
- Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| | - Min Xue
- Department of Neurology, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan First People's Hospital, 203 Huaibin Road, Huainan, Anhui Province 232007, China.
| | - Beibei Zhu
- Department of Neurology, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan First People's Hospital, 203 Huaibin Road, Huainan, Anhui Province 232007, China.
| | - Xiaosi Cheng
- Department of Neurology, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan First People's Hospital, 203 Huaibin Road, Huainan, Anhui Province 232007, China.
| | - Chuanqing Yu
- Department of Neurology, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan First People's Hospital, 203 Huaibin Road, Huainan, Anhui Province 232007, China.
| | - Jun Lu
- School of Public Health, Anhui University of Science and Technology, 168 Taifeng Road, Huainan, Anhui Province 232001, China.
| |
Collapse
|
3
|
Xu H, Wang Z, Chen D, Zhang H, Ge J, Li J. PCSK-9 Inhibitors Can Significantly Improve the Coronary Slow Flow Caused by Elevated Lipoprotein (a) in ST-Elevation Myocardial Infarction Patients With Chronic Kidney Disease. Catheter Cardiovasc Interv 2025. [PMID: 40277036 DOI: 10.1002/ccd.31543] [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: 01/26/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Coronary slow flow and no reflow significantly predict poor prognosis in acute myocardial infarction (AMI) patients, especially those with chronic kidney disease (CKD). Early identification of factors contributing to these conditions can mitigate ischemic events and improve outcomes. AIMS This study aimed to investigate the association between elevated lipoprotein (a) [Lp(a)] levels and proprotein convertase subtilisin/kexin Type 9 (PCSK-9) inhibitor therapy with coronary slow flow or no reflow after percutaneous coronary intervention (PCI) in AMI patients with CKD. METHODS A total of 323 ST-elevation myocardial infarction (STEMI) patients who underwent PCI between October 2017 and June 2023 were included. Patients were divided into CKD (n = 132) and non-CKD (n = 191) groups. Lp(a) levels and the prevalence of coronary slow flow or no reflow after PCI were evaluated. STEMI patients with CKD were further categorized into elevated Lp(a) (n = 81) and normal Lp(a) (n = 51) subgroups. Logistic analysis identified risk factors for coronary slow flow/no reflow after PCI. The impact of PCSK-9 inhibitors on outcomes was also assessed in the elevated Lp(a) subgroup. RESULTS STEMI patients with CKD had significantly higher Lp(a) levels compared to those without CKD (median 36.75 vs. 15.90 mg/dL, p = 0.0001). CKD patients with elevated Lp(a) had a higher prevalence of coronary slow flow/no reflow after PCI than those with normal Lp(a) (38.3% vs. 13.7%, p = 0.002). Logistic regression analysis identified elevated Lp(a) as an independent risk factor for slow flow/no reflow after PCI in STEMI patients with CKD (OR = 2.985, p = 0.027). In CKD patients with elevated Lp(a), PCSK-9 inhibitors significantly improved post-PCI coronary flow and reduced composite cardiovascular events during 1-year follow-up (22.2% vs. 51.1%, p = 0.008). CONCLUSIONS Elevated Lp(a) is an independent risk factor for coronary slow flow or no reflow after PCI in STEMI patients with CKD. PCSK-9 inhibitors improve coronary blood flow and reduce cardiovascular events in these patients.
Collapse
Affiliation(s)
- Hao Xu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| | - Ziqing Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| | - Dan Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| | - Haojie Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| | - Junhua Ge
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| | - Jian Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), Qingdao, Shandong Province, China
| |
Collapse
|
4
|
Kamanu C, Karalis DG. The Role of Non-Statin Lipid Lowering Therapies to Reduce ASCVD Events in Primary Prevention. Curr Atheroscler Rep 2025; 27:46. [PMID: 40172616 PMCID: PMC11965143 DOI: 10.1007/s11883-025-01283-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/04/2025]
Abstract
PURPOSE OF REVIEW Atherosclerotic cardiovascular disease (ASCVD) remains a leading global health challenge, with low-density lipoprotein (LDL) cholesterol a pivotal risk factor. While statins are cornerstone therapy for lowering LDL cholesterol, many high-risk primary prevention patients are unable to tolerate statin therapy and do not achieve their guideline directed LDL cholesterol goal. For these patients, non-statin therapies offer complementary and alternative approaches to LDL cholesterol reduction. RECENT FINDINGS Recent advancements in non-statin therapies have expanded the options available to clinicians to lower LDL cholesterol in high-risk primary prevention patients. Yet these medications are often under-utilized in clinical practice. Observational studies, Mendelian randomization studies, and randomized clinical trials support the role of non-statin LDL cholesterol lowering therapies in the primary prevention of ASCVD. This review summarizes the evidence supporting their use for the primary prevention of ASCVD and offers practical suggestions as to how clinicians can integrate these medications into their clinical practice.
Collapse
Affiliation(s)
- Chukwuemezie Kamanu
- Department of Cardiology, Jefferson University Hospital, Sidney Kimmel Medical College, 227 North Broad Street, Suite 200, Philadelphia, PA, 19107, USA
| | - Dean G Karalis
- Department of Cardiology, Jefferson University Hospital, Sidney Kimmel Medical College, 227 North Broad Street, Suite 200, Philadelphia, PA, 19107, USA.
| |
Collapse
|
5
|
Carnero LAR, Bedinger D, Cocklin S, Li J, Erasmus MF, D'Angelo S, Leal-Lopes C, Teixeira AAR, Ferrara F, Bradbury ARM. Identification of polyreactive antibodies by high throughput enzyme-linked immunosorbent assay and surface Plasmon resonance. J Immunol Methods 2025; 539:113855. [PMID: 40157637 DOI: 10.1016/j.jim.2025.113855] [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: 11/21/2024] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
The assessment of polyreactivity is usually carried out by enzyme linked immunosorbent assay (ELISA) using biochemically diverse target antigens with different biochemical properties, including charge and hydrophobicity, and comprising proteins, carbohydrates, nucleic acids and lipids, some of which are heterogenous in nature. Here we explored polyreactivity ELISAs based on probes of defined molecular weight, which we were also able to directly transition to a polyreactivity assay using surface plasmon resonance (SPR). Using a panel of previously characterized clinical antibodies we obtain results compatible with previous polyreactivity studies, but with potential for high throughput analysis following kinetic measurements in the early discovery process. We find ELISA is more sensitive for the detection of polyreactivity in antibodies, and with potential lower throughput, compared to SPR, but may lack the linear sensitivity of SPR.
Collapse
Affiliation(s)
| | | | - Simon Cocklin
- Specifica Inc, an IQVIA Laboratories Company, Santa Fe 87501, USA
| | - Jianquan Li
- Specifica Inc, an IQVIA Laboratories Company, Santa Fe 87501, USA
| | - M Frank Erasmus
- Specifica Inc, an IQVIA Laboratories Company, Santa Fe 87501, USA
| | - Sara D'Angelo
- Specifica Inc, an IQVIA Laboratories Company, Santa Fe 87501, USA
| | - Camila Leal-Lopes
- New Mexico Consortium, Los Alamos 87504, USA; Sanofi, Large Molecule Research, 02141 Cambridge, USA
| | | | | | | |
Collapse
|
6
|
Zhang Y, Li K, Bo X, Zhang Y, Xiao T, Liu H, Villamil OIRC, Chen K, Ding J. Effects of residual inflammatory and cholesterol risks on cardiovascular events with evolocumab in patients with acute coronary syndrome undergoing percutaneous coronary intervention. Lipids Health Dis 2025; 24:123. [PMID: 40165297 PMCID: PMC11956451 DOI: 10.1186/s12944-025-02537-2] [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: 12/26/2024] [Accepted: 03/17/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Evolocumab has shown significant reductions in low-density lipoprotein cholesterol (LDL-C) levels and incident cardiovascular events among acute coronary syndrome (ACS) patients undergoing percutaneous coronary intervention (PCI). Nonetheless, the potential modification of evolocumab's effectiveness by baseline inflammatory risk remains unclear. We aimed to assess evolocumab's effectiveness based on baseline neutrophil-to-lymphocyte ratio (NLR) and evaluate residual inflammatory and cholesterol-related risks across varying on-treatment NLR and LDL-C levels. METHODS This multicentric, retrospective analysis enrolled consecutive patients with ACS undergoing PCI and exhibiting elevated LDL-C at the First Affiliated Hospital of Zhengzhou University and Zhongda Hospital Southeast University between March 2019 and August 2021. Patients were categorized into evolocumab and standard-of-care treatment groups based on evolocumab administration. Hazard ratios for the primary composite outcome-including myocardial infarction, ischemic stroke, cardiac death, unplanned coronary revascularization, and hospitalization due to unstable angina-comparing baseline NLR quartiles were computed using multivariable Cox regression. We assessed evolocumab's impact on the primary outcome across median-based NLR dichotomization and evaluated the outcome across 1-month NLR and LDL-C levels. RESULTS The median baseline NLR was 2.99 (IQR: 2.14-4.69), remaining stable following evolocumab therapy. Each NLR quartile increase heightened the risk of primary outcome by 29% (95% CI, 17-42%; P < 0.01). The relative risk reductions with evolocumab were consistent across NLR categories (P-interaction > 0.05), but absolute risk reductions were higher in high-NLR patients (2.9% vs. 6.2%). Residual inflammatory and cholesterol risks, indicated by on-treatment NLR and LDL-C, independently correlated with the primary outcome (P < 0.001). CONCLUSIONS Higher baseline NLR is associated with increased cardiovascular risk in ACS/PCI patients. Relative risk reductions with evolocumab were consistent across NLR categories, while absolute risk reductions were more significant in high-NLR patients. Minimized risk is observed in patients with the lowest on-treatment NLR and LDL-C levels.
Collapse
Affiliation(s)
- Yahao Zhang
- Department of Cardiology, National Key Clinical Specialty, Zhongda Hospital, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Kairu Li
- Department of Cardiology, Tinghu People's Hospital of Yancheng City, Yancheng, 224000, China
| | - Xiangwei Bo
- Department of Cardiology, National Key Clinical Specialty, Zhongda Hospital, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Yanghui Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Tingting Xiao
- Department of Cardiology, National Key Clinical Specialty, Zhongda Hospital, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Huan Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Orion I R Chiara Villamil
- Department of Cardiology, National Key Clinical Specialty, Zhongda Hospital, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Kui Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jiandong Ding
- Department of Cardiology, National Key Clinical Specialty, Zhongda Hospital, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China.
| |
Collapse
|
7
|
Huang JXF, Yousaf A, Moon J, Ahmed R, Uppal K, Pemminati S. Recent Advances in the Management of Dyslipidemia: A Systematic Review. Cureus 2025; 17:e81034. [PMID: 40264627 PMCID: PMC12013775 DOI: 10.7759/cureus.81034] [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: 03/22/2025] [Indexed: 04/24/2025] Open
Abstract
Dyslipidemia refers to abnormal levels of lipids in the bloodstream, typically exhibiting an increased pattern. Total cholesterol, high-density lipoprotein-cholesterol (HDL-C), low-density lipoprotein-cholesterol (LDL-C), and triglycerides (TGs) are all contributing factors to this disorder. This leads to an increased risk of atherosclerosis and cardiovascular diseases, such as coronary artery disease, which elevates the likelihood of morbidity. Dyslipidemia can be managed via the use of numerous classes of drugs and treatments. The conventional pharmacological agents comprising 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, selective cholesterol absorption inhibitors, proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), bile acid sequestrants, monoclonal antibodies, and nutritional supplementation, such as inhibitors of cholesterol synthesis and absorption, and promoters of LDL-C excretion, are also discussed. Furthermore, conventional pharmacological treatment of dyslipidemia may elicit a variety of adverse side effects that are detrimental to the quality of life of the user. These side effects include muscle pain, weakness, liver enzyme elevations, and hyperglycemia. This systematic review further analyzes the pharmacological actions of novel lipid-lowering agents such as adenosine triphosphate-citrate lyase inhibitors (ACLi), selective peroxisome proliferator-activated receptor alpha (PPARα) modulators, cholesteryl ester transfer protein inhibitors (CETPi), antisense oligonucleotides (ASO), and angiopoietin-like protein 3 inhibitors (ANGPTL3i) as well as their efficacy in treating dyslipidemia while sparing the user of potentially severe side effects. Compared to existing treatments, novel therapies have shown significantly greater effectiveness in managing dyslipidemia-related lipid profiles and exhibit fewer systemic adverse effects. Some of the recent therapies discussed are alternative treatments that offer patients promising efficacy and improved tolerability. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed to ensure a robust and transparent search process, aiming to minimize bias and maximize the retrieval of pertinent studies for review. Thus, this systematic review provides an overview of current and novel treatments for dyslipidemia, describing their efficacy, mechanism of action, safety, and side effects. As experimental investigations and clinical research progress, there is a possibility that a combination of newly tested medications and traditional ones may emerge as a promising treatment option for dyslipidemia in the future.
Collapse
Affiliation(s)
- Jacky Xiao Feng Huang
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Adil Yousaf
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Julie Moon
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Ramiz Ahmed
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Krishma Uppal
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Sudhakar Pemminati
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| |
Collapse
|
8
|
Patel SB, Wyne KL, Afreen S, Belalcazar LM, Bird MD, Coles S, Marrs JC, Peng CCH, Pulipati VP, Sultan S, Zilbermint M. American Association of Clinical Endocrinology Clinical Practice Guideline on Pharmacologic Management of Adults With Dyslipidemia. Endocr Pract 2025; 31:236-262. [PMID: 39919851 DOI: 10.1016/j.eprac.2024.09.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 02/09/2025]
Abstract
OBJECTIVE To review the evidence and provide updated and new recommendations for the pharmacologic management of adults with dyslipidemia to prevent adverse cardiovascular outcomes. These recommendations are intended for use by clinicians, health care team members, patients, caregivers, and other stakeholders. METHODS This guideline was developed by a multidisciplinary task force of content experts and guideline methodologists based on systematic reviews of randomized controlled trials or cohort studies from database inception to November 7, 2023. An updated literature search was completed for any additional articles published by May 31, 2024. Clinical questions addressing nonstatin medications and patient-important outcomes were prioritized. The task force assessed the certainty of the evidence and developed recommendations using the Grading of Recommendations Assessment, Development, and Evaluation framework. All recommendations were based on the consideration of the certainty of the evidence across patient-important outcomes, in addition to issues of feasibility, acceptability, equity, and patient preferences and values. RESULTS This guideline update includes 13 evidence-based recommendations for the pharmacologic management of adults with dyslipidemia focused on patient-important outcomes of atherosclerotic cardiovascular disease (ASCVD) risk reduction. The task force issued a good practice statement to assess the risk of ASCVD events for primary prevention in adults with dyslipidemia. The task force suggested the use of alirocumab, evolocumab, or bempedoic acid for adults who have ASCVD or who are at increased risk for ASCVD in addition to standard care. The task force suggested against the use of these medications in adults without ASCVD. There was insufficient evidence to recommend for or against the addition of inclisiran. For adults with hypertriglyceridemia and ASCVD or increased risk of ASCVD, the task force suggested the use of eicosapentaenoic acid but not eicosapentaenoic acid plus docosahexaenoic acid and strongly recommended against the use of niacin. There was insufficient evidence for recommendations regarding pharmacologic management in adults with severe hypertriglyceridemia (≥500 mg/dL). The task force suggested a low-density lipoprotein cholesterol treatment goal of <70 mg/dL in adults with dyslipidemia and ASCVD or at increased risk of ASCVD. CONCLUSIONS Pharmacotherapy is recommended in adults with dyslipidemia to reduce the risk of ASCVD events. There are several effective and safe treatment options for adults with dyslipidemia who have ASCVD or at increased risk of ASCVD who need additional lipid-lowering medications. Shared decision-making discussions are essential to determine the best option for each individual.
Collapse
Affiliation(s)
- Shailendra B Patel
- University of Cincinnati, Cincinnati, and Cincinnati Veterans Affairs Medical Center, Ohio
| | - Kathleen L Wyne
- The Ohio State University Wexner Medical Center, Columbus, Ohio
| | | | | | - Melanie D Bird
- American Association of Clinical Endocrinology, Jacksonville, Florida
| | - Sarah Coles
- North Country HealthCare, Flagstaff, Arizona
| | - Joel C Marrs
- University of Tennessee Health Sciences Center, Nashville, Tennessee
| | | | | | | | - Mihail Zilbermint
- Johns Hopkins University School of Medicine, Baltimore, Maryland; Johns Hopkins Community Physicians, Baltimore, Maryland
| |
Collapse
|
9
|
Kim YH, Jeong S, Cho KA, Woo SY, Han SH, Ryu KH. Reduction of Low-Density Lipoprotein Cholesterol by Mesenchymal Stem Cells in a Mouse Model of Exogenous Cushing's Syndrome. Tissue Eng Regen Med 2025; 22:237-248. [PMID: 39873947 PMCID: PMC11794754 DOI: 10.1007/s13770-024-00697-3] [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: 10/18/2024] [Revised: 12/11/2024] [Accepted: 12/22/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Exogenous Cushing's syndrome, which results from prolonged glucocorticoid treatment, is associated with metabolic abnormalities. Previously, we reported the inhibitory effect of tonsil-derived mesenchymal stem cell conditioned medium (T-MSC CM) on glucocorticoid signal transduction. In this study, we investigated the therapeutic efficacy of T-MSCs in a mouse model of exogenous Cushing's syndrome. METHODS Exogenous Cushing's syndrome model mice was generated by corticosterone administration in the drinking water for 5 weeks, and T-MSCs were injected intraperitoneally twice during the third week. Serum lipid profiles were measured using a chemistry analyzer. HepG2 cells were treated with dexamethasone and co-cultured with T-MSCs. Expression levels of genes involved in cholesterol metabolism were examined using real-time PCR. Low-density lipoprotein receptor (LDLR) protein levels were determined using western blotting and immunohistochemistry. Liver RNA extracted from the CORT and CORT + MSC mouse groups was used for transcriptome sequencing analysis and protein-protein interaction analysis. RESULTS Weight reduction and improvements in dyslipidemia by T-MSC administration were observed only in female mice. T-MSCs reduce circulating LDL cholesterol levels by downregulating liver X receptor α (LXRα) and inducible degrader of LDLR (IDOL) expression, thereby stabilizing LDLRs in the liver. Transcriptome analysis of liver tissue revealed pathways that are regulated by T-MSCs administration. CONCLUSION Administration of MSCs to female mice receiving chronic corticosterone treatment reduced the circulating LDL cholesterol level by downregulating the LXRα-IDOL axis in hepatocytes. These results suggest that T-MSCs may offer a novel therapeutic strategy for managing exogenous Cushing's syndrome by regulating cholesterol metabolism.
Collapse
Affiliation(s)
- Yu-Hee Kim
- Advance Biomedical Research Institute, Ewha Womans University Seoul Hospital, Seoul, South Korea
| | - Seonghee Jeong
- Ewha Medical Academy, Ewha Womans University Medical Center, Seoul, South Korea
| | - Kyung-Ah Cho
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - So-Youn Woo
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - Seung-Ho Han
- Ewha Medical Academy, Ewha Womans University Medical Center, Seoul, South Korea
- Department of Anatomy, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, 07804, South Korea.
| |
Collapse
|
10
|
Li W, Wu C, Li W, Li L. LDL-cholesterol lowering agents (statins and PCSK9 inhibitors) and the risk of intracerebral hemorrhage: A network meta-analysis. J Stroke Cerebrovasc Dis 2025; 34:108224. [PMID: 39755190 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 12/21/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND AND PURPOSE Statin therapy reduces the risk of ischemic stroke; however, certain studies have observed an increased incidence of intracerebral hemorrhage (ICH). Moreover, proprotein convertase subtilisin/kexin type 9(PCSK-9) inhibitors have emerged as a powerful class of lipid-lowering medications, potentially with a lower propensity for causing hemorrhagic events. To investigate this matter further, we conducted a network meta-analysis of randomized controlled trials (RCTs) involving statins and PCSK-9 inhibitors that reported occurrences of ICH. METHODS We performed a literature search of Medline, Web of Science, and The Cochrane Library from database inception until August 2023. All randomized controlled trials of statin therapy and PCSK-9 inhibitors that reported ICH or hemorrhagic stroke were included. The primary outcome variable was ICH. The risk of bias of each included study was assessed by using the Cochrane Handbook for Systematic Reviews of Interventions. We performed network meta-analysis to compare and rank statin and PCSK-9 inhibitors therapies. This study is registered (2023110026. inplasy.com). RESULTS A total of 26251 citations were identified by the search, and 38 potentially eligible articles were included. In total, data from 271411 individuals were analyzed. The data showed that there was not a significant increased risk of intracerebral hemorrhage for all statins and PCSK-9 inhibitors compared with placebo. atorvastatin and rosuvastatin were associated with a lower risk of death than placebo (ORs ranging between 0.79 and 0.82). For risk of intracerebral hemorrhage and mortality. there was not a significant increased risk among all drugs. CONCLUSIONS LDL-Cholesterol lowering agents (statins and PCSK-9 inhibitors) was not associated with a significant increased risk of ICH. Our network meta-analysis provides strong evidence for the safety of statins and PCSK-9 inhibitors, but more studies are needed to further validate this conclusion.
Collapse
Affiliation(s)
- Wangwen Li
- Department of Neurology, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing 400016, China; School of Medicine, Chongqing University, Chongqing 404010, China.
| | - Chuyue Wu
- Department of Neurology, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing 400016, China; School of Medicine, Chongqing University, Chongqing 404010, China; Chongqing Municipality Clinical Research Center for Geriatric diseases, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing 400016, China.
| | - Wenkui Li
- Department of Neurology, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing 400016, China; School of Medicine, Chongqing University, Chongqing 404010, China.
| | - Li Li
- Department of Neurology, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing 400016, China; School of Medicine, Chongqing University, Chongqing 404010, China.
| |
Collapse
|
11
|
Lian PA, Zhu WQ, Zhao WX, Huang PP, Ran JL, Tang YX, Huang XS, Li R. Lipoprotein(a) in atherosclerotic cardiovascular disease and proprotein convertase subtilisin/kexin-type 9 inhibitors. Clin Chim Acta 2025; 565:119982. [PMID: 39366516 DOI: 10.1016/j.cca.2024.119982] [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: 02/08/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
High plasma lipoprotein(a) (Lp(a)) levels increase the cardiovascular risk in populations with atherosclerotic cardiovascular disease (ASCVD). Apolipoprotein (a) [apo(a)], a unique protein component of Lp(a), plays an important role in the pathogenesis of atherosclerosis. Statins, the primary medication in managing ASCVD, lower low-density lipoprotein cholesterol (LDL-C) but concurrently elevate plasma Lp(a) levels, contributing to an increased residual cardiovascular risk. In turn, proprotein convertase subtilisin/kexin-type 9 (PCSK9) inhibitors, a novel class of LDL-C lowering drugs, effectively reduce plasma Lp(a) levels, which is believed to decrease residual cardiovascular risk. However, the mechanism by which PCSK9 inhibitors reduce Lp(a) levels remains unknown. In addition, there are some clinical limitations of PCSK9 inhibitors. Here, we systematically review the past, present, and prospects of studies pertaining to Lp(a), PCSK9 inhibitors, and ASCVD.
Collapse
Affiliation(s)
- Ping-An Lian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen-Qiang Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei-Xin Zhao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Piao-Piao Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan-Li Ran
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya-Xin Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xian-Sheng Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cardiovascular Medicine, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China
| | - Rong Li
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Stomatology, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China.
| |
Collapse
|
12
|
Movahedan M, Ellis UM, Barry AR. Efficacy of Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitors in Patients with Heterozygous Familial Hypercholesterolemia: A Meta-analysis. Am J Cardiovasc Drugs 2025; 25:47-55. [PMID: 39304616 DOI: 10.1007/s40256-024-00682-0] [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] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Patients with heterozygous familial hypercholesterolemia (HeFH) are at high risk of major adverse cardiovascular events (MACE) and mortality. Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), including monoclonal antibodies (alirocumab, evolocumab) and small interfering RNA (inclisiran), substantially reduce lipid levels. This meta-analysis aimed to evaluate the efficacy of both types of PCSK9i specifically in patients with HeFH. METHODS A librarian-assisted systematic search of MEDLINE, Embase, CENTRAL, and ClinicalTrials.gov was performed from 2013 to 2023. Randomized controlled trials of PCSK9i versus control in patients with HeFH were included. No language restrictions were applied. Cochrane Risk-of-Bias tool 2 was used to assess quality of evidence. Meta-analyses were performed using Cochrane ReviewManager. Outcomes included change in atherogenic lipids, MACE, and all-cause death. RESULTS Seven trials were included (N = 2196). Overall risk of bias was mostly low or with some concerns. Median follow-up was 24 weeks. PCSK9i had an uncertain effect on MACE (odds ratio [OR] 1.25, 95% confidence interval [CI] 0.69-2.26) and all-cause death (OR 2.47, 95% CI 0.33-18.26) due to the low event rate and short follow-up. However, PCSK9i significantly reduced low-density lipoprotein cholesterol (LDL-C) by 54% (95% CI 49-58), apolipoprotein B by 43% (95% CI 37-49), and lipoprotein(a) by 20% (95% CI 13-28). CONCLUSIONS In patients with HeFH, PCSK9i significantly reduced atherogenic lipids (LDL-C, apolipoprotein B, and lipoprotein[a]). Despite this, the effect on MACE or all-cause death was unclear. Larger-scale randomized controlled trials of longer duration are needed to validate whether this short-term reduction in lipid levels translates into a reduction in clinically meaningful outcomes.
Collapse
Affiliation(s)
- Mahsa Movahedan
- St. Paul's Hospital, Lower Mainland Pharmacy Services, Vancouver, BC, Canada
| | - Ursula M Ellis
- Woodward Library, The University of British Columbia, Vancouver, BC, Canada
| | - Arden R Barry
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Jim Pattison Outpatient Care and Surgery Centre, Lower Mainland Pharmacy Services, Surrey, BC, Canada.
| |
Collapse
|
13
|
Tong M, Palmer N, Dailamy A, Kumar A, Khaliq H, Han S, Finburgh E, Wing M, Hong C, Xiang Y, Miyasaki K, Portell A, Rainaldi J, Suhardjo A, Nourreddine S, Chew WL, Kwon EJ, Mali P. Robust genome and cell engineering via in vitro and in situ circularized RNAs. Nat Biomed Eng 2025; 9:109-126. [PMID: 39187662 DOI: 10.1038/s41551-024-01245-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 07/24/2024] [Indexed: 08/28/2024]
Abstract
Circularization can improve RNA persistence, yet simple and scalable approaches to achieve this are lacking. Here we report two methods that facilitate the pursuit of circular RNAs (cRNAs): cRNAs developed via in vitro circularization using group II introns, and cRNAs developed via in-cell circularization by the ubiquitously expressed RtcB protein. We also report simple purification protocols that enable high cRNA yields (40-75%) while maintaining low immune responses. These methods and protocols facilitate a broad range of applications in stem cell engineering as well as robust genome and epigenome targeting via zinc finger proteins and CRISPR-Cas9. Notably, cRNAs bearing the encephalomyocarditis internal ribosome entry enabled robust expression and persistence compared with linear capped RNAs in cardiomyocytes and neurons, which highlights the utility of cRNAs in these non-dividing cells. We also describe genome targeting via deimmunized Cas9 delivered as cRNA and a long-range multiplexed protein engineering methodology for the combinatorial screening of deimmunized protein variants that enables compatibility between persistence of expression and immunogenicity in cRNA-delivered proteins. The cRNA toolset will aid research and the development of therapeutics.
Collapse
Affiliation(s)
- Michael Tong
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Nathan Palmer
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Amir Dailamy
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Aditya Kumar
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Hammza Khaliq
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Sangwoo Han
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Emma Finburgh
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Madeleine Wing
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Camilla Hong
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Yichen Xiang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Katelyn Miyasaki
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Andrew Portell
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Joseph Rainaldi
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Amanda Suhardjo
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Sami Nourreddine
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Wei Leong Chew
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ester J Kwon
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Prashant Mali
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
14
|
Garwood CL, Cabral KP, Brown R, Dixon DL. Current and emerging PCSK9-directed therapies to reduce LDL-C and ASCVD risk: A state-of-the-art review. Pharmacotherapy 2025; 45:54-65. [PMID: 39679827 PMCID: PMC11755694 DOI: 10.1002/phar.4635] [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: 08/21/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of death worldwide. Lowering low-density lipoprotein cholesterol (LDL-C) levels is a primary strategy to reduce ASCVD risk. Although statin therapy remains the initial therapy of choice to reduce LDL-C and ASCVD risk, statin intolerance and suboptimal LDL-C lowering response prompts the need for additional non-statin therapies. Ezetimibe and bempedoic acid are reasonable options but they modestly reduce LDL-C levels (15% to 25%). Therapies directed at the proprotein convertase subtilisin/kexin type 9 (PCSK9) enzyme, however, reduce LDL-C levels by 50%-60% when added to background statin therapy. PCSK9 is an enzyme synthesized by the liver that facilitates the degradation of LDL receptors and prevents their recycling to the hepatocyte surface to remove LDL-C from circulation. Approaches to inhibit this effect have centered on monoclonal antibodies (mAbs) (alirocumab, evolocumab) targeting PCSK9 functionality and small interfering RNA (siRNA) therapies (inclisiran) targeting the hepatic synthesis of PCSK9. Randomized controlled trials have demonstrated beneficial cardiovascular outcomes of PCSK9 mAbs, but such evidence is not yet available for inclisiran. Current clinical practice guidelines generally recommend PCSK9-directed therapies for higher-risk patients with established ASCVD and those with familial hypercholesterolemia. This approach is, in part, due to their cost and uncertain economic value, but also because these therapies require subcutaneous administration, which is not preferred by some patients. Oral therapies targeting PCSK9 are, however, in development. This scoping review covers the development of current and emerging PCSK9-directed therapies, their efficacy, safety, and role in clinical practice.
Collapse
Affiliation(s)
- Candice L. Garwood
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health SciencesWayne State UniversityDetroitMichiganUSA
- Department of PharmacyHarper University Hospital, Detroit Medical CenterDetroitMichiganUSA
| | - Katherine P. Cabral
- Department of Pharmacy PracticeAlbany College of Pharmacy & Health SciencesAlbanyNew YorkUSA
- Capital Cardiology AssociatesAlbanyNew YorkUSA
| | - Roy Brown
- Health Sciences LibraryVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Dave L. Dixon
- Department of Pharmacotherapy & Outcomes ScienceVirginia Commonwealth University School of PharmacyRichmondVirginiaUSA
| |
Collapse
|
15
|
Zeng W, Tomlinson B. Statin alternatives for the treatment of hypercholesterolemia - a safety evaluation. Expert Opin Drug Saf 2025; 24:17-24. [PMID: 39485240 DOI: 10.1080/14740338.2024.2424411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/19/2024] [Accepted: 10/28/2024] [Indexed: 11/03/2024]
Abstract
INTRODUCTION Statins are well established as the first-line treatment to reduce low-density-lipoprotein cholesterol (LDL-C) and cardiovascular (CV) events, but some patients are unable to tolerate effective doses or sometimes any dose of statins and alternative treatments may be required. AREAS COVERED In this review we summarize the relevant published literature obtained from a PubMed search on the safety of statin alternatives for the treatment of hypercholesterolemia. EXPERT OPINION The main alternatives to statins are ezetimibe, the proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, evolocumab, alirocumab, and inclisiran, and the recently approved bempedoic acid. These have all shown an excellent safety profile and have not been associated with skeletal muscle symptoms or with increased risk of new onset diabetes and they have no major drug interactions. The injectable PCSK9 inhibitors are associated with a small increase in injection site reactions which are usually of mild or moderate intensity. Bempedoic acid is associated with a small increase in plasma uric acid and slightly increased frequency of episodes of gout in susceptible subjects. The cost and availability and the degree of lowering of LDL-C required are more likely to determine the choice of statin alternatives than the safety issues.
Collapse
Affiliation(s)
- Weiwei Zeng
- Department of Pharmacy, Shenzhen Longgang Second People's Hospital, Shenzhen, China
| | - Brian Tomlinson
- Faculty of Medicine, Macau University of Science & Technology, Macau, China
| |
Collapse
|
16
|
Tomlinson B. An up-to-date review of emerging biologic therapies for hypercholesterolemia. Expert Opin Biol Ther 2025; 25:69-78. [PMID: 39668448 DOI: 10.1080/14712598.2024.2442455] [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: 09/01/2024] [Revised: 11/20/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Hypercholesterolemia and other lipid disorders are major causes of atherosclerotic cardiovascular disease (ASCVD). Statins have been the mainstay of lipid-lowering therapy for many years, but they may not be adequate to achieve the target low-density lipoprotein (LDL) cholesterol levels and there are other residual lipid risk factors. AREAS COVERED This article reviews the biologic therapies in development for hypercholesterolemia identified by a PubMed search. Inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) is a major focus, but the drugs targeting apolipoprotein C3 (apoC3) and angiopoietin-like 3 (ANGPTL3) that were originally developed to reduce the levels of triglyceride-rich lipoproteins are now being explored to reduce cardiovascular events in a wider range of patients. A brief overview of biologic therapies targeting lipoprotein(a) [Lp(a)] is also proved. EXPERT OPINION Inhibition of PCSK9 remains an attractive target. In addition to the currently available monoclonal antibodies (mAbs) and small interfering RNA (siRNA), new mAbs and the adenectin lerodalcibep are promising therapies. The antisense oligonucleotide (ASO) and siRNA inhibitors of apoC3 and ANGPTL3 are effective in severe hypertriglyceridemia and homozygous familial hypercholesterolemia, respectively, and may prove to have wider applications. ASO and siRNA inhibitors of Lp(a) are currently in cardiovascular outcome studies.
Collapse
Affiliation(s)
- Brian Tomlinson
- Faculty of Medicine, Macau University of Science & Technology, Macau, China
| |
Collapse
|
17
|
Wang Y, Fang X, Liu J, Lv X, Lu K, Lu Y, Jiang Y. PCSK9 in T-cell function and the immune response. Biomark Res 2024; 12:163. [PMID: 39736777 DOI: 10.1186/s40364-024-00712-8] [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: 08/16/2024] [Accepted: 12/19/2024] [Indexed: 01/01/2025] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) was first reported in 2003 and confirmed to be strongly associated with familial hypercholesterolemia. Small-molecule inhibitors targeting PCSK9 provide an effective and safe method for managing hypercholesterolemia and reducing the cardiovascular risk. In recent years, increasing evidence has indicated other important roles for PCSK9 in inflammation, tumors, and even immune regulation. PCSK9 might be an attractive regulator of T-cell activation and expansion. It might mediate inflammation and regulate other types of immune cells. In this review, we summarize the current advances in the field of PCSK9 and provide a narrative of the biological processes associated with PCSK9. The relationships between PCSK9 and different T cells were investigated in depth. Finally, the signaling pathways associated with PCSK9 and the immune response are also summarized in this review.
Collapse
Affiliation(s)
- Yuying Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Jiarui Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Xiao Lv
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Kang Lu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Yingxue Lu
- Department of Nephrology, Shandong Second Provincial General Hospital, Jinan , Shandong, 250021, China
| | - Yujie Jiang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China.
| |
Collapse
|
18
|
Gibson CM, Duffy D, Bahit MC, Chi G, White H, Korjian S, Alexander JH, Lincoff AM, Heise M, Kingwell BA, Nicolau JC, Lopes RD, Cornel JH, Lewis BS, Vinereanu D, Goodman SG, Bode C, Steg PG, Libby P, Sacks FM, Bainey KR, Ridker PM, Mahaffey KW, Aylward P, Nicholls SJ, Pocock SJ, Mehran R, Harrington RA. Apolipoprotein A-I infusions and cardiovascular outcomes in acute myocardial infarction according to baseline LDL-cholesterol levels: the AEGIS-II trial. Eur Heart J 2024; 45:5023-5038. [PMID: 39221651 DOI: 10.1093/eurheartj/ehae614] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/08/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS In the AEGIS-II trial (NCT03473223), CSL112, a human apolipoprotein A1 derived from plasma that increases cholesterol efflux capacity, did not significantly reduce the risk of the primary endpoint through 90 days vs. placebo after acute myocardial infarction (MI). Nevertheless, given the well-established relationship between higher low-density lipoprotein cholesterol (LDL-C) and plaque burden, as well as greater risk reductions seen with PCSK9 inhibitors in patients with baseline LDL-C ≥ 100 mg/dL on statin therapy, the efficacy of CSL112 may be influenced by baseline LDL-C. METHODS Overall, 18 219 patients with acute MI, multivessel coronary artery disease, and additional risk factors were randomized to either four weekly infusions of 6 g CSL112 or placebo. This exploratory post-hoc analysis evaluated cardiovascular outcomes by baseline LDL-C in patients prescribed guideline-directed statin therapy at the time of randomization (n = 15 731). RESULTS As baseline LDL-C increased, the risk of the primary endpoint at 90 days lowered in those treated with CSL112 compared with placebo. In patients with LDL-C ≥ 100 mg/dL at randomization, there was a significant risk reduction of cardiovascular death, MI, or stroke in the CSL112 vs. placebo group at 90, 180, and 365 days [hazard ratio .69 (.53-.90), .71 (.57-.88), and .78 (.65-.93)]. In contrast, there was no difference between treatment groups among those with LDL-C < 100 mg/dL at baseline. CONCLUSIONS In this population, treatment with CSL112 compared to placebo was associated with a significantly lower risk of recurrent cardiovascular events among patients with a baseline LDL-C ≥ 100 mg/dL. Further studies need to confirm that CSL112 efficacy is influenced by baseline LDL-C.
Collapse
Affiliation(s)
- C Michael Gibson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 930 Commonwealth Avenue, Boston, MA 02215, USA
| | - Danielle Duffy
- Clinical Development, CSL Behring, King of Prussia, PA, USA
| | | | - Gerald Chi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 930 Commonwealth Avenue, Boston, MA 02215, USA
| | - Harvey White
- Health New Zealand - Te Whatu Ora, Te Toka Tumai, Green Lane Cardiovascular Service, Auckland City Hospital, Auckland 1142, New Zealand
| | - Serge Korjian
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 930 Commonwealth Avenue, Boston, MA 02215, USA
| | - John H Alexander
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Duke Clinical Research Institute, Duke Health, Durham, NC, USA
| | - A Michael Lincoff
- The Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Mark Heise
- Clinical Development, CSL Behring, King of Prussia, PA, USA
- Deparment of Biostatistics, CSL Behring, King of Prussia, PA, USA
| | - Bronwyn A Kingwell
- Deparment of Research and Development, CSL Limited, Melbourne, Australia
| | - Jose C Nicolau
- Instituto do Coracao (InCor), Cardiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Renato D Lopes
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Duke Clinical Research Institute, Duke Health, Durham, NC, USA
- Brazilian Clinical Research Institute, Sao Paulo, SP, Brazil
| | - Jan H Cornel
- Radboud University Medical Center, Nijmegen and Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | | | - Dragos Vinereanu
- University of Medicine and Pharmacy Carol Davila, University and Emergency Hospital, Bucharest, Romania
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Canada
- Division of Cardiology, Department of Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
- Clinical Trials and Translation Unit, Peter Munk Cardiac Center, University Health Network, Toronto, Canada
| | - Christoph Bode
- Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ph Gabriel Steg
- Université Paris-Cité, INSERM U_1148, FACT and AP-HP, Hôpital Bichat, Paris, France
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Frank M Sacks
- Cardiovascular Disease Prevention, Department of Nutrition, Harvard T.H. Chan School of Public Health, Brigham and Women's Hospital, Boston, MA, USA
| | - Kevin R Bainey
- Walter Mackenzie Health Sciences Centre, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Boston, MA, USA
| | - Kenneth W Mahaffey
- Stanford Center for Clinical Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Philip Aylward
- Department of Cardiology, South Australian Health and Medical Research Institute/SAHMRI, Adelaide, Australia
| | - Stephen J Nicholls
- Department of Cardiology, Victorian Heart Institute, Monash University, Melbourne, VIC, Australia
| | - Stuart J Pocock
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Roxana Mehran
- Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, Zena and Michael A. Wiener Cardiovascular Institute, New York, NY, USA
| | | |
Collapse
|
19
|
Zendjebil S, Steg PG. PCSK9 Monoclonal Antibodies Have Come a Long Way. Curr Atheroscler Rep 2024; 26:721-732. [PMID: 39384735 DOI: 10.1007/s11883-024-01243-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 10/11/2024]
Abstract
PURPOSE OF THE REVIEW This review examines the pivotal role of monoclonal antibodies against PCSK9 in lipid-lowering therapy, emphasizing their biological and clinical impact. RECENT FINDINGS Randomized controlled trials have validated that PCSK9 monoclonal antibodies (Mabs) effectively reduce LDL-c levels by approximately 50%, even when added to maximal statin therapy. They moreover produce a notable 15-20% relative decrease in major cardiovascular events, with a greater reduction among high-risk patients and no evidence for serious adverse effects, assuaging previous concerns. This review highlights the benefits of PCSK9 Mabs in high cardiovascular risk patients. Despite their efficacy and safety, these therapies are hindered by limited access, and require broader integration into clinical practice to optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Sandra Zendjebil
- Université Paris-Cité, Paris, France
- French Alliance for Cardiovascular Trials (FACT), INSERM U_1148/LVTS, AP-HP, Hôpital Bichat, Paris, France
- Département de Cardiologie, Hôpital Bichat, AP-HP 46 Rue Henri Huchard, 75018, Paris, France
| | - Philippe Gabriel Steg
- Université Paris-Cité, Paris, France.
- French Alliance for Cardiovascular Trials (FACT), INSERM U_1148/LVTS, AP-HP, Hôpital Bichat, Paris, France.
- Département de Cardiologie, Hôpital Bichat, AP-HP 46 Rue Henri Huchard, 75018, Paris, France.
| |
Collapse
|
20
|
Li Z, Zhu L, Xu Y, Zhang Y, Liu Y, Sun H, Li S, Wang M, Jiang T, Zhou J, Deng Q. Pleiotropic Effects of PCSK9 Inhibitors on Cardio-Cerebrovascular Diseases. Biomedicines 2024; 12:2729. [PMID: 39767636 PMCID: PMC11726846 DOI: 10.3390/biomedicines12122729] [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: 10/31/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Cardiovascular disease (CVD) and ischemic stroke (IS) are the primary causes of mortality worldwide. Hypercholesterolemia has been recognized as an independent risk factor for CVD and IS. Numerous clinical trials have unequivocally demonstrated that reducing levels of low-density lipoprotein cholesterol (LDL-C) significantly mitigates the risk of both cardiac and cerebral vascular events, thereby enhancing patient prognosis. Consequently, LDL-C reduction remains a pivotal therapeutic strategy for CVD and IS. However, despite intensive statin therapy, a significant proportion of high-risk hypercholesterolemic patients fail to achieve sufficient reductions in LDL-C levels. In response to this challenge, an inhibitor targeting proprotein convertase subtilisin-kexin type 9 (PCSK9) has been developed as a therapeutic intervention for hyperlipidemia. Numerous randomized controlled trials (RCTs) have conclusively demonstrated that the combination of PCSK9 inhibitors and statins significantly enhances prognosis not only in patients with CVD, but also in those afflicted with symptomatic intracranial artery stenosis (sICAS). PCSK9 inhibitors significantly reduce LDL-C levels by binding to the PCSK9 molecule and preventing its interaction with LDLRs. This prevents degradation of the receptor and increases uptake of LDL-C, thereby decreasing its concentration in blood. Besides significantly reducing LDL-C levels, PCSK9 inhibitors also demonstrate anti-inflammatory and anti-atherosclerotic properties while promoting plaque stabilization and inhibiting platelet aggregation and thrombosis. This article aims to provide a comprehensive review based on the relevant literature regarding the evolving understanding of pleiotropic effects associated with PCSK9 inhibitors, particularly focusing on their impact on the cardiovascular system and central nervous system.
Collapse
Affiliation(s)
- Zhenzhen Li
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (Z.L.); (L.Z.); (Y.Z.); (Y.L.); (S.L.); (M.W.); (T.J.)
| | - Lin Zhu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (Z.L.); (L.Z.); (Y.Z.); (Y.L.); (S.L.); (M.W.); (T.J.)
| | - Yeqiong Xu
- Central Laboratory of Changshu Medical Examination Institute, Changshu 215500, China;
| | - Yiting Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (Z.L.); (L.Z.); (Y.Z.); (Y.L.); (S.L.); (M.W.); (T.J.)
| | - Yukai Liu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (Z.L.); (L.Z.); (Y.Z.); (Y.L.); (S.L.); (M.W.); (T.J.)
| | - Huiling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China;
| | - Shuo Li
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (Z.L.); (L.Z.); (Y.Z.); (Y.L.); (S.L.); (M.W.); (T.J.)
| | - Meng Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (Z.L.); (L.Z.); (Y.Z.); (Y.L.); (S.L.); (M.W.); (T.J.)
| | - Teng Jiang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (Z.L.); (L.Z.); (Y.Z.); (Y.L.); (S.L.); (M.W.); (T.J.)
| | - Junshan Zhou
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (Z.L.); (L.Z.); (Y.Z.); (Y.L.); (S.L.); (M.W.); (T.J.)
| | - Qiwen Deng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (Z.L.); (L.Z.); (Y.Z.); (Y.L.); (S.L.); (M.W.); (T.J.)
| |
Collapse
|
21
|
Jeswani BM, Sharma S, Rathore SS, Nazir A, Bhatheja R, Kapoor K. PCSK9 Inhibitors: The Evolving Future. Health Sci Rep 2024; 7:e70174. [PMID: 39479289 PMCID: PMC11522611 DOI: 10.1002/hsr2.70174] [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: 06/03/2024] [Revised: 09/14/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024] Open
Abstract
Introduction PCSK9 inhibitors are a novel class of medications that lower LDL cholesterol (LDL-C) by increasing LDL receptor activity, promoting clearance of LDL-C from the bloodstream. Over the years, PCSK9 inhibitors have been explored as adjunct therapies to statins or as monotherapy in high-risk cardiovascular patients. Aim This review aims to provide an updated perspective on PCSK9 inhibitors, assessing their clinical efficacy, safety, and significance, especially in light of recent clinical trials. Methods The review examines the role of PCSK9 in cholesterol regulation and summarizes the results of major cardiovascular trials, including FOURIER, SPIRE-1, SPIRE-2, and ODYSSEY Outcomes. It also discusses emerging treatments like small interfering RNA (siRNA) therapies and evaluates PCSK9 inhibitor effects on LDL-C and lipoprotein(a) levels. Results Clinical trials have shown PCSK9 inhibitors reduce LDL-C by up to 60%. In the FOURIER trial, evolocumab reduced LDL-C by 59% and major cardiovascular events by 15%-20%. The SPIRE-2 trial, despite early termination, showed a 21% risk reduction in the primary composite endpoint with bococizumab. The ODYSSEY Outcomes trial reported a 57% LDL-C reduction with alirocumab, alongside a 15% reduction in adverse events. Emerging treatments like Inclisiran offer long-term LDL-C control with fewer doses. PCSK9 inhibitors are generally well-tolerated, with the most common side effect being injection site reactions. Conclusion PCSK9 inhibitors significantly lower LDL-C and reduce cardiovascular events, offering promising therapies for high-risk patients, including those with familial hypercholesterolemia (FH) and those who cannot tolerate statins. Future research will focus on optimizing these inhibitors, integrating complementary therapies, and exploring gene-editing technologies to improve patient outcomes.
Collapse
Affiliation(s)
- Bijay Mukesh Jeswani
- Department of MedicineGCS Medical College, Hospital & Research CentreAhmedabadIndia
| | | | | | - Abubakar Nazir
- Department of MedicineKing Edward Medical UniversityLahorePakistan
- Department of MedicineOli Health Magazine Organization, Research, and EducationKigaliRwanda
| | | | - Kapil Kapoor
- Cardiology, AdventHealth OrlandoOrlandoFloridaUSA
| |
Collapse
|
22
|
Xiao G, Gao S, Xie Y, Wang Z, Shu M. Efficacy and Safety of Evolocumab and Alirocumab as PCSK9 Inhibitors in Pediatric Patients with Familial Hypercholesterolemia: A Systematic Review and Meta-Analysis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1646. [PMID: 39459433 PMCID: PMC11509226 DOI: 10.3390/medicina60101646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/25/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: The proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitors evolocumab and alirocumab are recently developed promising drugs used for treatment of familial hypercholesterolemia (FH). This systematic review and meta-analysis aimed to thoroughly evaluate the efficacy and safety of evolocumab and alirocumab among pediatric patients with FH. Materials and Methods: A comprehensive search was conducted in PubMed, Embase, CENTRAL (Cochrane Central Register of Controlled Trials), and ClinicalTrials.gov from inception through July 2024 to identify primary interventional studies among pediatric patients with FH. Meta-analyses were performed if appropriate. Statistics were analyzed using Review Manager version 5.4 and Stata version 16.0. Results: Fourteen articles reporting nine unique studies were included. There were three randomized controlled trials (RCTs) assessing evolocumab or alirocumab involving a total of 320 pediatric patients, one cross-over trial and five single-arm or observational studies. Pooled results showed significant efficacy of evolocumab/alirocumab in reducing low-density lipoprotein cholesterol (LDL-C) (weighted mean difference [WMD]: -37.92%, 95% confidence interval [CI]: -43.06% to -32.78%; I2 = 0.0%, p = 0.60), apolipoprotein B (WMD: -33.67%, 95% CI: -38.12% to -29.22%; I2 = 0.0%, p = 0.71), and also lipoprotein(a) (WMD: -16.94%, 95% CI: -26.20% to -7.69%; I2 = 0.0%, p = 0.71) among pediatric patients with FH. The efficacies of evolocumab/alirocumab on LDL-C reduction within pediatric patients with heterozygous FH (HeFH) were consistent between studies, whereas in patients with homozygous FH (HoFH), it varied dramatically. Pediatric patients with the null/null variant may respond to the treatment. PCSK9 inhibitors were generally well tolerated within most pediatric patients, in line with previous studies among adult populations. Conclusions: The PCSK9 inhibitors evolocumab/alirocumab significantly reduced LDL-C and some other lipid parameters, such as apolipoprotein B, in pediatric patients with HeFH. These drugs may be appropriate as a potential therapy for pediatric patients with HoFH who cannot achieve LDL-C targets with other treatments. Evolocumab/alirocumab was generally well tolerated in the pediatric population.
Collapse
Affiliation(s)
- Guoguang Xiao
- Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu 610041, China; (G.X.); (S.G.); (Y.X.)
- Department of Pediatrics, West China Xiamen Hospital of Sichuan University, Xiamen 361022, China
| | - Shan Gao
- Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu 610041, China; (G.X.); (S.G.); (Y.X.)
| | - Yongmei Xie
- Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu 610041, China; (G.X.); (S.G.); (Y.X.)
| | - Zhiling Wang
- Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu 610041, China; (G.X.); (S.G.); (Y.X.)
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Min Shu
- Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu 610041, China; (G.X.); (S.G.); (Y.X.)
- Department of Pediatrics, West China Xiamen Hospital of Sichuan University, Xiamen 361022, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| |
Collapse
|
23
|
Bimal T, Szema A, Pavlovic M, Karalis D, Gianos E. Life-Threatening Reaction to Lifesaving Medication: Stepwise Approach to Severe Adverse Reactions to PCSK-9 Monoclonal Antibodies. JACC Case Rep 2024; 29:102614. [PMID: 39484310 PMCID: PMC11522809 DOI: 10.1016/j.jaccas.2024.102614] [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: 06/20/2024] [Accepted: 08/13/2024] [Indexed: 11/03/2024]
Abstract
Typical side effects of proprotein convertase subtilisin/kexin type 9 monoclonal antibodies including influenza-like illness and injection site reactions, are minor and well tolerated. This case, however, highlights a less common but severe reaction, indicating the need for clinicians to understand and manage potential rare side effects noted with biologics.
Collapse
Affiliation(s)
- Tia Bimal
- Northwell, New Hyde Park, New York, Cardiovascular Institute, Lenox Hill Hospital, Northwell Health, New York, New York, USA
| | - Anthony Szema
- Northwell, New Hyde Park, New York, Mather Hospital, Northwell Health, Port Jefferson, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Maia Pavlovic
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Dean Karalis
- Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Eugenia Gianos
- Northwell, New Hyde Park, New York, Cardiovascular Institute, Lenox Hill Hospital, Northwell Health, New York, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
24
|
Marco-Benedí V, Sánchez-Hernández RM, Díaz JL, Jarauta E, Suárez-Tembra M, Pintó X, Morillas C, Plana N, Pedro-Botet J, Civeira F. PCSK9 inhibitors on the management of primary and secondary cardiovascular prevention. Lipids Health Dis 2024; 23:290. [PMID: 39256734 PMCID: PMC11386113 DOI: 10.1186/s12944-024-02283-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/01/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) have represented an important change in the management of hypercholesterolemia, although, until now, they have barely been used. Without PCSK9i, many patients with atherosclerotic cardiovascular disease (CVD) or those at very high risk do not reach their therapeutic LDLc objectives. OBJECTIVE The analysis aimed to examine the clinical and biochemical characteristics of subjects receiving PCSK9i treatment in the Dyslipidemia Registry of the Spanish Atherosclerosis Society. METHODS All consecutive subjects aged ≥ 18 years from different Lipid Units included in the Dyslipidemia Registry of the SEA were analyzed. Inclusion criteria consisted of unrelated patients aged ≥ 18 at the time of inclusion with hypercholesterolemia (LDL-C ≥ 130 mg/dL or non-HDL-C ≥ 160 mg/dL after the exclusion of secondary causes) who were studied for at least two years after inclusion. Participants' baseline and final visit clinical and biochemical characteristics were analyzed based on whether they were on primary or secondary prevention and whether they were taking PCSK9i at the end of follow-up. RESULTS Eight hundred twenty-nine patients were analyzed, 7014 patients in primary prevention and 1281 in secondary prevention at baseline. 4127 subjects completed the required follow-up for the final analysis. The median follow-up duration was 7 years (IQR 3.0-10.0). Five hundred patients (12.1%) were taking PCSK9i at the end of the follow-up. The percentage of PCSK9i use reached 35.6% (n = 201) and 8.7% (n = 318) in subjects with and without CVD, respectively. Subjects on PCSK9i and oral lipid-lowering agents with and without CVD achieved LDLc reductions of 80.3% and 75.1%, respectively, concerning concentrations without lipid-lowering drugs. Factors associated with PCSK9i use included increasing age, LDLc without lipid-lowering drugs and the Dutch Lipid Clinic Network (DLCN) score. However, hypertension, diabetes, smoking, and LDLc after oral lipid-lowering drugs were not independent factors associated with PCSK9i prescription. In subjects with CVD, the use of PCSK9i was higher in men than in women (an odds ratio of 1.613, P = 0.048). CONCLUSIONS Approximately one-third of CVD patients received PCSK9i at the end of follow-up. The use of PCSK9i was more focused on baseline LDLc concentrations rather than on CVD risk. Women received less PCSK9i in secondary prevention compared to men.
Collapse
Affiliation(s)
- Victoria Marco-Benedí
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Saragossa, Spain.
- Unidad de Lípidos, Hospital Universitario Miguel Servet, Avda Isabel La Católica 1-3, 50009, Saragossa, Spain.
| | - Rosa M Sánchez-Hernández
- Sección de Endocrinología y Nutrición, Complejo Hospitalario Universitario Insular Materno-Infantil, Instituto Universitario de Investigaciones Biomédicas y Sanitarias de La Universidad de Las Palmas de Gran Canaria, de Las Palmas de Gran Canaria, Spain
| | - José Luis Díaz
- Unidad de Lípidos y Riesgo Cardiovascular, Medicina Interna, Hospital Universitario A Coruña, A Coruña, Spain
| | - Estíbaliz Jarauta
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Saragossa, Spain
- Unidad de Lípidos, Hospital Universitario Miguel Servet, Avda Isabel La Católica 1-3, 50009, Saragossa, Spain
| | | | - Xavier Pintó
- Unidad de Lípidos y Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge, CIBEROBN, Barcelona, Spain
| | - Carlos Morillas
- Unidad de Lípidos Clínico-Epidemiológica, Hospital Universitario Dr. Peset, Valencia, Spain
| | - Núria Plana
- Unitat de Medicina Vascular I Metabolisme, Hospital Sant Joan de Reus, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Juan Pedro-Botet
- Unidad de Lípidos y Riesgo Vascular, Servicio de Endocrinología y Nutrición, Hospital del Mar, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Fernando Civeira
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Saragossa, Spain.
- Unidad de Lípidos, Hospital Universitario Miguel Servet, Avda Isabel La Católica 1-3, 50009, Saragossa, Spain.
| |
Collapse
|
25
|
Kastrati L, Raeisi-Dehkordi H, Llanaj E, Quezada-Pinedo HG, Khatami F, Ahanchi NS, Llane A, Meçani R, Muka T, Ioannidis JPA. Agreement Between Mega-Trials and Smaller Trials: A Systematic Review and Meta-Research Analysis. JAMA Netw Open 2024; 7:e2432296. [PMID: 39240561 PMCID: PMC11380108 DOI: 10.1001/jamanetworkopen.2024.32296] [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: 06/08/2024] [Accepted: 07/12/2024] [Indexed: 09/07/2024] Open
Abstract
Importance Mega-trials can provide large-scale evidence on important questions. Objective To explore how the results of mega-trials compare with the meta-analysis results of trials with smaller sample sizes. Data Sources ClinicalTrials.gov was searched for mega-trials until January 2023. PubMed was searched until June 2023 for meta-analyses incorporating the results of the eligible mega-trials. Study Selection Mega-trials were eligible if they were noncluster nonvaccine randomized clinical trials, had a sample size over 10 000, and had a peer-reviewed meta-analysis publication presenting results for the primary outcome of the mega-trials and/or all-cause mortality. Data Extraction and Synthesis For each selected meta-analysis, we extracted results of smaller trials and mega-trials included in the summary effect estimate and combined them separately using random effects. These estimates were used to calculate the ratio of odds ratios (ROR) between mega-trials and smaller trials in each meta-analysis. Next, the RORs were combined using random effects. Risk of bias was extracted for each trial included in our analyses (or when not available, assessed only for mega-trials). Data analysis was conducted from January to June 2024. Main Outcomes and Measures The main outcomes were the summary ROR for the primary outcome and all-cause mortality between mega-trials and smaller trials. Sensitivity analyses were performed with respect to the year of publication, masking, weight, type of intervention, and specialty. Results Of 120 mega-trials identified, 41 showed a significant result for the primary outcome and 22 showed a significant result for all-cause mortality. In 35 comparisons of primary outcomes (including 85 point estimates from 69 unique mega-trials and 272 point estimates from smaller trials) and 26 comparisons of all-cause mortality (including 70 point estimates from 65 unique mega-trials and 267 point estimates from smaller trials), no difference existed between the outcomes of the mega-trials and smaller trials for primary outcome (ROR, 1.00; 95% CI, 0.97-1.04) nor for all-cause mortality (ROR, 1.00; 95% CI, 0.97-1.04). For the primary outcomes, smaller trials published before the mega-trials had more favorable results than the mega-trials (ROR, 1.05; 95% CI, 1.01-1.10) and subsequent smaller trials published after the mega-trials (ROR, 1.10; 95% CI, 1.04-1.18). Conclusions and Relevance In this meta-research analysis, meta-analyses of smaller studies showed overall comparable results with mega-trials, but smaller trials published before the mega-trials gave more favorable results than mega-trials. These findings suggest that mega-trials need to be performed more often given the relative low number of mega-trials found, their low significant rates, and the fact that smaller trials published prior to mega-trial report more beneficial results than mega-trials and subsequent smaller trials.
Collapse
Affiliation(s)
- Lum Kastrati
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Hamidreza Raeisi-Dehkordi
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Erand Llanaj
- Epistudia, Bern, Switzerland
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- German Centre for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Hugo G. Quezada-Pinedo
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Farnaz Khatami
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
- Community Medicine Department, Tehran University of Medical Sciences, Tehran, Iran
| | - Noushin Sadat Ahanchi
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
- Department of Internal Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | | | - Renald Meçani
- Epistudia, Bern, Switzerland
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Taulant Muka
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California
- Epistudia, Bern, Switzerland
| | - John P. A. Ioannidis
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California
- Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California
- Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, California
| |
Collapse
|
26
|
Repp ML, Edwards MD, Burch CS, Rao A, Chinyere IR. PCSK9 Inhibitors and Anthracyclines: The Future of Cardioprotection in Cardio-Oncology. HEARTS 2024; 5:375-388. [PMID: 39268545 PMCID: PMC11391951 DOI: 10.3390/hearts5030027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024] Open
Abstract
The field of cardio-oncology is an expanding frontier within cardiovascular medicine, and the need for evidence-based guidelines is apparent. One of the emerging focuses within cardio-oncology is the concomitant use of medications for cardioprotection in the setting of chemotherapy regimens that have known cardiovascular toxicity. While clinical trials focusing on cardioprotection during chemotherapy are sparse, an inaugural trial exploring the prophylactic potential of Sodium-Glucose Cotransporter-2 inhibitors (SGLT2is) for anthracycline (ANT)-induced cardiotoxicity has recently commenced. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, though less studied in this oncology demographic, have exhibited promise in preclinical studies for conferring cardiac protection during non-ischemic toxic insults. While primarily used to reduce low-density lipoprotein, PCSK9 inhibitors exhibit pleiotropic effects, including the attenuation of inflammation, reactive oxygen species, and endothelial dysfunction. In ANT-induced cardiotoxicity, these same processes are accelerated, resulting in premature termination of treatment, chronic cardiovascular sequelae, heart failure, and/or death. This review serves a dual purpose: firstly, to provide a concise overview of the mechanisms implicated in ANT-induced cardiotoxicity, and, finally, to summarize the existing preclinical data supporting the theoretical possibility of the cardioprotective effects of PCSK9 inhibition in ANT-induced cardiotoxicity.
Collapse
Affiliation(s)
- Matthew L Repp
- Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Mark D Edwards
- Department of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christopher S Burch
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Amith Rao
- Department of Medicine, Banner University Medicine, Tucson, AZ 85724, USA
| | - Ikeotunye Royal Chinyere
- Department of Medicine, Banner University Medicine, Tucson, AZ 85724, USA
- Sarver Heart Center, University of Arizona, 1501 North Campbell Avenue, Room 6154, Tucson, AZ 85724, USA
| |
Collapse
|
27
|
Bosco G, Mszar R, Piro S, Sabouret P, Gallo A. Cardiovascular Risk Estimation and Stratification Among Individuals with Hypercholesterolemia. Curr Atheroscler Rep 2024; 26:537-548. [PMID: 38965183 DOI: 10.1007/s11883-024-01225-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
PURPOSE OF THE REVIEW This review aims to assess the variability in considering hypercholesterolemia for cardiovascular risk stratification in the general population. Recent literature on the integration of hypercholesterolemia into clinical risk scores and its interaction with other risk factors will be explored. RECENT FINDINGS The impact of hypercholesterolemia on risk estimation varies among different cardiovascular risk calculators. Elevated lipid levels during early life stages contribute to atherosclerotic plaque development, influencing disease severity despite later treatment initiation. The interplay between low-density lipoprotein cholesterol (LDLc), inflammatory markers and non-LDL lipid parameters enhances cardiovascular risk stratification. Studies have also examined the role of coronary artery calcium (CAC) score as a negative risk marker in populations with severe hypercholesterolemia. Furthermore, polygenic risk scores (PRS) may aid in diagnosing non-monogenic hypercholesterolemia, refining cardiovascular risk stratification and guiding lipid-lowering therapy strategies. Understanding the heterogeneity in risk estimation and the role of emerging biomarkers and imaging techniques is crucial for optimizing cardiovascular risk prediction and guiding personalized treatment strategies in individuals with hypercholesterolemia.
Collapse
Affiliation(s)
- Giosiana Bosco
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Sorbonne Université, INSERM UMR1166, Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié-Salpètriêre, 47/83 Boulevard de L'Hôpital, 75013, Paris, France
| | - Reed Mszar
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Salvatore Piro
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Pierre Sabouret
- Heart Institute, Cardiology Department, Sorbonne University, 47-83 Boulevard de L'Hôpital, 75013, Paris, FR, France
- National College of French Cardiologists, 13 Rue Niepce, 75014, Paris, FR, France
| | - Antonio Gallo
- Sorbonne Université, INSERM UMR1166, Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié-Salpètriêre, 47/83 Boulevard de L'Hôpital, 75013, Paris, France.
| |
Collapse
|
28
|
Ou Z, Wang F, Chen Y, Liu X, Ran B, Yin Y, Cui K. Comparative Efficacy of Colchicine and Intensive Low-density Lipoprotein Cholesterol Lowering in Patients with Atherosclerotic Diseases receiving Statins: A Network Meta-analysis of Randomized Controlled Trials. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07622-9. [PMID: 39207624 DOI: 10.1007/s10557-024-07622-9] [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] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
AIMS Adding intensive low-density lipoprotein cholesterol (LDL-C)-lowering agents or colchicine to statin has been shown to result in additional cardiovascular benefits for patients with atherosclerotic cardiovascular diseases (ASCVD). We aimed to compare the efficacy and safety of these supplementary agents in patients with ASCVD receiving statin. METHODS We performed a systematic review and frequentist network meta-analysis of randomized controlled trials. The primary efficacy endpoint was the main adverse cardiovascular event (MACE), and the secondary efficacy endpoints were myocardial infarct, stroke, coronary revascularization, cardiovascular death, and all-cause mortality, respectively. The safety endpoints were treatment discontinuation and non-cardiovascular death. We obtained estimates for efficacy outcomes and safety endpoints and presented these estimates as risk ratio (RR) with 95% confidence intervals. We ranked the comparative efficacy and safety of all drugs with P-scores. RESULTS Seventeen trials totaling 85,823 participants treated with colchicine (5926 participants), intensive LDL-C lowering (37,854 participants) via proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor, Niemann-Pick C1-like 1 protein (NPC1L1) inhibitor or ATP citrate lyase (ACL) inhibitor, or statin alone (42,043 participants) were included. Colchicine was associated with a greater reduction in the risk of MACE (RR 0.72, 0.69-0.91), stroke (RR 0.55, 0.33-0.92), and coronary revascularization (RR 0.73, 0.60-0.90) compared with NPC1L1 inhibitor, and it provided a larger reduction in the risk of MACE (RR 0.79, 0.69-0.91) compared to PCSK9 inhibitor. However, colchicine was associated with increased risk of non-cardiovascular death compared with NPC1L1 inhibitor (RR 1.48, 1.04-2.10) and PCSK9 inhibitor (RR 1.57, 1.08-2.27). Although no regimen prolonged survival, colchicine had worse performance on non-cardiovascular death and all-cause mortality. CONCLUSIONS In patients with ASCVD receiving statin, colchicine seems to be more effective than intensive LDL-C-lowering therapy with PCSK9 inhibitor or NPC1L1 inhibitor for cardiovascular prevention. However, using colchicine as an alternative to intensive LDL-C-lowering therapy may need to be weighed against the cardiovascular benefits and the potential harms of higher non-cardiovascular death. TRIAL REGISTRATION PROSPERO Identifier: CRD42023441385.
Collapse
Affiliation(s)
- Zhenhong Ou
- Department of Cardiology, Chongqing General Hospital of Chongqing University, Chongqing, China
- Department of Cardiology, Chongqing General Hospital, No. 118 Xingguang Avenue, Liangjiang New Area, Chongqing, 401120, China
| | - Fangchao Wang
- Department of Cardiology, Chongqing General Hospital of Chongqing University, Chongqing, China
- Department of Cardiology, Chongqing General Hospital, No. 118 Xingguang Avenue, Liangjiang New Area, Chongqing, 401120, China
| | - Yunlin Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xueyuan Liu
- Department of Cardiology, Graduate School of Medicine, Keio University, Tokyo, Japan
| | - Boli Ran
- Department of Cardiology, Chongqing General Hospital of Chongqing University, Chongqing, China
- Department of Cardiology, Chongqing General Hospital, No. 118 Xingguang Avenue, Liangjiang New Area, Chongqing, 401120, China
| | - Yuehui Yin
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kun Cui
- Department of Cardiology, Chongqing General Hospital of Chongqing University, Chongqing, China.
- Department of Cardiology, Chongqing General Hospital, No. 118 Xingguang Avenue, Liangjiang New Area, Chongqing, 401120, China.
| |
Collapse
|
29
|
Barbati G, Gregorio C, Scagnetto A, Indennidate C, Cappelletto C, Di Lenarda A. Effectiveness of PCSK9 inhibitors: A Target Trial Emulation framework based on Real-World Electronic Health Records. PLoS One 2024; 19:e0309470. [PMID: 39173034 PMCID: PMC11341039 DOI: 10.1371/journal.pone.0309470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Low-Density Lipoprotein (LDL) cholesterol is one of the main target for cardiovascular (CV) prevention and therapy. In the last years, Proprotein Convertase Subtilisin-Kexin type 9 inhibitors (PCSK9-i) has emerged as a key therapeutic target to lower LDL and were introduced for prevention of CV events. Recently (June 2022) the Italian Medicines Agency (AIFA) modified the eligibility criteria for the use of PCSK9-i. We designed an observational study to estimate the prevalence of eligible subjects and evaluate the effectiveness of PCSK9-i applying a Target Trial Emulation (TTE) approach based on Electronic Health Records (EHR). Subjects meeting the eligibility criteria were identified from July 2017 (when PCSK9-i became available) to December 2020. Outcomes were all-cause death and the first hospitalization. Among eligible subjects, we identified those treated at date of the first prescription. Inverse Probability of Treatment Weights (IPTW) were estimated including demographic and clinical covariates, history of treatment with statins and the month/year eligibility date. Competing risk models on weighted cohorts were used to derive the Average Treatment Effect (ATE) and the Conditional Average Treatment Effect (CATE) in subgroups of interest. Out of 1976 eligible subjects, 161 (8%) received treatment with PCSK9-i. Treated individuals were slightly younger, predominantly male, had more severe CV conditions, and were more often treated with statin compared to the untreated subjects. The latter exhibited a higher prevalence of non-CV comorbidities. A significant absolute and relative risk reduction of death and a lower relative risk for the first hospitalization was observed. The risk reduction for death was confirmed in CATE analysis. PCSk9-i were prescribed to a minority of eligible subjects. Within the TTE framework, the analysis confirmed the association between PCSK9-i and lower risk of events, aligning with findings from randomized clinical trials (RCTs). In our study, PCSK9-i provided protection specifically against all-cause death, expanding upon the evidence from RCTs that had primarily focused on composite CV outcomes.
Collapse
Affiliation(s)
- Giulia Barbati
- Biostatistics Unit, Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Caterina Gregorio
- Biostatistics Unit, Department of Medical Sciences, University of Trieste, Trieste, Italy
- MOX—Modelling and Scientific Computing Laboratory, Department of Mathematics, Politecnico di Milano, Milano, Italy
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Arjuna Scagnetto
- Cardiovascular Center, Territorial Specialistic Department, University Hospital and Health Services of Trieste, Trieste, Italy
| | - Carla Indennidate
- Cardiovascular Center, Territorial Specialistic Department, University Hospital and Health Services of Trieste, Trieste, Italy
| | - Chiara Cappelletto
- Cardiovascular Center, Territorial Specialistic Department, University Hospital and Health Services of Trieste, Trieste, Italy
| | - Andrea Di Lenarda
- Cardiovascular Center, Territorial Specialistic Department, University Hospital and Health Services of Trieste, Trieste, Italy
| |
Collapse
|
30
|
Bashir B, Schofield J, Downie P, France M, Ashcroft DM, Wright AK, Romeo S, Gouni-Berthold I, Maan A, Durrington PN, Soran H. Beyond LDL-C: unravelling the residual atherosclerotic cardiovascular disease risk landscape-focus on hypertriglyceridaemia. Front Cardiovasc Med 2024; 11:1389106. [PMID: 39171323 PMCID: PMC11335737 DOI: 10.3389/fcvm.2024.1389106] [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: 02/20/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024] Open
Abstract
Aims Historically, atherosclerotic cardiovascular disease (ASCVD) risk profile mitigation has had a predominant focus on low density lipoprotein cholesterol (LDL-C). In this narrative review we explore the residual ASCVD risk profile beyond LDL-C with a focus on hypertriglyceridaemia, recent clinical trials of therapeutics targeting hypertriglyceridaemia and novel modalities addressing other residual ASCVD risk factors. Findings Hypertriglyceridaemia remains a significant ASCVD risk despite low LDL-C in statin or proprotein convertase subtilisin/kexin type 9 inhibitor-treated patients. Large population-based observational studies have consistently demonstrated an association between hypertriglyceridaemia with ASCVD. This relationship is complicated by the co-existence of low high-density lipoprotein cholesterol. Despite significantly improving atherogenic dyslipidaemia, the most recent clinical trial outcome has cast doubt on the utility of pharmacologically lowering triglyceride concentrations using fibrates. On the other hand, purified eicosapentaenoic acid (EPA), but not in combination with docosahexaenoic acid (DHA), has produced favourable ASCVD outcomes. The outcome of these trials suggests alternate pathways involved in ASCVD risk modulation. Several other pharmacotherapies have been proposed to address other ASCVD risk factors targeting inflammation, thrombotic and metabolic factors. Implications Hypertriglyceridaemia poses a significant residual ASCVD risk in patients already on LDL-C lowering therapy. Results from pharmacologically lowering triglyceride are conflicting. The role of fibrates and combination of EPA and DHA is under question but there is now convincing evidence of ASCVD risk reduction with pure EPA in a subgroup of patients with hypertriglyceridaemia. Clinical guidelines should be updated in line with recent clinical trials evidence. Novel agents targeting non-conventional ASCVD risks need further evaluation.
Collapse
Affiliation(s)
- Bilal Bashir
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Department of Endocrinology, Diabetes & Metabolism, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- NIHR/Wellcome Trust Clinical Research Facility, Manchester, United Kingdom
| | - Jonathan Schofield
- Department of Endocrinology, Diabetes & Metabolism, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Paul Downie
- Department of Clinical Biochemistry, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Michael France
- Department of Clinical Biochemistry, Central Manchester University Hospitals, NHS Foundation Trust, Manchester, United Kingdom
| | - Darren M. Ashcroft
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Centre for Pharmacoepidemiology and Drug Safety, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Alison K. Wright
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Centre for Pharmacoepidemiology and Drug Safety, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
- Cardiology Department, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ioanna Gouni-Berthold
- Centre for Endocrinology, Diabetes and Preventive Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Akhlaq Maan
- Department of Endocrinology, Diabetes & Metabolism, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Paul N. Durrington
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Handrean Soran
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Department of Endocrinology, Diabetes & Metabolism, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- NIHR/Wellcome Trust Clinical Research Facility, Manchester, United Kingdom
| |
Collapse
|
31
|
Chen T, Liu N. How safe are proprotein convertase subtilisinekexin type 9 inhibitors in diabetes? Curr Opin Lipidol 2024; 35:187-194. [PMID: 38527426 DOI: 10.1097/mol.0000000000000934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
PURPOSE OF REVIEW To examine the safety of proprotein convertase subtilisinekexin type 9 (PCSK9) inhibitors in patients with diabetes, specifically focusing on their impact on glucose metabolism. RECENT FINDINGS Patients with diabetes often require intensified lipid-lowering therapy. PCSK9 inhibitors can reduce low-density lipoprotein cholesterol (LDL-C) concentrations by approximately 60%, and significantly reduce cardiovascular risk when added to statin therapy. Some studies have suggested an association between low LDL-C levels and an increased risk of new-onset diabetes, and genetics has almost consistently shown an increased glucose concentration and risk of diabetes. Most clinical trials have not demonstrated a deterioration in glycaemic control in patients with diabetes after the use of PCSK9 inhibitors, and they do not lead to other significant treatment-emergent adverse events. SUMMARY Although the majority of patients with diabetes are undergoing background statin therapy, which may mask potential adverse effects of PCSK9 inhibitors on glycaemic control, current data suggest that the benefits outweigh the risks for diabetic patients using PCSK9 inhibitors. Considering the different nature of genetic studies and of clinical trials, close monitoring of glucose parameters is necessary, especially in individuals with prediabetes.
Collapse
Affiliation(s)
- Tian Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | | |
Collapse
|
32
|
De Oliveira-Gomes D, Joshi PH, Peterson ED, Rohatgi A, Khera A, Navar AM. Apolipoprotein B: Bridging the Gap Between Evidence and Clinical Practice. Circulation 2024; 150:62-79. [PMID: 38950110 PMCID: PMC11219008 DOI: 10.1161/circulationaha.124.068885] [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: 07/03/2024]
Abstract
Despite data suggesting that apolipoprotein B (apoB) measurement outperforms low-density lipoprotein cholesterol level measurement in predicting atherosclerotic cardiovascular disease risk, apoB measurement has not become widely adopted into routine clinical practice. One barrier for use of apoB measurement is lack of consistent guidance for clinicians on how to interpret and apply apoB results in clinical context. Whereas guidelines have often provided clear low-density lipoprotein cholesterol targets or triggers to initiate treatment change, consistent targets for apoB are lacking. In this review, we synthesize existing data regarding the epidemiology of apoB by comparing guideline recommendations regarding use of apoB measurement, describing population percentiles of apoB relative to low-density lipoprotein cholesterol levels, summarizing studies of discordance between low-density lipoprotein cholesterol and apoB levels, and evaluating apoB levels in clinical trials of lipid-lowering therapy to guide potential treatment targets. We propose evidence-guided apoB thresholds for use in cholesterol management and clinical care.
Collapse
Affiliation(s)
- Diana De Oliveira-Gomes
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Parag H Joshi
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric D Peterson
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anand Rohatgi
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amit Khera
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ann Marie Navar
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
33
|
Gregorio C, Rea F, Ieva F, Scagnetto A, Indennidate C, Cappelletto C, Di Lenarda A, Barbati G. Flexible Approaches Based on Multistate Models and Microsimulation to Perform Real-World Cost-Effectiveness Analyses: An Application to Proprotein Convertase Subtilisin-Kexin Type 9 Inhibitors. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:897-906. [PMID: 38548178 DOI: 10.1016/j.jval.2024.03.008] [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: 09/12/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/26/2024]
Abstract
OBJECTIVES This study aims to show the application of flexible statistical methods in real-world cost-effectiveness analyses applied in the cardiovascular field, focusing specifically on the use of proprotein convertase subtilisin-kexin type 9 inhibitors for hyperlipidemia. METHODS The proposed method allowed us to use an electronic health database to emulate a target trial for cost-effectiveness analysis using multistate modeling and microsimulation. We formally established the study design and provided precise definitions of the causal measures of interest while also outlining the assumptions necessary for accurately estimating these measures using the available data. Additionally, we thoroughly considered goodness-of-fit assessments and sensitivity analyses of the decision model, which are crucial to capture the complexity of individuals' healthcare pathway and to enhance the validity of this type of health economic models. RESULTS In the disease model, the Markov assumption was found to be inadequate, and a "time-reset" timescale was implemented together with the use of a time-dependent variable to incorporate past hospitalization history. Furthermore, the microsimulation decision model demonstrated a satisfying goodness of fit, as evidenced by the consistent results obtained in the short-term horizon compared with a nonmodel-based approach. Notably, proprotein convertase subtilisin-kexin type 9 inhibitors revealed their favorable cost-effectiveness only in the long-term follow-up, with a minimum willingness to pay of 39 000 Euro/life years gained. CONCLUSIONS The approach demonstrated its significant utility in several ways. Unlike nonmodel-based or alternative model-based methods, it enabled to (1) investigate long-term cost-effectiveness comprehensively, (2) use an appropriate disease model that aligns with the specific problem under study, and (3) conduct subgroup-specific cost-effectiveness analyses to gain more targeted insights.
Collapse
Affiliation(s)
- Caterina Gregorio
- MOX-Modelling and Scientific Computing Laboratory, Department of Mathematics, Politecnico di Milano, Milano, Italy; Biostatistics Unit, Department of Medical Sciences, University of Trieste, Trieste, Italy; Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden.
| | - Federico Rea
- Department of Statistics and Quantitative Methods, Unit of Biostatistics, Epidemiology and Public Health, University of Milano-Bicocca, Milan, Italy; National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, Milan, Italy
| | - Francesca Ieva
- MOX-Modelling and Scientific Computing Laboratory, Department of Mathematics, Politecnico di Milano, Milano, Italy; HDS, Health Data Science center, Human Technopole, Milan, Italy
| | - Arjuna Scagnetto
- Cardiovascular Center, Territorial Specialistic Department, University Hospital and Health Services of Trieste, Italy
| | - Carla Indennidate
- Cardiovascular Center, Territorial Specialistic Department, University Hospital and Health Services of Trieste, Italy
| | - Chiara Cappelletto
- Cardiovascular Center, Territorial Specialistic Department, University Hospital and Health Services of Trieste, Italy
| | - Andrea Di Lenarda
- Cardiovascular Center, Territorial Specialistic Department, University Hospital and Health Services of Trieste, Italy
| | - Giulia Barbati
- Biostatistics Unit, Department of Medical Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Two large cardiovascular outcomes trials of monoclonal antibodies against proprotein convertase subtilisin/kexin type 9 (PCSK9) demonstrated that therapeutic inhibition of extracellular PCSK9 markedly reduces LDL cholesterol concentration and cardiovascular risk. Several novel strategies to inhibit PCSK9 function are in development. Different mechanisms of action may determine specific properties with potential relevance for patient care. RECENT FINDINGS For the monoclonal antibodies evolocumab und alirocumab as first-generation PCSK9 inhibitors, follow-up data of up to 8 years of exposure complement the information on efficacy and safety available from outcome trials. For the small-interfering RNA inclisiran as second-generation PCSK9 inhibitor, several phase III trials have been published and a cardiovascular outcome trial has completed recruitment and is ongoing. Third-generation PCSK9 inhibitors encompass, among others, orally available drugs such as MK-0616 and the fusion protein lerodalcibep. Additional strategies to inhibit PCSK9 include vaccination and gene editing. SUMMARY Long-term inhibition of PCSK9 with monoclonal antibodies is safe and conveys sustained cardiovascular benefit. Novel strategies to inhibit PCSK9 function such as orally available drugs, RNA targeting, and one-time treatment with gene editing may further enhance the therapeutic armamentarium and enable novel preventive strategies.
Collapse
Affiliation(s)
- Julius L Katzmann
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | | |
Collapse
|
35
|
Mhaimeed O, Burney ZA, Schott SL, Kohli P, Marvel FA, Martin SS. The importance of LDL-C lowering in atherosclerotic cardiovascular disease prevention: Lower for longer is better. Am J Prev Cardiol 2024; 18:100649. [PMID: 38576462 PMCID: PMC10992711 DOI: 10.1016/j.ajpc.2024.100649] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/25/2024] [Accepted: 03/09/2024] [Indexed: 04/06/2024] Open
Abstract
Cumulative exposure to low-density lipoprotein cholesterol (LDL-C) is a key driver of atherosclerotic cardiovascular disease (ASCVD) risk. An armamentarium of therapies to achieve robust and sustained reduction in LDL-C can reduce ASCVD risk. The gold standard for LDL-C assessment is ultracentrifugation but in routine clinical practice LDL-C is usually calculated and the most accurate calculation is the Martin/Hopkins equation. For primary prevention, consideration of estimated ASCVD risk frames decision making regarding use of statins and other therapies, and tools such as risk enhancing factors and coronary artery calcium enable tailoring of risk assessment and decision making. In patients with diabetes, lipid lowering therapy is recommended in most patients to reduce ASCVD risk with an opportunity to tailor therapy based on other risk factors. Patients with primary hypercholesterolemia and familial hypercholesterolemia (FH) with baseline LDL-C greater than or equal to 190 mg/dL are at elevated risk, and LDL-C lowering with high-intensity statin therapy is often combined with non-statin therapies to prevent ASCVD. Secondary prevention of ASCVD, including in patients with prior myocardial infarction or stroke, requires intensive lipid lowering therapy and lifestyle modification approaches. There is no established LDL-C level below which benefit ceases or safety concerns arise. When further LDL-C lowering is required beyond lifestyle modifications and statin therapy, additional medications include oral ezetimibe and bempedoic acid, or injectables such as PCSK9 monoclonal antibodies or siRNA therapy. A novel agent that acts independently of hepatic LDL receptors is evinacumab, which is approved for patients with homozygous FH. Other emerging agents are targeted at Lp(a) and CETP. In light of the expanding lipid treatment landscape, this manuscript reviews the importance of early, intensive, and sustained LDL-C-lowering for primary and secondary prevention of ASCVD.
Collapse
Affiliation(s)
- Omar Mhaimeed
- Department of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Zain A Burney
- Department of Medicine, Cleveland Clinic, Cleveland, OH, United States
| | - Stacey L Schott
- Department of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Payal Kohli
- Department of Cardiology, University of Colorado Anschutz, Aurora, CO, United States
- Department of Cardiology, Veterans Affairs Hospital, Aurora, CO, United States
- Cherry Creek Heart, Aurora, CO, United States
- Tegna Broadcasting, MD, United States
| | - Francoise A Marvel
- Department of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Seth S Martin
- Department of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
36
|
Abbasi S, Khan A, Choudhry MW. New Insights Into the Treatment of Hyperlipidemia: Pharmacological Updates and Emerging Treatments. Cureus 2024; 16:e63078. [PMID: 38919858 PMCID: PMC11196920 DOI: 10.7759/cureus.63078] [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: 06/24/2024] [Indexed: 06/27/2024] Open
Abstract
Cardiovascular diseases are the leading causes of global mortality and morbidity. Hyperlipidemia is a significant risk factor for atherosclerosis and subsequent cardiovascular diseases. Hyperlipidemia is characterized by imbalances in blood cholesterol levels, particularly elevated low-density lipoprotein cholesterol and triglycerides, and is influenced by genetic and environmental factors. Current management consists of lifestyle modifications and pharmacological interventions most commonly consisting of statins. This review paper explores pathophysiology, management strategies, and pharmacotherapies including commonly used well-established medications including statins, fibrates, and ezetimibe, exciting novel therapies including proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, and RNA interference therapies (inclisiran), lomitapide, and bempedoic acid, highlighting their mechanisms of action, clinical efficacy, and safety profiles. Additionally, emerging therapies under clinical trials including ApoC-III inhibitors, DGAT2 inhibitors, ACAT2 Inhibitors, and LPL gene therapies are examined for their potential to improve lipid homeostasis and cardiovascular outcomes. The evolving landscape of hyperlipidemia management underscores the importance of continued research into both established therapies and promising new candidates, offering hope for more effective treatment strategies in the future.
Collapse
Affiliation(s)
| | - Adnan Khan
- Cardiology, St. Joseph's Medical Center, Stockton, USA
| | | |
Collapse
|
37
|
Song R, Li J, Xiong Y, Huang H, Liu X, Li Q. Efficacy and safety of proprotein convertase subtilisin kexin type (PCSK9) inhibitors in patients with acute coronary syndrome: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e38360. [PMID: 39259104 PMCID: PMC11142774 DOI: 10.1097/md.0000000000038360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The effect of proprotein convertase subtilisin kexin type (PCSK9) inhibitors on blood lipids and major adverse cardiovascular events (MACEs) is still controversial for acute coronary syndrome (ACS) patients. This study aimed to evaluate the efficacy and safety of PCSK9 inhibitors for ACS patients. METHODS We searched the following databases until March 2023: PubMed, Embase, Cochrane, Web of Science, CNKI, Chongqing VIP Database and Wan Fang Database. Finally, all randomized controlled trials, retrospective studies and prospective studies were included in the analysis. RESULTS A total of 20 studies involving 48,621 patients were included in this meta-analysis. The results demonstrated that PCSK9 inhibitors group was more beneficial for ACS patients compared to control group (receiving statins alone or placebo). The meta-analysis showed: there was no significant difference in high density lipoprotein cholesterol between PCSK9 inhibitors group and control group (standard mean difference = 0.17, 95% confidence interval [CI]: -0.02 to 0.36, P = .08), while the level of low density lipoprotein cholesterol in PCSK9 inhibitors group was lower than that in control group (standard mean difference = -2.32, 95% CI: -2.81 to -1.83, P < .00001). Compared with the control group, the PCSK9 inhibitors group also decreased the levels of total cholesterol and triglycerides (mean difference = -1.24, 95% CI: -1.40 to -1.09, P < .00001, mean difference = -0.36, 95% CI: -0.56 to -0.16, P = .0004). Moreover, compared with the control group, PCSK9 inhibitors group could reduce the incidence of MACEs (relative risk [RR] = 0.87, 95% CI: 0.83-0.91; P < .00001). However, this study showed that the incidence of drug-induced adverse events in PCSK9 inhibitors group was higher than that in the control group (RR = 1.15, 95% CI: 1.05-1.25, P < .0001). CONCLUSION Although this study demonstrates that PCSK9 inhibitors have higher drug-induced adverse events, they can not only reduce low-density lipoprotein cholesterol levels but also reduce the incidence of MACEs simultaneously. However, these findings needed to be further verified through large sample, multicenter, double-blind randomized controlled trials.
Collapse
Affiliation(s)
- Ruohong Song
- Department of Cardiology, Sichuan Tianfu New District People’s Hospital, Chengdu, China
| | - Jinsong Li
- Department of Cardiology, Sichuan Provincial People’s Hospital, Chengdu, China
| | - Yan Xiong
- Department of Cardiology, Sichuan Provincial People’s Hospital, Chengdu, China
| | - Hui Huang
- Department of Cardiology, Sichuan Provincial People’s Hospital, Chengdu, China
| | - Xiaojian Liu
- Department of Cardiology, Sichuan Tianfu New District People’s Hospital, Chengdu, China
| | - Qiyong Li
- Department of Cardiology, Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
38
|
Ji J, Wei X, Wan D, Wu L, Chen G, Liu H. Predictive Value of Plasma PCSK9 Levels for Degree of Atherosclerosis and Major Adverse Cardiovascular and Cerebrovascular Events in Older Adult Patients with Non-Alcoholic Fatty Liver Disease. Int J Gen Med 2024; 17:2177-2186. [PMID: 38770364 PMCID: PMC11104394 DOI: 10.2147/ijgm.s454633] [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: 01/10/2024] [Accepted: 04/15/2024] [Indexed: 05/22/2024] Open
Abstract
Purpose This study investigated the influence of plasma proprotein convertase subtilisin/kexin 9 (PCSK9) levels on the degree of atherosclerosis and major adverse cardiovascular and cerebrovascular events (MACCE) in older adults with non-alcoholic fatty liver disease. Methods The degree of atherosclerosis severity was assessed by the standard Gensini score quartile method. According to the degree of atherosclerosis, patients were divided into mild (0-24 points; n=84), moderate (25-53 points; n=86), and severe groups (≥54 points; n=84) and then categorized as MACCE (n=30) or non-MACCE (n=224) according to 6-month follow-up data. The patients' age, sex, smoking history, medical history, and early morning fasting venous blood, for measuring biochemical indexes, were collected. Clinical data were compared between groups and the relationship between Gensini scores and PCSK9 was evaluated. Results Compared with the mild group, the moderate and severe groups had higher high-sensitivity C-reactive protein(hs-CRP), PCSK9, triglycerides(TG), low-density lipoprotein cholesterol (LDL-C), and lipoprotein(a)[Lp(a)] levels and lower high-density lipoprotein cholesterol(HDL-C) levels (all P<0.05). Moreover, PCSK9 positively correlated with Gensini scores (r=0.657, P<0.01). The MACCE and non-MACCE groups had significantly different ages, statin use, Gensini scores, PCSK9, and LDL-C (all P<0.05). Multi-factorial Cox risk regression analysis showed the Gensini score (HR=1.018, 95% CI: 1.006~1.029) and PCSK9 (HR=1.147, 95% CI: 1.038~1.287) were independent risk factors for MACCE. Conclusion The Gensini score and PCSK9 levels can be used as predictive indicators for the degree of illness and occurrence of MACCE in older NAFLD patients.
Collapse
Affiliation(s)
- Jinrui Ji
- Clinical Medical Department, Faculty of Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
- Department of Cardiology, People’s Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Xiaoyun Wei
- Clinical Medical Department, Faculty of Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
- Department of Cardiology, People’s Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Dongyun Wan
- Clinical Medical Department, Faculty of Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
- Department of Cardiology, People’s Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Lei Wu
- Clinical Medical Department, Faculty of Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
- Department of Cardiology, People’s Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Guangyao Chen
- Clinical Medical Department, Faculty of Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
- Zhengzhou Institute of Gastroenterology, People’s Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Hengliang Liu
- Clinical Medical Department, Faculty of Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
- Department of Cardiology, People’s Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| |
Collapse
|
39
|
Wang P, Liu XL, Jiang ZZ, Long Y, Gao CL, Huang W, Tan XZ, Ma XM, Xu Y. Effect of proanthocyanidins on blood lipids: A systematic review and meta-analysis. Phytother Res 2024; 38:2154-2164. [PMID: 38391003 DOI: 10.1002/ptr.8162] [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: 03/17/2023] [Revised: 12/07/2023] [Accepted: 02/04/2024] [Indexed: 02/24/2024]
Abstract
Proanthocyanidins (PCs) are natural antioxidant polyphenols and their effect on the regulation of blood lipids is still controversial. This study was conducted to evaluate the effect of PCs on lipid metabolism. We searched PubMed, Embase, Web of Science, Chinese biomedical literature service system, China National Knowledge Internet, and Wanfang Data with no time restriction until March 18, 2022, using various forms of "proanthocyanidins" and "blood lipid" search terms. Randomized controlled trials investigating the relationship between PCs and lipid metabolism were included. The standard system of Cochrane Collaboration was used to assess the quality of studies. We standardized mean differences (SMDs) with 95% confidence interval (CI) using the random-effects model, Cohen approach. Seventeen studies (17 trials, N = 1138) fulfilled the eligibility criteria. PCs significantly reduced triglyceride, and increased recombinant apolipoprotein A1. Subgroup analysis showed a significant reduction in triglycerides in older adults (≥60 years) and total cholesterol for participants who were not overweight or obese (body mass index <24). An intervention duration of greater than 8 weeks reduced triglyceride and low-density lipoprotein cholesterol levels but increased high-density lipoprotein cholesterol. Different doses of PCs could regulate triglycerides, high-density lipoprotein cholesterol and total cholesterol. PCs have beneficial effects on circulating lipids and may represent a new approach for treating or preventing lipid metabolism disorders. However, more high-quality studies are needed to confirm these results.
Collapse
Affiliation(s)
- Peng Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, PR China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, PR China
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, PR China
| | - Xue Lian Liu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, PR China
- Sichuan College of Traditional Chinese Medicine, Mianyang, PR China
| | - Zong Zhe Jiang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, PR China
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, PR China
| | - Chen Lin Gao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, PR China
| | - Wei Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, PR China
| | - Xiao Zhen Tan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, PR China
| | - Xiu Mei Ma
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, PR China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, PR China
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, PR China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, PR China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, PR China
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, PR China
| |
Collapse
|
40
|
Luo F, Das A, Khetarpal SA, Fang Z, Zelniker TA, Rosenson RS, Qamar A. ANGPTL3 inhibition, dyslipidemia, and cardiovascular diseases. Trends Cardiovasc Med 2024; 34:215-222. [PMID: 36746257 DOI: 10.1016/j.tcm.2023.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/07/2023]
Abstract
Optimal management of low-density lipoprotein cholesterol (LDL-C) is a central tenet in the primary and secondary prevention of atherosclerotic cardiovascular disease (ASCVD). However, significant residual cardiovascular risk remains despite achieving guideline-directed LDL-C levels, in part due to mixed hyperlipidemia with elevated fasting and non-fasting triglyceride-rich lipoprotein levels. Advances in human genetics have identified angiopoietin-like 3 (ANGPTL3) as a promising therapeutic target to lower cardiovascular risk. Evidence accrued from genetic epidemiological studies demonstrate that ANGPTL3 loss of function is strongly associated with lowering of circulating LDL-C, triglyceride-rich lipoproteins and concurrent risk reduction in development of coronary artery disease. Pharmacological inhibition of ANGPTL3 with monoclonal antibodies, antisense oligonucleotides and gene editing are in development with early studies showing their safety and efficacy in lowering in both, LDL-C and TGs, circumventing a key limitation of previous therapies. Monoclonal antibodies targeting ANGPTL3 are approved for clinical use in homozygous familial hypercholesteremia in USA and Europe. Although promising, future studies focusing on long-term beneficial effect in reducing cardiovascular events with inhibition of ANGPTL3 are warranted.
Collapse
Affiliation(s)
- Fei Luo
- Department of Cardiovascular Medicine, Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Avash Das
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Sumeet A Khetarpal
- Division of Cardiology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Zhenfei Fang
- Department of Cardiovascular Medicine, Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Thomas A Zelniker
- Division of Cardiology, Vienna General Hospital and Medical University of Vienna, Austria
| | - Robert S Rosenson
- Metabolism and Lipids Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R Kravis Center for Cardiovascular Health, Mount Sinai Icahn School of Medicine, New York, NY, United States
| | - Arman Qamar
- Section of Interventional Cardiology & Vascular Medicine, NorthShore University Health System, University of Chicago Pritzker School of Medicine, 2650 Ridge Avenue, Evanston, IL, United States.
| |
Collapse
|
41
|
Nicholls SJ, Nelson AJ. New targets and mechanisms of action for lipid-lowering and anti-inflammatory therapies in atherosclerosis: where does the field stand? Expert Opin Ther Targets 2024; 28:375-384. [PMID: 38815057 DOI: 10.1080/14728222.2024.2362644] [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: 12/20/2023] [Accepted: 05/29/2024] [Indexed: 06/01/2024]
Abstract
INTRODUCTION Atherosclerotic cardiovascular disease remains a leading cause of morbidity and mortality worldwide, despite widespread use of statins. There is a need to develop additional therapeutic strategies that will complement statins to achieve more effective reductions in cardiovascular risk. AREAS COVERED This review provides a comprehensive summary of current areas of therapeutic development targeting both lipid and inflammatory factors implicated in the pathogenesis of atherosclerosis. In addition to develop of novel approaches that will produce more effective lowering of low-density lipoprotein cholesterol, clinical trials are currently evaluating the potential to target other atherogenic lipid parameters such as triglyceride-rich lipoproteins and Lp(a), in addition to promoting the biological properties of high-density lipoproteins. Targeting inflammation within the vascular wall has emerged as a new frontier in cardiovascular prevention, with early evidence that use of anti-inflammatory agents have the potential to reduce cardiovascular risk. EXPERT OPINION Clinical practice has an increasing array of therapeutic tools to achieve more effective lowering of low-density lipoprotein cholesterol for high-risk patients. In addition, clinical trials have the potential to deliver a range of additional agents to the clinic, that target alternative lipid and inflammatory mediators. This will permit the potential to personalize cardiovascular prevention.
Collapse
Affiliation(s)
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, Melbourne, Australia
| |
Collapse
|
42
|
Jurin I, Jurišić A, Rudež I, Kurtić E, Skorić I, Čikara T, Šipić T, Rudan D, Manola Š, Hadžibegović I. Outcomes of Patients with Normal LDL-Cholesterol at Admission for Acute Coronary Syndromes: Lower Is Not Always Better. J Cardiovasc Dev Dis 2024; 11:120. [PMID: 38667738 PMCID: PMC11050419 DOI: 10.3390/jcdd11040120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND AND AIM There are few prospective data on the prognostic value of normal admission low-density lipoprotein cholesterol (LDL-C) in statin-naïve patients with acute coronary syndromes (ACS) who are treated with a preemptive invasive strategy. We aimed to analyze the proportion of patients with normal LDL-C at admission for ACS in our practice, and their characteristics and clinical outcomes in comparison to patients with high admission LDL-C. PATIENTS AND METHODS Two institutions' prospective registries of patients with confirmed ACS from Jan 2017 to Jan 2023 were used to identify 1579 statin-naïve patients with no history of prior coronary artery disease (CAD), and with available LDL-C admission results, relevant clinical and procedural data, and short- and long-term follow-up data. Normal LDL-C at admission was defined as lower than 2.6 mmol/L. All demographic, clinical, procedural, and follow-up data were compared between patients with normal LDL-C and patients with a high LDL-C level (≥2.6 mmol/L) at admission. RESULTS There were 242 (15%) patients with normal LDL-C at admission. In comparison to patients with high LDL-cholesterol at admission, they were significantly older (median 67 vs. 62 years) with worse renal function, had significantly more cases of diabetes mellitus (DM) (26% vs. 17%), peripheral artery disease (PAD) (14% vs. 9%), chronic obstructive pulmonary disease (COPD) (8% vs. 2%), and psychological disorders requiring medical attention (19% vs. 10%). There were no significant differences in clinical type of ACS. Complexity of CAD estimated by coronary angiography was similar between the two groups (median Syntax score 12 for both groups). There were no significant differences in rates of complete revascularization (67% vs. 72%). Patients with normal LDL-C had significantly lower left ventricular ejection fraction (LVEF) at discharge (median LVEF 52% vs. 55%). Patients with normal LDL-C at admission had both significantly higher in-hospital mortality (5% vs. 2%, RR 2.07, 95% CI 1.08-3.96) and overall mortality during a median follow-up of 43 months (27% vs. 14%, RR 1.86, 95% CI 1.45-2.37). After adjusting for age, renal function, presence of diabetes mellitus, PAD, COPD, psychological disorders, BMI, and LVEF at discharge in a multivariate Cox regression analysis, normal LDL-C at admission remained significantly and independently associated with higher long-term mortality during follow-up (RR 1.48, 95% CI 1.05-2.09). CONCLUSIONS A spontaneously normal LDL-C level at admission for ACS in statin-naïve patients was not rare and it was an independent risk factor for both substantially higher in-hospital mortality and mortality during long-term follow-up. Patients with normal LDL-C and otherwise high total cardiovascular risk scores should be detected early and treated with optimal medical therapy. However, additional research is needed to reveal all the missing pieces in their survival puzzle after ACS-beyond coronary anatomy, PCI optimization, numerical LDL-C levels, and statin therapy.
Collapse
Affiliation(s)
- Ivana Jurin
- Department for Cardiovascular Diseases, Dubrava University Hospital, 10000 Zagreb, Croatia; (I.J.); (A.J.); (T.Š.); (D.R.); (Š.M.)
| | - Anđela Jurišić
- Department for Cardiovascular Diseases, Dubrava University Hospital, 10000 Zagreb, Croatia; (I.J.); (A.J.); (T.Š.); (D.R.); (Š.M.)
| | - Igor Rudež
- Department of Cardiac and Transplant Surgery, Dubrava University Hospital, 10000 Zagreb, Croatia;
| | - Ena Kurtić
- Division of Cardiology, Department of Medicine, University Hospital Merkur, 10000 Zagreb, Croatia;
| | - Ivan Skorić
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Tomislav Čikara
- Department for Cardiovascular Diseases, Dubrava University Hospital, 10000 Zagreb, Croatia; (I.J.); (A.J.); (T.Š.); (D.R.); (Š.M.)
| | - Tomislav Šipić
- Department for Cardiovascular Diseases, Dubrava University Hospital, 10000 Zagreb, Croatia; (I.J.); (A.J.); (T.Š.); (D.R.); (Š.M.)
| | - Diana Rudan
- Department for Cardiovascular Diseases, Dubrava University Hospital, 10000 Zagreb, Croatia; (I.J.); (A.J.); (T.Š.); (D.R.); (Š.M.)
- University North, Trg dr. Zarka Dolinara 1, 48000 Koprivnica, Croatia
| | - Šime Manola
- Department for Cardiovascular Diseases, Dubrava University Hospital, 10000 Zagreb, Croatia; (I.J.); (A.J.); (T.Š.); (D.R.); (Š.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Irzal Hadžibegović
- Department for Cardiovascular Diseases, Dubrava University Hospital, 10000 Zagreb, Croatia; (I.J.); (A.J.); (T.Š.); (D.R.); (Š.M.)
- Faculty of Dental Medicine and Health Care, Josip Juraj Strossmayer University, 31000 Osijek, Croatia
| |
Collapse
|
43
|
Sato R, Matsuzawa Y, Yoshii T, Akiyama E, Konishi M, Nakahashi H, Minamimoto Y, Kimura Y, Okada K, Maejima N, Iwahashi N, Kosuge M, Ebina T, Kimura K, Tamura K, Hibi K. Impact of Low-Density Lipoprotein Cholesterol Levels at Acute Coronary Syndrome Admission on Long-Term Clinical Outcomes. J Atheroscler Thromb 2024; 31:444-460. [PMID: 37821363 PMCID: PMC10999725 DOI: 10.5551/jat.64368] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/21/2023] [Indexed: 10/13/2023] Open
Abstract
AIM Low-density lipoprotein cholesterol (LDL-C) level reduction is highly effective in preventing the occurrence of a cardiovascular event. Contrariwise, an inverse association exists between LDL-C levels and prognosis in some patients with cardiovascular diseases-the so-called "cholesterol paradox." This study aimed to investigate whether the LDL-C level on admission affects the long-term prognosis in patients who develop acute coronary syndrome (ACS) and to examine factors associated with poor prognosis in patients with low LDL-C levels. METHODS We enrolled 410 statin-naïve patients with ACS, whom we divided into low- and high-LDL-C groups based on an admission LDL-C cut-off (obtained from the Youden index) of 122 mg/dL. Endothelial function was assessed using the reactive hyperemia index 1 week after statin initiation. The primary composite endpoint included all-cause death, as well as myocardial infarction and ischemic stroke occurrences. RESULTS During a median follow-up period of 6.1 years, 76 patients experienced the primary endpoint. Multivariate Cox regression analysis revealed that patients in the low LDL-C group had a 2.3-fold higher risk of experiencing the primary endpoint than those in the high LDL-C group (hazard ratio, 2.34; 95% confidence interval, 1.29-4.27; p=0.005). In the low LDL-C group, slow gait speed (frailty), elevated chronic-phase high-sensitivity C-reactive protein levels (chronic inflammation), and endothelial dysfunction were significantly associated with the primary endpoint. CONCLUSIONS Patients with low LDL-C levels at admission due to ACS had a significantly worse long-term prognosis than those with high LDL-C levels; frailty, chronic inflammation, and endothelial dysfunction were poor prognostic factors.
Collapse
Affiliation(s)
- Ryosuke Sato
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Yasushi Matsuzawa
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
- Department of Cardiovascular Medicine, Kumamoto University Hospital, Kumamoto, Japan
| | - Tomohiro Yoshii
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Eiichi Akiyama
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Masaaki Konishi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Hidefumi Nakahashi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Yugo Minamimoto
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Yuichiro Kimura
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Kozo Okada
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Nobuhiko Maejima
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Noriaki Iwahashi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Masami Kosuge
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Toshiaki Ebina
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Kazuo Kimura
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| |
Collapse
|
44
|
Moustafa B, Oparowski D, Testai S, Guman I, Trifan G. Efficacy and safety of PCSK9 inhibitors for stroke prevention: Systematic review and meta-analysis. J Stroke Cerebrovasc Dis 2024; 33:107633. [PMID: 38336118 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107633] [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: 12/24/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024] Open
Abstract
OBJECTIVE Investigate the efficacy and safety of proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) on stroke prevention. BACKGROUND PCSK9i reduce low-density lipoprotein cholesterol (LDL-C) and lipoprotein a (LpA) levels. Their efficacy in reducing the risk of major cardiovascular events has been shown in multiple randomized clinical trials (RCT). However, clinical equipoise remains on the magnitude and mechanisms by which PCSK9i decrease the risk of stroke. METHODS We performed a systematic search of biomedical databases from inception to January 15, 2024, to identify RCTs that investigated the efficacy of PCSK9i versus placebo for major cardiovascular event prevention. The primary outcome was total stroke. The safety outcome was the risk of adverse neurological events, as defined by each trial. Effect size was represented by risk ratio (RR), and analysis was done using random-effects meta-analysis. Heterogeneity was assessed by I2 and Cochrane Q statistics. Meta-regression analyses were performed to assess the association between LDL-C and LpA reduction and stroke risk. RESULTS Overall, 20 studies with 93,093 patients were included. The quality of the evidence was moderate and heterogeneity for all comparisons was low (I2 < 25 %). The mean age was 60.1 years for the PCSK9i group and 59.6 years for the placebo group, with a mean follow-up time of 60.1 weeks. PCSK9i reduced the LDL-C levels by 11 % and LpA levels by 8 %. PCSK9i were associated with a significant reduction in stroke risk (RR 0.75, 95 % CI 0.66-0.86, I2 = 0 %), without an increase in mortality (RR 0.97, 95 % CI 0.87-1.08, I2 = 0 %). The risk of adverse neurological events was similar between groups (RR 0.99, 95 % CI 0.84-1.18, I2 = 11 %). In meta-regression analyses, the stroke risk was not associated with the magnitude of the effect of PCSK9i on LDL-C (LDL C β = -0.01, 95 % CI = -0.03-0.02) and LpA (β = -0.01, 95 % CI = -0.06-0.04) levels. CONCLUSIONS PCSK9i significantly reduced the stroke risk, without increasing mortality or the risk of adverse neurological events. Our findings also suggest that the beneficial effect of PCSK9i on stroke risk is mediated by LDL-C- and LpA-independent mechanisms.
Collapse
Affiliation(s)
- Bayan Moustafa
- Mayo Clinic Health System-Eau Claire, Eau Claire, WI, United States.
| | | | - Sofia Testai
- Latin School of Chicago, Chicago, IL, United States
| | - Ilan Guman
- Glenbrook North High Sch, Northbrook, IL, United States
| | - Gabriela Trifan
- Department of Neurology and Rehabilitation, University of Illinois Chicago, College of Medicine, Chicago, IL, United States
| |
Collapse
|
45
|
Dankar R, Wehbi J, Refaat MM. Tailoring Treatment in Cardiovascular Diseases: The Role of Targeted Therapies. Pharmaceutics 2024; 16:461. [PMID: 38675122 PMCID: PMC11054164 DOI: 10.3390/pharmaceutics16040461] [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: 02/26/2024] [Revised: 03/19/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality around the globe. To address this public health burden, innovative therapeutic agents are being developed to specifically target molecular and genetic markers. Various therapeutic modalities have been implemented, including vaccines, monoclonal or bispecific antibodies, and gene-based therapies. Such drugs precisely target the underlying disease pathophysiology, aiming at notable molecules such as lipid metabolism regulators, proinflammatory cytokines, and growth factors. This review focuses on the latest advancements in different targeted therapies. It provides an insightful overview of the current landscape of targeted cardiovascular therapies, highlighting promising strategies with potential to transform the treatment of CVDs into an era of precision medicine.
Collapse
Affiliation(s)
- Razan Dankar
- Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon; (R.D.); (J.W.)
- Department of Internal Medicine, Division of Cardiology, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon
| | - Jad Wehbi
- Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon; (R.D.); (J.W.)
- Department of Internal Medicine, Division of Cardiology, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon
| | - Marwan M. Refaat
- Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon; (R.D.); (J.W.)
- Department of Internal Medicine, Division of Cardiology, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon
| |
Collapse
|
46
|
Iqbal S, Sabbour HM, Ashraf T, Santos RD, Buckley A. First Report of Inclisiran Utilization for Hypercholesterolemia Treatment in Real-world Clinical Settings in a Middle East Population. Clin Ther 2024; 46:186-193. [PMID: 38220483 DOI: 10.1016/j.clinthera.2023.12.003] [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: 09/11/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 01/16/2024]
Abstract
PURPOSE Inclisiran is the first small interfering RNA-based treatment approved for reducing pro-atherogenic lipoproteins in patients with heterozygous familial hypercholesterolemia or clinical atherosclerotic cardiovascular disease, who require additional lowering of low-density lipoprotein cholesterol (LDL-C). We report the first evaluation of the effects of inclisiran in a Middle East population. METHODS We conducted a retrospective review of patients initiating inclisiran treatment at an outpatient diabetology, endocrinology, and cardiology center between May 2021 and December 2022. All patients followed up for 90 days or more, or with at least 1 lipid determination post initiation were included. Participants were categorized into primary prevention (n = 57) and secondary prevention (n = 89) groups according to previous atherosclerotic cardiovascular disease. FINDINGS Inclisiran was initiated in 146 patients; mean (SD) age was 54.8 (12.12) years, 82 patients (56.2%) were male, 28 patients (19.2%) had received a diagnosis of familial hypercholesterolemia, 89 patients (61%) had received a diagnosis of diabetes mellitus, and 35 patients (23.9%) had a statin intolerance. Median (interquartile range) follow-up was 137 (90 to 193) days. At 90 days, median (interquartile range) reductions in serum LDL-C and triglycerides were -37.9% (-9.5% to -51.2%) and -12.0% (-9.8% to -40.5%), respectively, in primary prevention and -54.1% (-17.1% to -71.4%) and -15.3% (-14% to -38.8%), respectively, in secondary prevention (all, P < 0.001). LDL-C goals were attained in 110 patients (75.3%). Nonattainment of LDL-C goal was attributed to system effect in 26 patients (72.2%), biological effect in 5 patients (13.9%), and discontinuation of treatment in 5 patients (13.9%). Therapy was well tolerated. IMPLICATIONS This study is the first from the Middle East and North Africa region that reported the real-world efficacy and safety profile of inclisiran in a mixed-risk population of patients with heterozygous familial hypercholesterolemia and other non-familial hypercholesterolemia indications. Clinically meaningful and sustained reductions in pro-atherogenic lipids with good tolerability were observed after inclisiran initiation. Fewer AEs were reported in this predominantly Arabic population, consistent with previous safety reports for inclisiran. It is important to note that no patient stopped inclisiran treatment due to AEs. Strengths of our study included an optimal cohort, patient heterogeneity, and high retention. In addition, we were able to report mean robust effects of inclisiran and good medication tolerability, quite like randomized studies and open-label extension periods. Despite this, our study had some limitations, including selection bias due to the retrospective design and the absence of a comparative group.
Collapse
Affiliation(s)
- Sajid Iqbal
- Imperial College London Diabetes Centre, Abu Dhabi, United Arab Emirates.
| | - Hani Mohamed Sabbour
- Imperial College London Diabetes Centre, Abu Dhabi, United Arab Emirates; Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | - Tanveer Ashraf
- Imperial College London Diabetes Centre, Abu Dhabi, United Arab Emirates
| | - Raul D Santos
- Heart Institute (InCor) University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil; Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Adam Buckley
- Imperial College London Diabetes Centre, Abu Dhabi, United Arab Emirates
| |
Collapse
|
47
|
Luo F, Lin Y, Zhang X, Li Y, Su L, Zhou S, Xu R, Gao Q, Chen R, Guo Z, Nie S, Xu X. Post-treatment level of LDL cholesterol and all-cause mortality in patients with atherosclerotic cardiovascular disease: evidence from real-world setting. Eur J Prev Cardiol 2024; 31:337-345. [PMID: 37966728 DOI: 10.1093/eurjpc/zwad354] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/04/2023] [Accepted: 10/29/2023] [Indexed: 11/16/2023]
Abstract
AIMS This study aimed to evaluate the safety of the currently recommended target of LDL cholesterol (LDL-C) control on mortality in patients with atherosclerotic cardiovascular disease (ASCVD). METHODS AND RESULTS Using deidentified electronic health record data, we conducted a multicentre retrospective cohort study involving individuals with documented ASCVD who had received statin treatment for at least 3 months across China. The primary outcomes assessed encompassed all-cause mortality, CV mortality, and non-CV mortality. Relationships between post-treatment LDL-C concentrations and outcomes were evaluated using restricted cubic spline curves based on Cox proportional hazards regression analyses. Additionally, competitive risk models were employed to explore associations between LDL-C levels and cause-specific mortality. Among 33 968 participants, we identified nearly linear associations of post-treatment LDL-C level with all-cause mortality and CV mortality during a median follow-up of 47 months. Notably, patients who achieved the recommended target of LDL-C (<1.4 mmol/L) were at significantly lower risks of all-cause mortality [hazard ratio (HR), 0.77; 95% confidence interval (CI), 0.69-0.86] and CV mortality (subdistribution HR, 0.68; 95% CI, 0.58-0.79), compared with those with LDL-C ≥ 3.4 mmol/L. This survival benefit was consistent in patients with different intensities of LDL-C reduction and other subgroup analyses. And no correlation was found between post-treatment LDL-C concentration and non-CV mortality. CONCLUSION Our findings supported the safety of currently recommended target of LDL-C control and the 'lower is better' principle in patients with ASCVD.
Collapse
Affiliation(s)
- Fan Luo
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Yuxin Lin
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Xiaodong Zhang
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Yanqin Li
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Licong Su
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Shiyu Zhou
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Ruqi Xu
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Qi Gao
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Ruixuan Chen
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Zhixin Guo
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Sheng Nie
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| | - Xin Xu
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 N Guangzhou Ave, Guangzhou 510515, China
| |
Collapse
|
48
|
Ferrell M, Wang Z, Anderson JT, Li XS, Witkowski M, DiDonato JA, Hilser JR, Hartiala JA, Haghikia A, Cajka T, Fiehn O, Sangwan N, Demuth I, König M, Steinhagen-Thiessen E, Landmesser U, Tang WHW, Allayee H, Hazen SL. A terminal metabolite of niacin promotes vascular inflammation and contributes to cardiovascular disease risk. Nat Med 2024; 30:424-434. [PMID: 38374343 PMCID: PMC11841810 DOI: 10.1038/s41591-023-02793-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 12/22/2023] [Indexed: 02/21/2024]
Abstract
Despite intensive preventive cardiovascular disease (CVD) efforts, substantial residual CVD risk remains even for individuals receiving all guideline-recommended interventions. Niacin is an essential micronutrient fortified in food staples, but its role in CVD is not well understood. In this study, untargeted metabolomics analysis of fasting plasma from stable cardiac patients in a prospective discovery cohort (n = 1,162 total, n = 422 females) suggested that niacin metabolism was associated with incident major adverse cardiovascular events (MACE). Serum levels of the terminal metabolites of excess niacin, N1-methyl-2-pyridone-5-carboxamide (2PY) and N1-methyl-4-pyridone-3-carboxamide (4PY), were associated with increased 3-year MACE risk in two validation cohorts (US n = 2,331 total, n = 774 females; European n = 832 total, n = 249 females) (adjusted hazard ratio (HR) (95% confidence interval) for 2PY: 1.64 (1.10-2.42) and 2.02 (1.29-3.18), respectively; for 4PY: 1.89 (1.26-2.84) and 1.99 (1.26-3.14), respectively). Phenome-wide association analysis of the genetic variant rs10496731, which was significantly associated with both 2PY and 4PY levels, revealed an association of this variant with levels of soluble vascular adhesion molecule 1 (sVCAM-1). Further meta-analysis confirmed association of rs10496731 with sVCAM-1 (n = 106,000 total, n = 53,075 females, P = 3.6 × 10-18). Moreover, sVCAM-1 levels were significantly correlated with both 2PY and 4PY in a validation cohort (n = 974 total, n = 333 females) (2PY: rho = 0.13, P = 7.7 × 10-5; 4PY: rho = 0.18, P = 1.1 × 10-8). Lastly, treatment with physiological levels of 4PY, but not its structural isomer 2PY, induced expression of VCAM-1 and leukocyte adherence to vascular endothelium in mice. Collectively, these results indicate that the terminal breakdown products of excess niacin, 2PY and 4PY, are both associated with residual CVD risk. They also suggest an inflammation-dependent mechanism underlying the clinical association between 4PY and MACE.
Collapse
Affiliation(s)
- Marc Ferrell
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Systems Biology and Bioinformatics Program, Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Zeneng Wang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James T Anderson
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xinmin S Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Marco Witkowski
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Joseph A DiDonato
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James R Hilser
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jaana A Hartiala
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arash Haghikia
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tomas Cajka
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, USA
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, USA
| | - Naseer Sangwan
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ilja Demuth
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Maximilian König
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - W H Wilson Tang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hooman Allayee
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stanley L Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
49
|
Fularski P, Hajdys J, Majchrowicz G, Stabrawa M, Młynarska E, Rysz J, Franczyk B. Unveiling Familial Hypercholesterolemia-Review, Cardiovascular Complications, Lipid-Lowering Treatment and Its Efficacy. Int J Mol Sci 2024; 25:1637. [PMID: 38338916 PMCID: PMC10855128 DOI: 10.3390/ijms25031637] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Familial hypercholesterolemia (FH) is a genetic disorder primarily transmitted in an autosomal-dominant manner. We distinguish two main forms of FH, which differ in the severity of the disease, namely homozygous familial hypercholesterolemia (HoFH) and heterozygous familial hypercholesterolemia (HeFH). The characteristic feature of this disease is a high concentration of low-density lipoprotein cholesterol (LDL-C) in the blood. However, the level may significantly vary between the two mentioned types of FH, and it is decidedly higher in HoFH. A chronically elevated concentration of LDL-C in the plasma leads to the occurrence of certain abnormalities, such as xanthomas in the tendons and skin, as well as corneal arcus. Nevertheless, a significantly more severe phenomenon is leading to the premature onset of cardiovascular disease (CVD) and its clinical implications, such as cardiac events, stroke or vascular dementia, even at a relatively young age. Due to the danger posed by this medical condition, we have investigated how both non-pharmacological and selected pharmacological treatment impact the course of FH, thereby reducing or postponing the risk of clinical manifestations of CVD. The primary objective of this review is to provide a comprehensive summary of the current understanding of FH, the effectiveness of lipid-lowering therapy in FH and to explain the anatomopathological correlation between FH and premature CVD development, with its complications.
Collapse
Affiliation(s)
- Piotr Fularski
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Hajdys
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Gabriela Majchrowicz
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Magdalena Stabrawa
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
50
|
Engin A. Protein Kinases in Obesity, and the Kinase-Targeted Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:199-229. [PMID: 39287853 DOI: 10.1007/978-3-031-63657-8_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The action of protein kinases and protein phosphatases is essential for multiple physiological responses. Each protein kinase displays its own unique substrate specificity and a regulatory mechanism that may be modulated by association with other proteins. Protein kinases are classified as dual-specificity kinases and dual-specificity phosphatases. Dual-specificity phosphatases are important signal transduction enzymes that regulate various cellular processes in coordination with protein kinases and play an important role in obesity. Impairment of insulin signaling in obesity is largely mediated by the activation of the inhibitor of kappa B-kinase beta and the c-Jun N-terminal kinase (JNK). Oxidative stress and endoplasmic reticulum (ER) stress activate the JNK pathway which suppresses insulin biosynthesis. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) are important for proper regulation of glucose metabolism in mammals at both the hormonal and cellular levels. Additionally, obesity-activated calcium/calmodulin dependent-protein kinase II/p38 suppresses insulin-induced protein kinase B phosphorylation by activating the ER stress effector, activating transcription factor-4. To alleviate lipotoxicity and insulin resistance, promising targets are pharmacologically inhibited. Nifedipine, calcium channel blocker, stimulates lipogenesis and adipogenesis by downregulating AMPK and upregulating mTOR, which thereby enhances lipid storage. Contrary to the nifedipine, metformin activates AMPK, increases fatty acid oxidation, suppresses fatty acid synthesis and deposition, and thus alleviates lipotoxicity. Obese adults with vascular endothelial dysfunction have greater endothelial cells activation of unfolded protein response stress sensors, RNA-dependent protein kinase-like ER eukaryotic initiation factor-2 alpha kinase (PERK), and activating transcription factor-6. The transcriptional regulation of adipogenesis in obesity is influenced by AGC (protein kinase A (PKA), PKG, PKC) family signaling kinases. Obesity may induce systemic oxidative stress and increase reactive oxygen species in adipocytes. An increase in intracellular oxidative stress can promote PKC-β activation. Activated PKC-β induces growth factor adapter Shc phosphorylation. Shc-generated peroxides reduce mitochondrial oxygen consumption and enhance triglyceride accumulation and lipotoxicity. Liraglutide attenuates mitochondrial dysfunction and reactive oxygen species generation. Co-treatment of antiobesity and antidiabetic herbal compound, berberine with antipsychotic drug olanzapine decreases the accumulation of triglyceride. While low-dose rapamycin, metformin, amlexanox, thiazolidinediones, and saroglitazar protect against insulin resistance, glucagon-like peptide-1 analog liraglutide inhibits palmitate-induced inflammation by suppressing mTOR complex 1 (mTORC1) activity and protects against lipotoxicity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|