1
|
Avenatti E, DiGregorio H, Hajj EE, Gullapelli R, Williams K, Shahid I, Bose B, Hagan K, Nicolas JC, Lahan S, Nwana N, Butt SA, Monga K, Karam LAR, Guevara M, Javed Z, Weber B, Al-Kindi S, Nasir K. Disparities in statin use in patients with ASCVD with vs without rheumatologic diseases in a large integrated healthcare system: Houston methodist CVD learning health system registry. Am J Prev Cardiol 2025; 22:100959. [PMID: 40290419 PMCID: PMC12023777 DOI: 10.1016/j.ajpc.2025.100959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/28/2025] [Accepted: 03/08/2025] [Indexed: 04/30/2025] Open
Abstract
Objective The comorbid presence of Rheumatologic Diseases (RDs) and Atherosclerotic Cardiovascular Disease (ASCVD) substantially accentuates cardiovascular risk. We aimed to compare rates of secondary prevention statin utilization in patients with established ASCVD both with and without underlying comorbid RDs- and to highlight any potential gender, racial, or ethnic disparities in statin use in a contemporary US cohort. Methods We queried the electronic medical record (EHR)-linked Houston Methodist Learning Health System Outpatient Registry containing data for approximately 1.2 million patients to identify patients with diagnosed ASCVD and RDs using ICD-10 codes. Statin prescription rates and dosage were evaluated via ATC codes. Results Among 113,021 patients with ASCVD, 7286 (6.4 %) had comorbid RDs. The majority (71.1 %) of patients with ASCVD were prescribed statins, with discernibly lower utilization in patients with comorbid RDs compared to the non-RD population (63.2 % vs. 71.7 %, p < 0.005). High-intensity statins were prescribed in 42,636 (37.7 %) of ASCVD patients, with similarly reduced utilization in RD vs non-RD patients (30.4 % vs. 38.2 %). These trends remained consistent across sociodemographic subgroups. Moreover, women were consistently less likely to receive high intensity statins in both RD and non-RD groups. Reduced statin utilization was not accounted for with non-statin lipid lowering therapies in RD vs non RD subgroups. Conclusion In this real-world study, co-morbid RDs were associated with significant lower utilization of secondary prevention statin therapy in patients with ASCVD. A multidisciplinary team approach may help to better understand key drivers of statin uptake in this clinically vulnerable population.
Collapse
Affiliation(s)
- Eleonora Avenatti
- Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Division of Cardiovascular Prevention, Houston Methodist Academic Institute, 6550 Fannin St Suite 1101, Houston, TX 77030, USA
| | - Helene DiGregorio
- Department of Internal Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Elia El Hajj
- Department of Internal Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Rakesh Gullapelli
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| | - Kenneth Williams
- Department of Internal Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Izza Shahid
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| | - Budhaditya Bose
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| | - Kobina Hagan
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| | - Juan C Nicolas
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| | - Shubham Lahan
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| | - Nwabunie Nwana
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| | - Sara Ayaz Butt
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| | - Kanika Monga
- Department of Medicine, Division of Rheumatology, Houston Methodist Hospital, Houston, TX, USA
| | - Lily Anne Romero Karam
- Department of Medicine, Division of Rheumatology, Houston Methodist Hospital, Houston, TX, USA
| | - Myriam Guevara
- Department of Medicine, Division of Rheumatology, Houston Methodist Hospital, Houston, TX, USA
| | - Zulqarnain Javed
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| | - Brittany Weber
- Department of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sadeer Al-Kindi
- Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Division of Cardiovascular Prevention, Houston Methodist Academic Institute, 6550 Fannin St Suite 1101, Houston, TX 77030, USA
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| | - Khurram Nasir
- Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Division of Cardiovascular Prevention, Houston Methodist Academic Institute, 6550 Fannin St Suite 1101, Houston, TX 77030, USA
- Center for Health Data Science and Analytics, Houston Methodist, Houston, TX, United States
| |
Collapse
|
2
|
Li Y, Shi Y, Zhu B, Chen Y, Shen B, Zhao S, Song N, Fang Y, Ding X. Association of chronic kidney disease and cardiovascular disease risk with all-cause mortality: an interaction, joint and mediation analysis in Chinese adults. BMC Public Health 2025; 25:1685. [PMID: 40335982 PMCID: PMC12057200 DOI: 10.1186/s12889-025-22924-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 04/24/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a global public health problem. This study aimed to evaluate the complex relationship of CKD and cardiovascular disease (CVD) risk with mortality in different age groups and the mediation effect of CVD risk among Chinese adults. METHODS A total of 7533 participants from the 2009 wave of China Health and Nutrition Survey (CHNS) cohort were included in this study and followed up to 2015. CKD was defined as the estimated glomerular filtration rate (eGFR) < 60 mL/min/1.73m2. Framingham risk score (FRS) was used to assess CVD risk. The interaction, joint association of CVD risk and CKD on mortality, and subsequent mediation effect were evaluated using multivariable Cox regression. RESULTS CHNS cohort recorded 266 deaths over a mean follow-up time of 5.04 years. The all-mortality rates among adults with CKD and high CVD risk were significantly higher than healthy controls (22.48 and 21.30 per 1000 person-years). After adjusting for covariates of age, gender, BMI, hypertension, diabetes, hyperuricemia, smoking status, and alcohol consumption, the adjusted hazard ratios (aHR) of CKD and high CVD risk were 1.70 (95% CI 1.27-2.28) and 1.62 (95%CI 1.26-2.09), respectively. Joint effect analysis revealed that mortality hazard was highest in CKD patients with high CVD risk (aHR = 3.15, 95% CI 1.92-5.16). Mediation analysis showed that significant partial mediation by SBP and fasting glucose, accounting for 19.2% (p < 0.001) and 3.52% (p = 0.012) of the total effect of CKD on mortality. CONCLUSIONS Comprehensive strategies including lifestyle modifications, diet restrictions, and cardio-nephrology multidisciplinary treatment for mitigating CVD risk in CKD patients should focus on middle-aged people and early disease detection.
Collapse
Affiliation(s)
- Yang Li
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Kidney Disease, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Yiqin Shi
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Kidney Disease, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Bowen Zhu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Kidney Disease, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Yafei Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Kidney Disease, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Bo Shen
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Kidney Disease, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Shuan Zhao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Kidney Disease, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Nana Song
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Kidney Disease, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Clinical Research Center for Kidney Disease, Shanghai, China.
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Clinical Research Center for Kidney Disease, Shanghai, China.
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.
| |
Collapse
|
3
|
Miida T, Hirayama S, Fukushima Y, Hori A, Ito S, Hinata M, Wakita M, Tabata H, Tamura Y, Watada H, Kawamori R, Vesper HW, Cobbaert CM. Harmonization of Lipoprotein(a) Immunoassays Using A Serum Panel Value Assigned with The IFCC-Endorsed Mass Spectrometry-Based Reference Measurement Procedure as A First Step Towards Apolipoprotein Standardization. J Atheroscler Thromb 2025; 32:580-595. [PMID: 39505497 PMCID: PMC12055511 DOI: 10.5551/jat.65238] [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: 07/04/2024] [Accepted: 09/23/2024] [Indexed: 11/08/2024] Open
Abstract
AIM Lipoprotein (a) [Lp(a)] is a well-established risk factor for cardiovascular disease independent of low-density lipoprotein-cholesterol (LDL-C). The Lp(a) concentrations were inconsistent between the immunoassays. This study aimed to investigate whether harmonization of Lp(a) measurements can be achieved using a serum panel value assigned with the IFCC-endorsed mass spectrometry-based reference measurement procedure (IFCC-MS-RMP). METHODS We measured the Lp(a) concentrations using five Lp(a) immunoassays in 40 panel sera provided by the Centers for Disease Control and Prevention (CDC), and 500 Japanese subjects enrolled in the Bunkyo Health Study. Of the five immunoassays, only the Roche Lp(a) assay was traceable to the WHO-IFCC reference material SRM2B. Lp(a) concentrations in CDC samples were also determined by IFCC-MS-RMP, provisionally calibrated to SRM2B. Lp(a) concentrations were expressed in mass units (mg/dL) for most reagents, but in SI units (nmol/L) for Roche's reagent and IFCC-MS-RMP. RESULTS In the CDC panel sera, all immunoassays, including Roche's reagent, showed good correlations with IFCC-MS-RMP. In the Bunkyo Health Study samples, all immunoassays showed good correlations with Roche's reagent (rs, 0.986-0.998) although the slopes of the regression lines ranged from 0.292 to 0.579. After recalibration with the CDC's panel sera, Lp(a) results of Bunkyo Health Study samples were converted to the equivalent values determined by the IFCC-MS-RMP, thus resulting in a marked reduction in the intermethod CV among the assays. CONCLUSION We achieved harmonization of Lp(a) measurements with five immunoassays using a serum panel value assigned with the IFCC-MS-RMP.
Collapse
Affiliation(s)
- Takashi Miida
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, Chiba, Japan
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Satoshi Hirayama
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Health Care Center, Tokyo Gakugei University, Tokyo, Japan
| | - Yoshifumi Fukushima
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Hori
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, Chiba, Japan
| | - Satomi Ito
- Clinical Laboratory, Juntendo University Hospital, Tokyo, Japan
| | - Masanobu Hinata
- Clinical Laboratory, Juntendo University Hospital, Tokyo, Japan
| | - Mitsuru Wakita
- Clinical Laboratory, Juntendo University Hospital, Tokyo, Japan
| | - Hiroki Tabata
- Juntendo Advanced Research Institute for Health Science, Tokyo, Japan
| | - Yoshifumi Tamura
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Sports Medicine and Sportology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryuzo Kawamori
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hubert W Vesper
- Protein Biomarker and Lipid Reference Laboratory, Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, GA, U.S.A
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
4
|
Kotani K. The Importance of the Harmonization and Standardization of Lipoprotein(a) Measurements. J Atheroscler Thromb 2025; 32:563-564. [PMID: 39971286 PMCID: PMC12055508 DOI: 10.5551/jat.ed279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 02/21/2025] Open
Affiliation(s)
- Kazuhiko Kotani
- Division of Community and Family Medicine, Center for Community Medicine, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
5
|
Lin S, Hsu YJ, Kim JS, Jackson JW, Segal JB. Predictive Factors of Apparent Treatment Resistant Hypertension Among Patients With Hypertension Identified Using Electronic Health Records. J Gen Intern Med 2025; 40:1265-1274. [PMID: 39358502 PMCID: PMC12045898 DOI: 10.1007/s11606-024-09068-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Early identification of a patient with resistant hypertension (RH) enables quickly intensified treatment, short-interval follow-up, or perhaps case management to bring his or her blood pressure under control and reduce the risk of complications. OBJECTIVE To identify predictors of RH among individuals with newly diagnosed hypertension (HTN), while comparing different prediction models and techniques for managing missing covariates using electronic health records data. DESIGN Risk prediction study in a retrospective cohort. PARTICIPANTS Adult patients with incident HTN treated in any of the primary care clinics of one health system between April 2013 and December 2016. MAIN MEASURES Predicted risk of RH at the time of HTN identification and candidate predictors for variable selection in future model development. KEY RESULTS Among 26,953 individuals with incident HTN, 613 (2.3%) met criteria for RH after 4.7 months (interquartile range, 1.2-11.3). Variables selected by the least absolute shrinkage and selection operator (LASSO), included baseline systolic blood pressure (SBP) and its missing indicator (a dummy variable created if baseline SBP is absent), use of antihypertensive medication at the time of cohort entry, body mass index, and atherosclerosis risk. The random forest technique achieved the highest area under the curve (AUC) of 0.893 (95% CI, 0.881-0.904) and the best calibration with a calibration slope of 1.01. Complete case analysis is not a valuable option (AUC = 0.625). CONCLUSIONS Machine learning techniques and traditional logistic regression exhibited comparable levels of predictive performance after handling the missingness. We suggest that the variables identified by this study may be good candidates for clinical prediction models to alert clinicians to the need for short-interval follow up and more intensive early therapy for HTN.
Collapse
Affiliation(s)
- Shanshan Lin
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yea-Jen Hsu
- Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ji Soo Kim
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John W Jackson
- Center for Drug Safety and Effectiveness, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jodi B Segal
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Center for Drug Safety and Effectiveness, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Division of General Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
6
|
Yang X, Wu W, Huang W, Fang J, Chen Y, Chen X, Lin X, He Y. Exosomes derived from baicalin‑pretreated mesenchymal stem cells mitigate atherosclerosis by regulating the SIRT1/NF‑κB signaling pathway. Mol Med Rep 2025; 31:126. [PMID: 40084693 PMCID: PMC11924171 DOI: 10.3892/mmr.2025.13491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/13/2025] [Indexed: 03/16/2025] Open
Abstract
Atherosclerosis (AS) is a disease with high global incidence and mortality rates. Currently, the treatment of AS in clinical practice carries a high risk of adverse effects and toxic side effects. The pretreatment of mesenchymal stem cells (MSCs) with drugs may enhance the bioactivity of MSC‑derived exosomes (MSC‑exos), which could be a promising candidate for inhibiting the progression of AS. The aim of the present study was to investigate the ability of exos derived from baicalin‑preconditioned MSCs (Ba‑exos) to exhibit an inhibitory effect on AS progression and to explore the potential molecular mechanisms. Exos were isolated from untreated MSCs and MSCs pretreated with Ba, and were characterized using transmission electron microscopy, nanoparticle tracking analysis and western blotting. Subsequently, Cell Counting Kit‑8 and Transwell assays, reverse transcription‑quantitative PCR, immunofluorescence, western blotting and ELISA were used to evaluate the effects of Ba‑exos on AS, and the possible molecular mechanisms. Oil Red O and Masson staining were used to assess AS pathological tissue in a high‑fat diet‑induced mouse model of AS. Notably, MSC‑exos and Ba‑exos were successfully isolated. Compared with MSC‑exos, Ba‑exos demonstrated superior inhibitory effects on the viability and migration, and the levels of inflammatory factors in oxidized low‑density lipoprotein (ox‑LDL)‑induced vascular smooth muscle cells (VSMCs). Additionally, compared with MSC‑exos, Ba‑exos significantly inhibited NF‑κB activation by upregulating sirtuin 1 (SIRT1), thereby suppressing inflammation in ox‑LDL‑induced VSMCs to a greater extent. In mice with high‑fat diet‑induced AS, Ba‑exos exhibited the ability to inhibit AS plaque formation and to alleviate AS progression by reducing the levels of inflammatory factors compared with MSC‑exos; however, the difference was not significant. In conclusion, Ba‑exos may serve as a potential strategy for treating AS by regulating the SIRT1/NF‑κB signaling pathway to suppress inflammation.
Collapse
Affiliation(s)
- Xiaochun Yang
- The First Clinical College of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Wei Wu
- The First Clinical College of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Weitian Huang
- Department of Rehabilitation, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong 510000, P.R. China
| | - Junfeng Fang
- Department of Emergency, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, P.R. China
| | - Yunli Chen
- Department of Rehabilitation, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong 510000, P.R. China
| | - Xiaoyan Chen
- Department of Rehabilitation, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong 510000, P.R. China
| | - Xiaolan Lin
- Department of Rehabilitation, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong 510000, P.R. China
| | - Yanbin He
- Department of Rehabilitation, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
7
|
Lee S, Kim B, Lee S, Kang SH, Yu KS. Comparative pharmacokinetics and bioequivalence of 145-mg fenofibrate formulations in healthy Korean participants. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04086-y. [PMID: 40304748 DOI: 10.1007/s00210-025-04086-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/21/2025] [Indexed: 05/02/2025]
Abstract
Fenofibrate is a serum lipid-modifying agent that is commonly used to treat dyslipidemia. This study aimed to compare the pharmacokinetics (PKs) and establish bioequivalence of two 145-mg fenofibrate formulations, AD-104 (test) and TRICOR® (reference). This randomized, open-label, two-sequence, two-period crossover study was conducted in healthy Korean participants. Forty participants were enrolled and received either the test or reference formulation during each period, with a 14-day washout between doses. Blood samples were collected pre-dose and up to 72 h post-dose. PK parameters were assessed using non-compartmental analysis with Phoenix WinNonlin®. Bioequivalence was determined if the 90% confidence intervals (CIs) for the geometric mean ratios (GMRs) of the maximum plasma concentration (Cmax) and the area under the concentration-time curve from time zero to the last measurable plasma concentration (AUClast) were within the bioequivalence limits of 0.80 to 1.25. Thirty-eight participants completed the study and were included in the PK analysis. The GMR and 90% CIs for the Cmax and AUClast of the test compared to the reference formulation were 0.8643 (0.8283-0.9019) and 0.9930 (0.9631-1.0239), respectively, both within the bioequivalence limits. No serious adverse events were reported during the study. This study demonstrates that the test formulation is bioequivalent to the reference formulation in healthy Korean participants. Both formulations were safe and well-tolerated; therefore, AD-104 is expected to benefit Korean patients with dyslipidemia. Clinical Research Information Service No. is as follows: KCT0009332 (April 12, 2024).
Collapse
Affiliation(s)
- Sujong Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Byungwook Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Seung-Hyun Kang
- Clinical Research Center, H-Plus Yangji Hospital, Seoul, Republic of Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Chen G, Chen Y, Yao Y, Ding L, Wu S, Wu W. High Cumulative Non-High-Density Lipoprotein-Cholesterol Concentration Increases the Risk of New-Onset Arterial Stiffness - A Prospective Cohort Study. Circ J 2025; 89:629-637. [PMID: 40090733 DOI: 10.1253/circj.cj-24-0921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
BACKGROUND The relationship between cumulative non-high-density lipoprotein-cholesterol (cum-non-HDL-C) and the risk of new-onset arterial stiffness has not been characterized. METHODS AND RESULTS A total of 6,852 participants with 3 consecutive measurements of total cholesterol and HDL-C and a baseline brachial-ankle pulse wave velocity (baPWV) <1,400 cm/s during 2010-2011, 2012-2013, and 2014-2015 were included. The cum-non-HDL-C concentrations were determined using time weighting, and the participants were grouped: G1 <130 mg/dL, G2 130-159 mg/dL, G3 160-189 mg/dL, and G4 ≥190 mg/dL. Cox models were used to characterize the relationships between cum-non-HDL-C and arterial stiffness by calculating hazard ratios (HRs) and 95% confidence intervals (CIs). Arterial stiffness (baPWV ≥1,800 cm/s) was present in 327 (4.77%) participants over a median follow-up period of 7.7 (interquartile range 7.2-8.2) years. After adjustment for multiple confounders, G2-4 had adjusted HRs (95% CIs) of 1.12 (0.85, 1.48), 1.45 (1.05, 1.99), and 2.52 (1.69, 3.74), respectively (P=0.0004), vs. G1. The adjusted HRs (95% CIs) for exposures of 2, 4, and 6 years were 1.17 (0.87, 1.58), 1.46 (1.96, 2.01), and 1.67 (1.14, 2.44), respectively (P=0.0029), vs. 0 years. Restricted cubic spline analysis revealed a linear dose-response relationship between cum-non-HDL-C and arterial stiffness risk. CONCLUSIONS A high cum-non-HDL-C concentration and prolonged exposure to this increase the risk of arterial stiffness. The monitoring and maintenance of appropriate cum-non-HDL-C may reduce the risk of arterial stiffness.
Collapse
Affiliation(s)
- Guanzhi Chen
- Peking Union Medical College, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Fuwai Hospital
| | - Yanjuan Chen
- Department of Endocrinology, Second Affiliated Hospital of Shantou University Medical College
| | - Yan Yao
- Peking Union Medical College, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Fuwai Hospital
| | - Ligang Ding
- Peking Union Medical College, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Fuwai Hospital
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital
| | - Weiqiang Wu
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College
| |
Collapse
|
9
|
Tsukada YT, Aoki-Kamiya C, Mizuno A, Nakayama A, Ide T, Aoyama R, Honye J, Hoshina K, Ikegame T, Inoue K, Bando YK, Kataoka M, Kondo N, Maemura K, Makaya M, Masumori N, Mito A, Miyauchi M, Miyazaki A, Nakano Y, Nakao YM, Nakatsuka M, Nakayama T, Oginosawa Y, Ohba N, Otsuka M, Okaniwa H, Saito A, Saito K, Sakata Y, Harada-Shiba M, Soejima K, Takahashi S, Takahashi T, Tanaka T, Wada Y, Watanabe Y, Yano Y, Yoshida M, Yoshikawa T, Yoshimatsu J, Abe T, Dai Z, Endo A, Fukuda-Doi M, Ito-Hagiwara K, Harima A, Hirakawa K, Hosokawa K, Iizuka G, Ikeda S, Ishii N, Izawa KP, Kagiyama N, Umeda-Kameyama Y, Kanki S, Kato K, Komuro A, Konagai N, Konishi Y, Nishizaki F, Noma S, Norimatsu T, Numao Y, Oishi S, Okubo K, Ohmori T, Otaki Y, Shibata T, Shibuya J, Shimbo M, Shiomura R, Sugiyama K, Suzuki T, Tajima E, Tsukihashi A, Yasui H, Amano K, Kohsaka S, Minamino T, Nagai R, Setoguchi S, Terada K, Yumino D, Tomoike H. JCS/JCC/JACR/JATS 2024 Guideline on Cardiovascular Practice With Consideration for Diversity, Equity, and Inclusion. Circ J 2025; 89:658-739. [PMID: 39971310 DOI: 10.1253/circj.cj-23-0890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Affiliation(s)
| | - Chizuko Aoki-Kamiya
- Department of Obstetrics and Gynecology, National Cerebral and Cardiovascular Center
| | - Atsushi Mizuno
- Department of Cardiology, St. Luke's International Hospital
| | | | - Tomomi Ide
- Department of Cardiovascular Medicine, Kyushu University
| | - Rie Aoyama
- Department of Cardiology, Heart and Vascular Institute, Funabashi Municipal Medical Center
| | - Junko Honye
- Cardiovascular Center, Kikuna Memorial Hospital
| | | | | | - Koki Inoue
- Department of Neuropsychiatry, Graduate School of Medicine, Osaka Metropolitan University
| | - Yasuko K Bando
- Department of Molecular Physiology and Cardiovascular Biology, Mie University Graduate School of Medicine
| | - Masaharu Kataoka
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Japan
| | - Naoki Kondo
- Department of Social Epidemiology, Graduate School of Medicine and School of Public Health, Kyoto University
| | - Koji Maemura
- Department of Cardiovascular Medicine, Nagasaki University Graduate School of Biomedical Sciences
| | | | - Naoya Masumori
- Department of Urology, Sapporo Medical University School of Medicine
| | - Asako Mito
- Division of Maternal Medicine, Center for Maternal-Fetal-Reproductive Medicine, National Center for Child Health and Development
| | - Mizuho Miyauchi
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Aya Miyazaki
- Department of Pediatric Cardiology, Department of Adult Congenital Heart Disease, Seirei Hamamatsu General Hospital
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences
| | - Yoko M Nakao
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University
| | - Mikiya Nakatsuka
- Faculty of Health Sciences, Okayama University Graduate School of Medicine
| | - Takeo Nakayama
- Department of Health Informatics, School of Public Health, Kyoto University
| | - Yasushi Oginosawa
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, Japan
| | | | - Maki Otsuka
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine
| | - Hiroki Okaniwa
- Department of Technology, Gunma Prefectural Cardiovascular Center
| | - Aya Saito
- Department of Surgery, Division of Cardiovascular Surgery, Yokohama City University, Graduate School of Medicine
| | - Kozue Saito
- Department of Neurology, Stroke Center, Nara Medical University
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | | | - Kyoko Soejima
- Department of Cardiovascular Medicine, Kyorin University School of Medicine
| | | | - Tetsuya Takahashi
- Department of Physical Therapy, Faculty of Health Science, Juntendo University
| | - Toshihiro Tanaka
- Department of Human Genetics and Disease Diversity, Tokyo Medical and Dental University
| | - Yuko Wada
- Division of Cardiovascular Surgery, Department of Surgery, Shinshu University School of Medicine
| | | | - Yuichiro Yano
- Department of General Medicine, Juntendo University Faculty of Medicine
| | - Masayuki Yoshida
- Department of Life Sciences and Bioethics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU)
| | - Toru Yoshikawa
- Research Center for Overwork-Related Disorders (RECORDs), National Institute of Occuatopnal Safety and Health, Japan (JNIOSH)
| | - Jun Yoshimatsu
- Department of Obstetrics and Gynecology, National Cerebral and Cardiovascular Center
| | - Takahiro Abe
- Department of Rehabilitation Medicine, Hokkaido University Hospital
| | - Zhehao Dai
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Ayaka Endo
- Department of Cardiology, Tokyo Saiseikai Central Hospital
| | - Mayumi Fukuda-Doi
- Department of Data Science, National Cerebral and Cardiovascular Center
- Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center
| | | | | | - Kyoko Hirakawa
- Department of Cardiovascular Medicine, Kumamoto University
| | | | | | - Satoshi Ikeda
- Stroke and Cardiovascular Diseases Support Center, Nagasaki University Hospital
| | - Noriko Ishii
- Department of Nursing, Sakakibara Heart Institute
| | - Kazuhiro P Izawa
- Department of Public Health, Graduate School of Health Sciences, Kobe University
| | - Nobuyuki Kagiyama
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | | | - Sachiko Kanki
- Department of Thoracic and Cardiovascular Surgery, Osaka Medical and Pharmaceutical University
| | - Katsuhito Kato
- Department of Hygiene and Public Health, Nippon Medical School
| | - Aya Komuro
- Department of Geriatric Medicine, The University of Tokyo Hospital
| | - Nao Konagai
- Department of Obstetrics and Gynecology, National Cerebral and Cardiovascular Center
| | - Yuto Konishi
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Fumie Nishizaki
- Department of Cardiology and Nephrology, Hirosaki University Graduate School of Medicine
| | - Satsuki Noma
- Department of Cardiovascular Medicine, Nippon Medical School
| | | | - Yoshimi Numao
- Department of Cardiology, Itabasih Chuo Medical Center
| | | | - Kimie Okubo
- Division of Cardiology, Department of Medicine, Nihon University School of Medicine Itabashi Hospital
| | | | - Yuka Otaki
- Department of Radiology, Sakakibara Heart Institute
| | | | - Junsuke Shibuya
- Division of Cardiovascular Intensive Care, Nippon Medical School Hospital
| | - Mai Shimbo
- Department of Cardiovascular Medicine, Department of Computational Diagnostic Radiology and Preventive Medicine, The University of Tokyo
| | - Reiko Shiomura
- Division of Cardiovascular Intensive Care, Nippon Medical School Hospital
| | | | - Takahiro Suzuki
- Department of Cardiovascular Medicine, St. Luke's International Hospital
| | - Emi Tajima
- Department of Cardiology, Tokyo General Hospital
| | - Ayako Tsukihashi
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Haruyo Yasui
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | | | - Shun Kohsaka
- Department of Cardiology, Keio University School of Medicine
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | | | - Soko Setoguchi
- Division of Education, Department of Medicine, Rutgers Robert Wood Johnson Medical School
- Division of Cardiovascular Disease and Hypertension, Department of Medicine, Rutgers Robert Wood Johnson Medical School
| | | | | | | |
Collapse
|
10
|
Glenn AJ, Tessier AJ, Kavanagh ME, Morgan GA, Clish CB, Salas-Salvado J, Malik VS, Hanley AJ, Bazinet RP, Comelli EM, El-Sohemy A, Liu S, Boucher BA, Kendall CWC, Jenkins DJA, Hu FB, Sievenpiper JL. Metabolomic profiling of a cholesterol lowering plant-based diet from two randomized controlled feeding trials. Eur J Clin Nutr 2025:10.1038/s41430-025-01625-x. [PMID: 40263496 DOI: 10.1038/s41430-025-01625-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Objective biomarkers of diet, such as metabolomics, may improve dietary assessment and provide additional insight into how diet influences disease risk. The portfolio diet, a cholesterol-lowering plant-based diet, is recommended for lowering low-density lipoprotein cholesterol (LDL-C). This diet is low in saturated fat and includes nuts, plant protein (legumes), viscous fiber, and phytosterols. OBJECTIVE We examined metabolomic profiles in response to the portfolio diet in two randomized controlled trials (RCTs), where all foods were provided to the participants, compared to a control vegetarian diet and the same control diet with a statin. METHODS The first RCT included 34 adults (age 58.4 ± 8.6 y) and the second RCT included 25 adults (age 61.0 ± 9.6 y), all with high LDL-C (>4.1 mmol/L). Plasma samples were obtained at baseline, week 2, and week 4 in both RCTs for metabolomics analysis using liquid chromatography-tandem mass spectrometry. Linear mixed models were used to examine effects of the interventions on the metabolites in each RCT, applying a Bonferroni correction. RESULTS Of 496 known metabolites, 145 and 63 metabolites significantly changed within the portfolio diet interventions in the first and second RCT, respectively. The majority were glycerophosphocholines (32%), triacylglycerols (20%), glycerophosphoethanolamines (14%), sphingomyelins (8%), and amino acids and peptides (8%) in the first RCT, and glycerophosphocholines (48%), glycerophosphoethanolamines (17%), and amino acids and peptides (8%) in the second RCT. Fifty-two metabolites were consistently changed in the same direction with the portfolio diet intervention across both RCTs, after Bonferroni correction. CONCLUSIONS Many of these metabolites likely reflect the plant-based nature, low saturated fat content, and cholesterol-lowering effects of the diet, such as increased N2-acetylornithine, L-pipecolic acid, lenticin, and decreased C18:0 lipids and cholesteryl esters. Further research is needed to validate these metabolites as biomarkers of a plant-based dietary pattern.
Collapse
Affiliation(s)
- Andrea J Glenn
- Department of Nutrition and Food Studies, New York University, New York, NY, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada.
| | - Anne-Julie Tessier
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
- Montreal Heart Institute, Montreal, QC, Canada
- Institut de Valorisation des Données (IVADO), Montreal, QC, Canada
| | - Meaghan E Kavanagh
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada
| | - Gloria A Morgan
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada
- School of Nutrition, Toronto Metropolitan University, Toronto, ON, Canada
| | - Clary B Clish
- Metabolomics Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jordi Salas-Salvado
- CIBER Fisiopatologıa de la Obesidad y Nutricion (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institut d'Investigacions Sanitàries Pere i Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Vasanti S Malik
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anthony J Hanley
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Elena M Comelli
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ahmed El-Sohemy
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Simin Liu
- Department of Epidemiology and Biostatistics, Joe C. Wen School of Population & Public Health, UC Irvine, Irvine, CA, USA
- Center for Global Cardiometabolic Health & Nutrition (CGCHN), Mary & Steve Wen Division of Cardiology, Department of Medicine, School of Medicine, UC Irvine, Irvine, CA, USA
- Division of Endocrinology Department of Medicine, and Division of Cardiothoracic Surgery Department of Surgery, Warren Alpert School of Medicine and Rhode Island Hospital, Providence, RI, USA
- Department of Epidemiology, Brown University, School of Public Health, Providence, RI, USA
| | - Beatrice A Boucher
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Cyril W C Kendall
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - David J A Jenkins
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Toronto, ON, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - John L Sievenpiper
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, ON, Canada.
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada.
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Toronto, ON, Canada.
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Vu S, Mahmoud N, Sivakumar S, Yazdanian F, Fisher M. Dilemmas in Secondary Stroke Prevention. Stroke 2025. [PMID: 40255157 DOI: 10.1161/strokeaha.124.050403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Secondary stroke prevention encompasses many approaches, including antithrombotic therapy, risk factor management, and a healthy lifestyle. Recommendations are typically based on the results of randomized clinical trials that provide evidence of benefit. However, in some situations, clinicians extrapolate the results of clinical trials into everyday practice, or trials have not provided sufficient information to make treatment decisions. This review will discuss 4 scenarios: dual-antiplatelet therapy, the perils of combining antiplatelet and anticoagulation, indications for statin therapy, and therapeutic considerations for patients with paroxysmal atrial fibrillation.
Collapse
Affiliation(s)
- Stephanie Vu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. (S.V., N.M., S.S., M.F.)
| | - Noor Mahmoud
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. (S.V., N.M., S.S., M.F.)
| | - Shravan Sivakumar
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. (S.V., N.M., S.S., M.F.)
| | - Forough Yazdanian
- Neurosurgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. (F.Y.)
| | - Marc Fisher
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. (S.V., N.M., S.S., M.F.)
| |
Collapse
|
12
|
M’barek L, Jin A, Pan Y, Lin J, Jiang Y, Meng X, Wang Y. Stroke Prognosis: The Impact of Combined Thrombotic, Lipid, and Inflammatory Markers. J Atheroscler Thromb 2025; 32:458-473. [PMID: 39462537 PMCID: PMC11973529 DOI: 10.5551/jat.64984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 08/25/2024] [Indexed: 10/29/2024] Open
Abstract
AIM D-dimer, lipoprotein (a) (Lp(a)), and high-sensitivity C-reactive protein (hs-CRP) are known predictors of vascular events; however, their impact on the stroke prognosis is unclear. This study used data from the Third China National Stroke Registry (CNSR-III) to assess their combined effect on functional disability and mortality after acute ischemic stroke (AIS). METHODS In total, 9,450 adult patients with AIS were enrolled between August 2015 and March 2018. Patients were categorized based on a cutoff value for D-dimer, Lp(a), and hs-CRP in the plasma. Adverse outcomes included poor functional outcomes (modified Rankin Scale (mRS score ≥ 3)) and one- year all-cause mortality. Logistic and multivariate Cox regression analyses were performed to investigate the relationship between individual and combined biomarkers and adverse outcomes. RESULTS Patients with elevated levels of all three biomarkers had the highest odds of functional disability (OR adjusted: 2.01; 95% CI (1.47-2.74); P<0.001) and mortality (HR adjusted: 2.93; 95% CI (1.55-5.33); P<0.001). The combined biomarkers improved the predictive accuracy for disability (C-statistic 0.80 vs.0.79, P<0.001) and mortality (C-statistic 0.79 vs.0.78, P=0.01). CONCLUSION Elevated D-dimer, Lp(a), and hs-CRP levels together increase the risk of functional disability and mortality one-year post-AIS more than any single biomarker.
Collapse
Affiliation(s)
- Lamia M’barek
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Aoming Jin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuesong Pan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinxi Lin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Jiang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xia Meng
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, 2019RU018, Beijing, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
13
|
Flo GL, Alzate Aguirre M, Gochanour BR, Hynes KJ, Scott CG, Fink AL, M Arruda-Olson A. Pharmacist-Initiated Team-Based Intervention for Optimizing Guideline-Directed Lipid Therapy of Hospitalized Patients With Acute Coronary Syndrome: Pilot Study Using a Stepped-Wedge Cluster Design. JMIR Cardio 2025; 9:e58837. [PMID: 40153788 PMCID: PMC11970799 DOI: 10.2196/58837] [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: 03/26/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 03/30/2025] Open
Abstract
Background Clinical guidelines recommend high-intensity statin therapy for patients with acute coronary syndrome (ACS). However, high-intensity statins have been underused in this population. Objective The objective of this study was to evaluate the feasibility of a pharmacist-initiated, team-based intervention for the delivery of individualized, guideline-directed, lipid-lowering therapy for patients with ACS. Methods Patients admitted with ACS to cardiology hospital services at Mayo Clinic from August 1, 2021, to June 19, 2022, were assigned to a pharmacist-initiated, team-based intervention group or control group using a stepped wedge cluster study design. For the intervention group, pharmacists reviewed electronic health records and provided recommendations for lipid lowering therapy in hospital and at follow-up. In the control group, patients received usual care. Neither care team, nor study team were blinded to study assignments. The primary outcome was the proportion of patients with ACS discharged on high-intensity statins in the intervention group compared to controls. Secondary outcomes were (1) proportion of patients in the intervention group with a specific templated pharmacist intervention note in their electronic health records, (2) frequency of low-density lipoprotein (LDL) measurements in hospital, (3) proportion of patients with information related to lipid follow-up in their discharge summary, and (4) proportion of patients that received LDL monitoring at the outpatient follow-up 4 to 12 weeks post discharge. Results There were 410 patients included in this study (median age 68, IQR 60-78 years) of whom 285 (69.5%) were male. Of the 402 patients alive at discharge, 355 (88.3%) were discharged taking a high-intensity statin, with no significant difference (P=.89) observed between groups. Lipid levels were measured in the hospital for 176/210 (83.8%) patients in the intervention group and 155/200 (77.5%) patients in the control group (P=.14). Fifty-four of 205 (26.3%) intervention patients alive at discharge had lipid-related recommendations in their discharge summary compared to 27/197 (13.7%) controls (P=.002). Forty-seven of 81 (58%) patients with lipid management recommendations provided in the discharge summary had LDL measured in the follow-up period compared with only 119/321 (37.1%) patients without these recommendations (P=.001). Of the 402 patients who survived to discharge, 166 (41.3%) had LDL measured at follow-up; the median LDL level was 63.5 (IQR 49-79) mg/dL, and distributions were similar by group (P=.95). Only 101/166 (60.8%) patients had follow-up LDL values below the target of 70 mg/dL. Conclusions During hospitalization, there was no group difference in the primary outcome of high-intensity statin therapy. Feasibility of an effective pharmacist-initiated intervention for improvement of lipid management was demonstrated by entry of recommendations in the discharge summary and related adjustment in outpatient statin therapy. The main opportunity for future improvement in lipid management of patients with ACS is in longitudinal patient follow-up.
Collapse
Affiliation(s)
- Gayle L Flo
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, United States, 1 507-284-2511
| | - Mateo Alzate Aguirre
- Ascension St. John Hospital, Detroit, MI, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Benjamin R Gochanour
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Kristin J Hynes
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, United States, 1 507-284-2511
- Pharmacy Services, Mayo Clinic, Rochester, MN, United States
| | - Christopher G Scott
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Angela L Fink
- Department of Nursing, Mayo Clinic, Rochester, MN, United States
| | - Adelaide M Arruda-Olson
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, United States, 1 507-284-2511
| |
Collapse
|
14
|
Li X, Lu L, Chen Y, Liu B, Liu B, Tian H, Yang H, Guo R. Association of atherogenic index of plasma trajectory with the incidence of cardiovascular disease over a 12-year follow-up: findings from the ELSA cohort study. Cardiovasc Diabetol 2025; 24:124. [PMID: 40108582 PMCID: PMC11924681 DOI: 10.1186/s12933-025-02677-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/08/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Atherogenic index of plasma (AIP) at baseline has been associated with increased morbidity and mortality from cardiovascular disease (CVD). However, the relationship between long-term AIP trajectories and CVD remains unclear. Therefore, this study aimed to investigate the associations between AIP trajectories and the incidence of CVD in the English population. METHOD The study data analysis was based on the English Longitudinal Study of Aging (ELSA) from 2004 to 2017. The study population consisted of individuals aged 50 years and older in England. AIP was calculated as log10 (triglycerides/high-density lipoprotein cholesterol). Group-based trajectory model (GBTM) was applied to identify the trajectory of the AIP index from Wave 2 to 8 over a 12-year follow-up. Cox proportional hazard models were then used to analyze the associations between different AIP index trajectory groups and the incidence of CVD. RESULTS A total of 3976 participants with completed AIP data in Wave 2 and more than two AIP measurements between Wave 2 and Wave 8 were enrolled in the ELSA cohort. The participants were divided into three groups [low-stable group (n = 1146), moderate-stable group (n = 2110), high-stable group (n = 720)] using a GBTM model. After adjusting for potential confounders, participants in the high-stable group indicated an increased risk of developing incident of CVD compared to those in the low-stable AIP group [Hazard Ratio (HR) 1.33; 95% Confidence Interval (CI) 1.02-1.74, P = 0.033]. However, no differences in the incidence of CVD (HR 1.20, 95%CI 0.98-1.48, P = 0.082) were observed in the moderate-stable group. Subgroup analysis indicated similar results for participants under 63 years old and those with high alcohol consumption. CONCLUSIONS A high and sustainable level of the AIP index may contribute to the incidence of CVD. The trajectories of the AIP index can help identify older English individuals at increased risk of CVD who deserve primitive preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Xicong Li
- Kunming Medical University, Kunming, 650032, Yunnan, China
- 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650032, Yunnan, China
| | - Lifei Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yubiao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Baiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Bei Liu
- 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650032, Yunnan, China
| | - Heshen Tian
- Department of Respiratory and Critical Care Medicine Key Laboratory of Respiratory Disease of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Huajing Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Ruiwei Guo
- 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650032, Yunnan, China.
- Department of Cardiology, 922th Hospital of Joint Logistics Support Force, PLA, Hengyang, 421000, Hunan, China.
| |
Collapse
|
15
|
Paraskevas T, Gakis G, Papapanou M, Sergentanis TN, Sotiriadis A, Siristatidis CS. Statins for preventing preeclampsia. Cochrane Database Syst Rev 2025; 3:CD016133. [PMID: 40099754 PMCID: PMC11915783 DOI: 10.1002/14651858.cd016133] [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: 03/20/2025]
Abstract
OBJECTIVES This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To evaluate the relative benefits and harms of statins for preeclampsia prevention in pregnant women.
Collapse
Affiliation(s)
| | - Georgios Gakis
- General University Hospital of Patras, University of Patras, Patras, Greece
| | - Michail Papapanou
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros N Sergentanis
- Department of Public Health Policy, School of Public Health, University of West Attica, Athens, Greece
| | - Alexandros Sotiriadis
- Second Department of Obstetrics and Gynaecology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Charalampos S Siristatidis
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
16
|
Wei Q, Cheng X, Li M, Wu K, Chen M, Zhang D. Associations between the cardiometabolic index and atherosclerotic cardiovascular disease acorss different glucose metabolism statuses: insights from NHANES, 1999-2020. Lipids Health Dis 2025; 24:93. [PMID: 40089751 PMCID: PMC11910843 DOI: 10.1186/s12944-025-02508-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/27/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND The cardiometabolic index (CMI) serves as a significant marker of diabetes mellitus (DM) and may predict the potential for cardiovascular disease development. Nevertheless, the correlation between CMI and atherosclerotic cardiovascular disease (ASCVD) among individuals exhibiting varying glucose metabolism statuses (GMS) continues to be unclear. METHODS Overall, 24,006 individuals aged 20 and above were enrolled in the research, drawn from the National Health and Nutrition Examination Survey (NHANES) database. Individuals in the study was classified into three distinct categories according to the level of fasting plasma glucose or glycated haemoglobin: normal glucose regulation, prediabetes, and DM. Multivariate logistic regression models and smoothed curve-fitting techniques were applied to investigate the correlation between CMI and ASCVD risk across varying GMS. Additionally, subgroup analyses stratified by relevant factors were performed to identify potential effect modifiers in this relationship. RESULTS Overall, 2352 participants (9.8%) with ASCVD were included. An increasing trend in ASCVD risk was observed for each successive CMI tertile. After adjusting for all related covariates, a significantly positive association was observed between CMI and ASCVD (P = 0.0004). Participants with DM in the highest CMI tertile had a 114% higher ASCVD risk compared to those in the lowest tertile (OR = 2.14; 95% CI = 1.30-3.53). Smoothed curve-fitting consistently confirmed the correlation between CMI and ASCVD across diverse GMS. Subgroup analyses and interaction tests highlighted statistically significant differences within the drinking status subgroup (P-interaction = 0.0479) and GMS subgroups (P-interaction = 0.0397). CONCLUSION This research suggests a positive association between ASCVD and CMI in adults in the United States, particularly among individuals with DM.
Collapse
Affiliation(s)
- Qiong Wei
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Hospital, No.5 Haiyuncang Hutong, Dongcheng District, Beijing, 100700, China
| | - Xu Cheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Hospital, No.5 Haiyuncang Hutong, Dongcheng District, Beijing, 100700, China
| | - Min Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Hospital, No.5 Haiyuncang Hutong, Dongcheng District, Beijing, 100700, China
| | - Keying Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Hospital, No.5 Haiyuncang Hutong, Dongcheng District, Beijing, 100700, China
| | - Meng Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Sunshine South Street, Fangshan District, Beijing, 100029, China.
| | - Dongmei Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Hospital, No.5 Haiyuncang Hutong, Dongcheng District, Beijing, 100700, China.
| |
Collapse
|
17
|
Kolossváry M, Schnittman SR, Zanni MV, Fitch KV, Fichtenbaum CJ, Aberg JA, Bloomfield GS, Malvestutto CD, Currier J, Diggs MR, deFilippi C, Eckard AR, Curran A, Centinbas M, Sadreyev R, Foldyna B, Mayrhofer T, Karady J, Taron J, McCallum S, Lu MT, Ribaudo HJ, Douglas PS, Grinspoon SK. Pitavastatin, Procollagen Pathways, and Plaque Stabilization in Patients With HIV: A Secondary Analysis of the REPRIEVE Randomized Clinical Trial. JAMA Cardiol 2025; 10:254-264. [PMID: 39661372 PMCID: PMC11771813 DOI: 10.1001/jamacardio.2024.4115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/29/2024] [Indexed: 12/12/2024]
Abstract
Importance In a mechanistic substudy of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) randomized clinical trial, pitavastatin reduced noncalcified plaque (NCP) volume, but specific protein and gene pathways contributing to changes in coronary plaque remain unknown. Objective To use targeted discovery proteomics and transcriptomics approaches to interrogate biological pathways beyond low-density lipoprotein cholesterol (LDL-C), relating statin outcomes to reduce NCP volume and promote plaque stabilization among people with HIV (PWH). Design, Setting, and Participants This was a post hoc analysis of the double-blind, placebo-controlled, REPRIEVE randomized clinical trial. Participants underwent coronary computed tomography angiography (CTA), plasma protein analysis, and transcriptomic analysis at baseline and 2-year follow-up. The trial enrolled PWH from April 2015 to February 2018 at 31 US research sites. PWH without known cardiovascular diseases taking antiretroviral therapy and with low to moderate 10-year cardiovascular risk were eligible. Data analyses were conducted from October 2023 to February 2024. Intervention Oral pitavastatin calcium, 4 mg per day. Main Outcomes and Measures Relative change in plasma proteomics, transcriptomics, and noncalcified plaque volume among those receiving treatment vs placebo. Results Among 558 individuals (mean [SD] age, 51 [6] years; 455 male [82%]) included in the proteomics assessment, 272 (48.7%) received pitavastatin and 286 (51.3%) received placebo. After adjusting for false discovery rates, pitavastatin increased abundance of procollagen C-endopeptidase enhancer 1 (PCOLCE), neuropilin 1 (NRP-1), major histocompatibility complex class I polypeptide-related sequence A (MIC-A) and B (MIC-B), and decreased abundance of tissue factor pathway inhibitor (TFPI), tumor necrosis factor ligand superfamily member 10 (TRAIL), angiopoietin-related protein 3 (ANGPTL3), and mannose-binding protein C (MBL2). Among these proteins, the association of pitavastatin with PCOLCE (a rate-limiting enzyme of collagen deposition) was greatest, with an effect size of 24.3% (95% CI, 18.0%-30.8%; P < .001). In a transcriptomic analysis, individual collagen genes and collagen gene sets showed increased expression. Among the 195 individuals with plaque at baseline (88 [45.1%] taking pitavastatin, 107 [54.9%] taking placebo), changes in NCP volume were most strongly associated with changes in PCOLCE (%change NCP volume/log2-fold change = -31.9%; 95% CI, -42.9% to -18.7%; P < .001), independent of changes in LDL-C level. Increases in PCOLCE related most strongly to change in the fibro-fatty (<130 Hounsfield units) component of NCP (%change fibro-fatty volume/log2-fold change = -38.5%; 95% CI, -58.1% to -9.7%; P = .01) with a directionally opposite, although nonsignificant, increase in calcified plaque (%change calcified volume/log2-fold change = 34.4%; 95% CI, -7.9% to 96.2%; P = .12). Conclusions and Relevance Results of this secondary analysis of the REPRIEVE randomized clinical trial suggest that PCOLCE may be associated with the atherosclerotic plaque stabilization effects of statins by promoting collagen deposition in the extracellular matrix transforming vulnerable plaque phenotypes to more stable coronary lesions. Trial Registration ClinicalTrials.gov Identifier: NCT02344290.
Collapse
Affiliation(s)
- Márton Kolossváry
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Samuel R. Schnittman
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
- Division of Infectious Diseases, Massachusetts General Hospital, Boston
| | - Markella V. Zanni
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Kathleen V. Fitch
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Carl J. Fichtenbaum
- Division of Infectious Diseases, Department of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Judith A. Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gerald S. Bloomfield
- Department of Medicine, Duke Global Health Institute, Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | | | - Judith Currier
- Division of Infectious Diseases, University of California, Los Angeles, Los Angeles, California
| | - Marissa R. Diggs
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | | | - Allison Ross Eckard
- Departments of Pediatrics and Medicine, Divisions of Infectious Diseases, Medical University of South Carolina, Charleston
| | - Adrian Curran
- Infectious Diseases Department, Vall d’Hebron Research Institute, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Murat Centinbas
- Department of Molecular Biology, Massachusetts General Hospital, Boston
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Julia Karady
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- Cardiovascular Imaging Research Group, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Jana Taron
- Department of Molecular Biology, Massachusetts General Hospital, Boston
- Department of Radiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Sara McCallum
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Michael T. Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Heather J. Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Pamela S. Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Steven K. Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
18
|
Di Fusco SA, Volpe M, Nardi F, Matteucci A, Aquilani S, Marino G, Aiello A, Colivicchi F. Reducing LDL-Cholesterol to Very Low Levels: Sailing Between Established Benefits and Potential Risks. High Blood Press Cardiovasc Prev 2025; 32:139-149. [PMID: 39998740 DOI: 10.1007/s40292-025-00708-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: 12/16/2024] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
In view of the growing evidence supporting more marked reductions of low-density lipoprotein cholesterol (LDL-C), according to the concept of "the lower is better" and with the availability of powerful and well tolerated lipid-lowering drugs, physicians are facing today with the clinical management of patients with very low LDL-C levels. The fear of potential risks linked to extreme reductions of LDL-C down to very low levels may lead to the de-escalation of treatments with consequent paradoxical unfavorable consequences due to the exposure to a higher cardiovascular risk. The aim of this review is to point out evidence of very low LDL-C clinical impact, with a focus on potential adverse effects. Research on cholesterol homeostasis has identified complex mechanisms which guarantee cell functions even when circulating cholesterol levels are very low. The almost complete self-sufficiency of the human body in terms of cholesterol needs is confirmed by evidence derived from genetically determined models with very low LDL-C levels. Studies on the potential harm of lowering LDL-C to very low concentrations do not confirm an increased risk of cancer or neurodegenerative disease attributable to lipid-lowering treatments, whereas evidence suggests a potential benefit in these settings. A potential increased risk of hemorrhagic stroke has been reported, suggesting tight monitoring and control of blood pressure should be implemented in patients with very low LDL-C levels. With regard to statin treatment, a dose-dependent increased risk of newly diagnosed diabetes has been reported. This adverse effect has not been found with more recently approved lipid-lowering drugs.
Collapse
Affiliation(s)
- Stefania Angela Di Fusco
- Clinical and Rehabilitation Cardiology Unit, Emergency Department, San Filippo Neri Hospital, ASL Rome 1, Rome, Italy.
- Clinical and Rehabilitation Cardiology Unit, San Filippo Neri Hospital, Via Martinotti 20, 00135, Rome, Italy.
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035, 00189, Rome, Italy
- IRCCS San Raffaele Roma, Rome, Italy
| | | | - Andrea Matteucci
- Clinical and Rehabilitation Cardiology Unit, Emergency Department, San Filippo Neri Hospital, ASL Rome 1, Rome, Italy
| | - Stefano Aquilani
- Clinical and Rehabilitation Cardiology Unit, Emergency Department, San Filippo Neri Hospital, ASL Rome 1, Rome, Italy
| | - Gaetano Marino
- Clinical and Rehabilitation Cardiology Unit, Emergency Department, San Filippo Neri Hospital, ASL Rome 1, Rome, Italy
| | - Alessandro Aiello
- Clinical and Rehabilitation Cardiology Unit, Emergency Department, San Filippo Neri Hospital, ASL Rome 1, Rome, Italy
| | - Furio Colivicchi
- Clinical and Rehabilitation Cardiology Unit, Emergency Department, San Filippo Neri Hospital, ASL Rome 1, Rome, Italy
| |
Collapse
|
19
|
Hu Y, Cui X, Lu M, Guan X, Li Y, Zhang L, Lin L, Zhang Z, Zhang M, Hao J, Wang X, Huan J, Li Y, Li C. Body Fat Distribution and Ectopic Fat Accumulation as Mediator of Diabetogenic Action of Lipid-Modifying Drugs: A Mediation Mendelian Randomization Study. Mayo Clin Proc 2025; 100:424-439. [PMID: 39918451 DOI: 10.1016/j.mayocp.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/06/2024] [Accepted: 10/25/2024] [Indexed: 05/08/2025]
Abstract
OBJECTIVE To investigate the causal relationship between various lipid-modifying drugs and new-onset diabetes, as well as the mediators contributing to this relationship. METHODS Mediation Mendelian randomization was performed to investigate the causal effect of lipid-modifying drug targets on type 2 diabetes (T2D) outcomes and the proportion of this association that is mediated through ectopic fat accumulation traits. Specific sets of variants in or near genes that encode 11 lipid-modifying drug targets (LDLR, HMGCR, NPC1L1, PCSK9, APOB, ABCG5/ABCG8, LPL, PPARA, ANGPTL3, APOC3, and CETP; for expansion of gene symbols, use search tool at www.genenames.org) were extracted. Random effects inverse variance weighted were performed to evaluate the causal effects among outcomes. Mediation analyses were performed to identify the mediators of the association between lipid-modifying drugs and T2D. The study was conducted from November 10, 2023, to April 2, 2024 RESULTS: The genetic mimicry of HMGCR and APOB inhibition was associated with an increased T2D risk, whereas the genetic mimicry of LPL enhancement was linked to a lower T2D risk. Gluteofemoral adipose tissue volume was a mediator for explaining 9.52% (P=.002), 16.90% (P=.03), and 10.50% (P=.003) of the total effect of HMGCR, APOB, and LPL on T2D susceptibility, respectively. Liver fat was a mediator for explaining 21.12% (P=.005), 12.28% (P=.03), and 9.84% (P=.005) of the total effect of HMGCR, APOB, and LPL on T2D susceptibility, respectively. CONCLUSION Our findings support the hypothesis that liver fat and gluteofemoral adipose tissue play a mediating role in the prodiabetic effects of HMGCR and APOB inhibition, as well as in the antidiabetic effects of LPL enhancement.
Collapse
Affiliation(s)
- Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinhai Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiuya Guan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuan Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Muxin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jiaqi Hao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaojie Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Jiaming Huan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yunlun Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China; Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
20
|
Yamashita S, Araki E, Arai H, Yokote K, Tanigawa R, Saito A, Suganami H, Minamikawa S, Ishibashi S. Clinical Pharmacology of Pemafibrate Extended-release Formulation in Patients with Hypertriglyceridemia-A Phase 2, Multicenter, Active-controlled, Randomized, Single-blind, Crossover study. J Atheroscler Thromb 2025; 32:367-384. [PMID: 39322570 PMCID: PMC11883216 DOI: 10.5551/jat.65001] [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: 03/28/2024] [Accepted: 08/05/2024] [Indexed: 09/27/2024] Open
Abstract
AIMS Efficacy, safety, and pharmacokinetics of the selective PPARα modulator pemafibrate as once-daily extended-release (XR) tablets were compared with those of twice-daily immediate-release (IR) tablets in patients with hypertriglyceridemia. METHODS A multicenter, randomized, single-blind, active-controlled crossover, phase 2 clinical pharmacology study was performed in patients with hypertriglyceridemia. Patients were randomly assigned to IR 0.2 mg/day, XR 0.4 mg/day, or XR 0.8 mg/day before/after meals (fasted/fed) and treated for a total of eight weeks. The primary endpoint was percentage change in fasting serum triglycerides (TG). RESULTS Of 63 randomized patients, 60 received the study drug. Patients were 78.3% male, mean age (±SD) 57.5±9.8 years, BMI 25.5±3.7 kg/m2, and fasting TG 221.3±68.1 mg/dL. Fasting serum TG decreased significantly from baseline in all groups (LS mean [95% CI];-43.6 [-47.7, -39.5] % for IR 0.2 mg/day, -41.1 [-45.1, -37.0] % for XR 0.4mg/day, -39.7 [-43.8, -35.6] % for XR 0.8 mg/day), indicating that XR 0.4 and XR 0.8 mg/day were not inferior to IR 0.2 mg/day. TG-lowering effects tended to be stronger for fed than fasted administration. MRTss, tmax, and t1/2 were longer for XR than for IR. Adverse events showed no major inter-group differences: 12.5% (5/40 patients) for IR 0.2, 17.5% (7/40) for XR 0.4, and 20.0% (8/40) for XR 0.8 mg/day. CONCLUSIONS In patients with hypertriglyceridemia, XR substantially lowered TG at all doses, with maximum effectiveness at 0.4 mg/day, the dose approved in Japan, to a level comparable to IR 0.2 mg/day. There were no safety concerns up to 0.8 mg/day.
Collapse
Affiliation(s)
| | - Eiichi Araki
- Kikuchi Medical Association Hospital
- Research Center for Health and Sports Sciences, Kumamoto Health Science University
| | | | | | | | - Ayumi Saito
- Global Clinical Development Department, Kowa Company Ltd
| | | | | | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University
- Ishibashi Diabetes and Endocrine Clinic
| |
Collapse
|
21
|
Fujino M, Di Giovanni G, Nicholls SJ. New Approaches to Lipoproteins for the Prevention of Cardiovascular Events. J Atheroscler Thromb 2025; 32:265-280. [PMID: 39756980 PMCID: PMC11883213 DOI: 10.5551/jat.rv22031] [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: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 01/07/2025] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is a leading global cause of mortality, and recent research has underscored the critical role of lipoproteins in modulating cardiovascular (CV) risk. Elevated low-density lipoprotein cholesterol (LDL-C) levels have been linked to increased CV events, and while numerous trials have confirmed the efficacy of lipid-lowering therapies (LLT), significant gaps remain between recommended LDL-C targets and real-world clinical practice. This review addresses care gaps in LLT, emphasizing the necessity for innovative approaches that extend beyond LDL-C management. It explores combination therapy approaches such as statins combined with ezetimibe or PCSK9 inhibitors, which have shown promise in enhancing LDL-C reduction and improving outcomes in high-risk patients. Additionally, this review discusses new approaches in lipid modification strategies, including bempedoic acid, inclisiran, and drugs that lower Lp(a), highlighting their potential for CV risk reduction. Furthermore, it emphasizes the potential of polygenic risk scores to guide LLT and lifestyle changes despite challenges in implementation and genetic testing ethics. This article discusses the current guidelines and proposes innovative approaches for optimizing lipoprotein management, ultimately contributing to improved patient outcomes in ASCVD prevention.
Collapse
|
22
|
Ding M, Chen H, Lin FC. A discrete-time split-state framework for multi-state modeling with application to describing the course of heart disease. BMC Med Res Methodol 2025; 25:54. [PMID: 40021971 PMCID: PMC11869649 DOI: 10.1186/s12874-025-02512-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
In chronic disease epidemiology, the investigation of disease etiology has largely focused on an endpoint, while the course of chronic disease is understudied, representing a knowledge gap. Multi-state models can be used to describe the course of chronic disease, such as Markov models which assume that the future state depends only on the present state, and semi-Markov models which allow transition rates to depend on the duration in the current state. However, these models are unsuitable for chronic diseases that are largely non-memoryless. We propose a Discrete-Time Split-State Framework that generates a process of substates by conditioning on past disease history and estimates discrete-time transition rates between substates as a function of duration in a (sub)state. Specifically, as the substates are created by conditioning on past history, they satisfy the Markov assumption, regardless of whether the original disease process is Markovian; and the transition rates are approximated by competing risks in a short time interval estimated from cause-specific Cox models. In the simulation study, we simulated a Markov process with an exponential distribution, a semi-Markov process with a Weibull distribution, and a non-Markov process with an exponential distribution. The coverage rate of transition rates estimated using our framework was 94% for the Markov process and 93% for the non-Markov process. However, the estimated transition rates were under coverage (72%) for the semi-Markov process, which is likely due to the approximation of transition rates in discrete time. In the application, we applied the framework to describe the course of heart disease in a large cohort study. In summary, the framework we proposed can be applied to both Markov and non-Markov processes and has potential to be applied to semi-Markov processes. For future research, as substates created using our framework track past disease history, the transition rates between substates have the potential to be used to derive summary estimates that characterize the disease course.
Collapse
Affiliation(s)
- Ming Ding
- Department of Emergency Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Haiyi Chen
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Feng-Chang Lin
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Li C, Liang Y, Lu Q, Lin Y, Wen S, Luo X, Huang S, Zhong X, Xu Z, Wang F. Protective effect of serum carotenoids on mortality among metabolic syndrome patients: attenuated by lipid-lowering drugs. Nutr J 2025; 24:27. [PMID: 39972496 PMCID: PMC11837722 DOI: 10.1186/s12937-025-01092-x] [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: 10/24/2024] [Accepted: 02/12/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Limited evidence exists about the relationship between serum carotenoid and mortality in metabolic syndrome (MetS) patients, and the effects of medication use on this association remains unclear. METHODS The study encompassed 2,521 MetS patients from the National Health and Nutrition Examination Survey (NHANES) 2001-2006 and 2017-2018. A total of 7 serum carotenoids were evaluated. Death data were sourced from the National Death Index, with causes assessed using ICD-10 codes. Bayesian kernel machine regression (BKMR) and random survival forest (RSF) were utilized to investigate serum carotenoid mixture on mortality and identify key carotenoids. "Qgcompint" R package was used to explore the modifying effects of medication use. RESULTS The serum carotenoid levels at baseline ranged from 0.04 to 1.37 µmol/L. During a follow-up of 15.1 years, there were 696 deaths (27.61%), with 247 (35.49%) by cardiovascular disease (CVD), 148 (21.26%) by cancer, and 301 (43.25%) by other diseases. Individual and combined serum carotenoids were negatively associated with all-cause mortality (HR range:0.70-0.88, 95%CI range:0.56-0.99, all P < 0.05). α-carotene (VIMP = 0.223 in RSF) and lutein/zeaxanthin (PIP = 1.000 in BKMR) emerged as the greatest contributors to all-cause mortality. Lipid-lowering drugs attenuate the negative effect of serum carotenoids on MetS patients' mortality (Pint = 0.014). CONCLUSION The present study identified a protective effect of serum carotenoid on mortality in MetS patients, which was probably weakened by lipid-lowering drugs. Early dietary interventions for MetS patients taking lipid-lowering drugs, particularly those rich in carotenoids like α-carotene and lutein/zeaxanthin, could help reduce mortality.
Collapse
Affiliation(s)
- Chunxiang Li
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yanlan Liang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Qiuyuan Lu
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuanxin Lin
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Shifeng Wen
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoyu Luo
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Shiping Huang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xue Zhong
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - ZhangJian Xu
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Fei Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
24
|
Kim JT, Lee JS, Kim H, Kim BJ, Kang J, Lee KJ, Park JM, Kang K, Lee SJ, Kim JG, Cha JK, Kim DH, Park TH, Lee K, Lee J, Hong KS, Cho YJ, Park HK, Lee BC, Yu KH, Oh MS, Kim DE, Choi JC, Kwon JH, Kim WJ, Shin DI, Yum KS, Sohn SI, Hong JH, Lee SH, Park MS, Ryu WS, Park KY, Lee J, Saver JL, Bae HJ. Comparative Effectiveness of Rosuvastatin Versus Atorvastatin in Acute Ischemic Stroke Treatment. J Am Heart Assoc 2025; 14:e038080. [PMID: 39895542 DOI: 10.1161/jaha.124.038080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND Research specifically addressing the efficacy of rosuvastatin versus atorvastatin in patients with ischemic stroke is insufficient. Using a large stroke registry, we investigated whether 2 commonly used statins, rosuvastatin and atorvastatin, differ in their effectiveness in reducing the risk of vascular events in patients with acute ischemic stroke. METHODS We analyzed data from a nationwide stroke registry in South Korea between January 2011 and April 2022. Patients with acute ischemic stroke within 7 days of onset who were prescribed either atorvastatin or rosuvastatin at discharge were included. The primary outcome was a composite of recurrent stroke (either hemorrhagic or ischemic), myocardial infarction, and all-cause mortality within 1 year. RESULTS A total of 43 512 patients (age, 69.2±12.5 years; male, 59.8%) were analyzed in this study. Atorvastatin was used in 84.8% (n=36 903), and rosuvastatin was used in 15.2% (n=6609). The 1-year cumulative event rate of the composite of recurrent stroke, myocardial infarction, and all-cause mortality was significantly lower in the rosuvastatin group than in the atorvastatin group (9.7% [95% CI, 9.0-10.5] versus 10.7% [95% CI, 10.4-11.0]; P=0.049). Cox proportional hazards analysis revealed that rosuvastatin, compared with atorvastatin, was significantly associated with less risk of 1-year composite of recurrent stroke, myocardial infarction, and all-cause mortality, with an absolute risk reduction of 1% [95% CI, -1.8 to -0.2] and a relative risk reduction of 11% (hazard ratio, 0.89 [95% CI, 0.82-0.97]). However, there were discrepancies in the statistical significance of the results between the propensity score matching and stabilized inverse probability of treatment weighting analysis. CONCLUSIONS The results of this analysis of a large cohort of patients with ischemic stroke suggested that, compared with atorvastatin, rosuvastatin was significantly associated with a reduced risk of a 1-year composite of recurrent stroke, myocardial infarction, and all-cause mortality in patients with acute ischemic stroke. However, in real clinical practice, rosuvastatin is used less than one-fifth as frequently as atorvastatin in patients with acute ischemic stroke. This study serves as a hypothesis-generating function.
Collapse
Affiliation(s)
- Joon-Tae Kim
- Department of Neurology Chonnam National University Hospital, Chonnam National University Medical School Gwangju Korea
| | - Ji Sung Lee
- Clinical Research Center Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine Seoul Korea
| | - Hyunsoo Kim
- Department of Neurology Chonnam National University Hospital, Chonnam National University Medical School Gwangju Korea
| | - Beom Joon Kim
- Department of Neurology Seoul National University College of Medicine, Seoul National University Bundang Hospital Seongnam Korea
| | - Jihoon Kang
- Department of Neurology Seoul National University College of Medicine, Seoul National University Bundang Hospital Seongnam Korea
| | - Keon-Joo Lee
- Department of Neurology Korea University Guro Hospital Seoul Korea
| | - Jong-Moo Park
- Department of Neurology Uijeongbu Eulji Medical Center, Eulji University School of Medicine Uijeongbu-si Korea
| | - Kyusik Kang
- Department of Neurology Nowon Eulji Medical Center, Eulji University School of Medicine Seoul Korea
| | - Soo Joo Lee
- Department of Neurology Daejeon Eulji Medical Center, Eulji University School of Medicine Daejeon Korea
| | - Jae Guk Kim
- Department of Neurology Daejeon Eulji Medical Center, Eulji University School of Medicine Daejeon Korea
| | - Jae-Kwan Cha
- Department of Neurology Dong-A University Hospital Busan Korea
| | - Dae-Hyun Kim
- Department of Neurology Dong-A University Hospital Busan Korea
| | - Tai Hwan Park
- Department of Neurology Seoul Medical Center Seoul Korea
| | - Kyungbok Lee
- Department of Neurology Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine Seoul Korea
| | - Jun Lee
- Department of Neurology Yeungnam University Hospital Daegu Korea
| | - Keun-Sik Hong
- Department of Neurology Ilsan Paik Hospital, Inje University Goyang Korea
| | - Yong-Jin Cho
- Department of Neurology Ilsan Paik Hospital, Inje University Goyang Korea
| | - Hong-Kyun Park
- Department of Neurology Ilsan Paik Hospital, Inje University Goyang Korea
| | - Byung-Chul Lee
- Department of Neurology Hallym University Sacred Heart Hospital Anyang Korea
| | - Kyung-Ho Yu
- Department of Neurology Hallym University Sacred Heart Hospital Anyang Korea
| | - Mi Sun Oh
- Department of Neurology Hallym University Sacred Heart Hospital Anyang Korea
| | - Dong-Eog Kim
- Department of Neurology Dongguk University Ilsan Hospital Goyang Korea
| | - Jay Chol Choi
- Department of Neurology Jeju National University Hospital, Jeju National University School of Medicine Jeju Korea
| | - Jee-Hyun Kwon
- Department of Neurology Ulsan University College of Medicine Ulsan Korea
| | - Wook-Joo Kim
- Department of Neurology Ulsan University College of Medicine Ulsan Korea
| | - Dong-Ick Shin
- Department of Neurology Chungbuk National University Hospital Cheongju Korea
| | - Kyu Sun Yum
- Department of Neurology Chungbuk National University Hospital Cheongju Korea
| | - Sung Il Sohn
- Department of Neurology Keimyung University Dongsan Medical Center Daegu Korea
| | - Jeong-Ho Hong
- Department of Neurology Keimyung University Dongsan Medical Center Daegu Korea
| | - Sang-Hwa Lee
- Department of Neurology Hallym University Chuncheon Sacred Heart Hospital Chuncheon-si Gangwon-do Korea
| | - Man-Seok Park
- Department of Neurology Chonnam National University Hospital, Chonnam National University Medical School Gwangju Korea
| | - Wi-Sun Ryu
- Artificial Intelligence Research Center JLK Inc. Seoul Korea
| | - Kwang-Yeol Park
- Department of Neurology Chung-Ang University College of Medicine, Chung-Ang University Hospital Seoul Korea
| | - Juneyoung Lee
- Department of Biostatistics Korea University College of Medicine Seoul Korea
| | - Jeffrey L Saver
- Department of Neurology and Comprehensive Stroke Center, David Geffen School of Medicine University of California Los Angeles CA
| | - Hee-Joon Bae
- Department of Neurology Seoul National University College of Medicine, Seoul National University Bundang Hospital Seongnam Korea
| |
Collapse
|
25
|
Li X, Li ZF, Wu NQ. Remnant Cholesterol and Residual Risk of Atherosclerotic Cardiovascular Disease. Rev Cardiovasc Med 2025; 26:25985. [PMID: 40026498 PMCID: PMC11868899 DOI: 10.31083/rcm25985] [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: 08/03/2024] [Revised: 09/23/2024] [Accepted: 10/17/2024] [Indexed: 03/05/2025] Open
Abstract
Remnant cholesterol (RC) is increasingly recognized as a key target in the treatment of atherosclerotic cardiovascular disease (ASCVD), addressing much of the residual risk that persists despite standard therapies. However, integrating RC into clinical practice remains challenging. Key issues, such as the development of accessible RC measurement methods, the identification of safe and effective medications, the determination of optimal target levels, and the creation of RC-based risk stratification strategies, require further investigation. This article explores the complex role of RC in ASCVD development, including its definition, metabolic pathways, and its association with both the overall risk and residual risk of ASCVD in primary and secondary prevention. It also examines the effect of current lipid-lowering therapies on RC levels and their influence on cardiovascular outcomes. Recent research has highlighted promising advancements in therapies aimed at lowering RC, which show potential for reducing major adverse cardiovascular events (MACEs). Inhibitors such as angiopoietin-like protein 3 (ANGPTL3), apolipoprotein C-III (apoCIII), and proprotein convertase subtilisin/kexin type 9 (PCSK9) have demonstrated their ability to modulate RC and reduce MACEs by targeting specific proteins involved in RC synthesis and metabolism. There is a pressing need for larger randomized controlled trials to clarify the role of RC in relevant patient populations. The development of targeted RC-lowering therapies holds the promise of significantly reducing the high rates of morbidity and mortality associated with ASCVD.
Collapse
Affiliation(s)
- Xi Li
- Cardiometabolic Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science, 100037 Beijing, China
| | - Zhi-Fan Li
- Cardiometabolic Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science, 100037 Beijing, China
| | - Na-Qiong Wu
- Cardiometabolic Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science, 100037 Beijing, China
| |
Collapse
|
26
|
Kattamuri L, Duggal S, Aparece JP, Sairam S. Cardiovascular Risk Factor and Atherosclerosis in Rheumatoid Arthritis (RA). Curr Cardiol Rep 2025; 27:31. [PMID: 39831939 DOI: 10.1007/s11886-025-02198-8] [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] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE OF REVIEW To highlight advancements in managing traditional and rheumatoid arthritis (RA) specific risk factors and the impact of RA treatments on cardiovascular outcomes. RECENT FINDINGS Advancements in rheumatoid arthritis management have paralleled declining trends in cardiovascular disease risks. Biomarkers like CRP, Lipoprotein(a), Apolipoprotein B 100, and imaging tools such as coronary artery calcium scoring enhance cardiovascular risk stratification, particularly in intermediate-risk RA patients. While effective RA treatments, have demonstrated substantial cardiovascular benefits, subclass differences were noted in high-risk patients. Increased risk of cardiovascular disease is driven by chronic inflammation, altered lipid metabolism, and traditional risk factors. Effective RA treatment significantly lowers cardiovascular events. Standard treatment of hypertension, diabetes and hypercholesterolemia are effective and lowers RA disease activity and inflammatory markers. While RA is considered a risk enhancing state in calculating CV risk scores, currently there exists no RA disease -specific blood pressure, blood sugar or lipid targets.
Collapse
Affiliation(s)
- Lakshmi Kattamuri
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, 4800 Alberta Ave, El Paso, TX, 79935, USA
| | - Shivangini Duggal
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, 4800 Alberta Ave, El Paso, TX, 79935, USA
| | - John Paul Aparece
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, 4800 Alberta Ave, El Paso, TX, 79935, USA
| | - Shrilekha Sairam
- Division of Rheumatology, Department of Internal Medicine, Texas Tech Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, TX, USA.
| |
Collapse
|
27
|
Shi Z, Han S. Personalized statin therapy: Targeting metabolic processes to modulate the therapeutic and adverse effects of statins. Heliyon 2025; 11:e41629. [PMID: 39866414 PMCID: PMC11761934 DOI: 10.1016/j.heliyon.2025.e41629] [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: 09/26/2024] [Revised: 12/31/2024] [Accepted: 01/01/2025] [Indexed: 01/28/2025] Open
Abstract
Statins are widely used for treating lipid disorders and cardiovascular diseases. However, the therapeutic efficiency and adverse effects of statins vary among different patients, which numerous clinical and epidemiological studies have attributed to genetic polymorphisms in statin-metabolizing enzymes and transport proteins. The metabolic processes of statins are relatively complex, involving spontaneous or enzyme-catalyzed interconversion between more toxic lactone metabolites and active acid forms in the liver and bloodstream, influenced by multiple factors, including the expression levels of many metabolic enzymes and transporters. Addressing the variable statin therapeutic outcomes is a pressing clinical challenge. Transcription factors and epigenetic modifications regulate the metabolic enzymes and transporters involved in statin metabolism and disposition and, therefore, hold promise as 'personalized' targets for achieving optimized statin therapy. In this review, we explore the potential for customizing therapy by targeting the metabolism of statin medications. The biochemical bases of adverse reactions to statin drugs and their correlation with polymorphisms in metabolic enzymes and transporters are summarized. Next, we mainly focus on the regulatory roles of transcription factors and epigenetic modifications in regulating the gene expression of statin biochemical machinery. The recommendations for future therapies are finally proposed by targeting the central regulatory factors of statin metabolism.
Collapse
Affiliation(s)
- Zhuangqi Shi
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, 830046, China
| | - Shuxin Han
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, 830046, China
| |
Collapse
|
28
|
Ansari A, Yadav PK, Zhou L, Prakash B, Ijaz L, Christiano A, Ahmad S, Rimbert A, Hussain MM. Casz1 and Znf101/Zfp961 differentially regulate apolipoproteins A1 and B, alter plasma lipoproteins, and reduce atherosclerosis. JCI Insight 2025; 10:e182260. [PMID: 39782688 PMCID: PMC11721306 DOI: 10.1172/jci.insight.182260] [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: 04/19/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
High apolipoprotein B-containing (apoB-containing) low-density lipoproteins (LDLs) and low apoA1-containing high-density lipoproteins (HDLs) are associated with atherosclerotic cardiovascular diseases. In search of a molecular regulator that could simultaneously and reciprocally control both LDL and HDL levels, we screened a microRNA (miR) library using human hepatoma Huh-7 cells. We identified miR-541-3p that both significantly decreases apoB and increases apoA1 expression by inducing mRNA degradation of 2 different transcription factors, Znf101 and Casz1. We found that Znf101 enhances apoB expression, while Casz1 represses apoA1 expression. The hepatic knockdown of Casz1 in mice increased plasma apoA1, HDL, and cholesterol efflux capacity. The hepatic knockdown of Zfp961, an ortholog of Znf101, reduced lipogenesis and production of triglyceride-rich lipoproteins and atherosclerosis, without causing hepatic lipid accumulation. This study identifies hepatic Znf101/Zfp961 and Casz1 as potential therapeutic targets to alter plasma lipoproteins and reduce atherosclerosis without causing liver steatosis.
Collapse
Affiliation(s)
- Abulaish Ansari
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Pradeep Kumar Yadav
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Liye Zhou
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | - Binu Prakash
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Laraib Ijaz
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Amanda Christiano
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Sameer Ahmad
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l’institut du thorax, F-44000 Nantes, France
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
- VA New York Harbor Healthcare System, Brooklyn, New York, USA
| |
Collapse
|
29
|
Li S, Liu HH, Li JJ. Moderate-Intensity Statin Plus Ezetimibe: Time to Rethink it as an Optimal Initial Lipid-Lowering Strategy. Drugs 2025; 85:51-65. [PMID: 39542994 PMCID: PMC11739249 DOI: 10.1007/s40265-024-02113-5] [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] [Accepted: 10/13/2024] [Indexed: 11/17/2024]
Abstract
Achievement of low-density lipoprotein cholesterol (LDL-C) targets is crucial for the prevention of cardiovascular disease (CVD) in individuals with dyslipidaemia who are at high risk. Current guidelines recommend high-intensity statins at the highest tolerated dose as initial treatment to achieve LDL-C goals. However, the real-world situation is dismal: high-intensity statins are underused and achievement of LDL-C goals is suboptimal. Various challenges exist in the implementation of the recommended initial treatment strategy, including hesitancy to use high-intensity statins, non-adherence, and side effects, and the response to high-intensity statins varies across individuals. Emerging studies have shown another line of lipid-lowering, moderate-intensity statins in combination with ezetimibe, presenting considerable efficacy/effectiveness, along with better safety and adherence compared to statin intensification alone. Here we review the clinical evidence, treatment guidelines and challenges associated with high-intensity statins, and summarise the evidence on the combination therapy, moderate-intensity statin plus ezetimibe, which is the core strategy recommended by the 2023 Chinese Guideline for Lipid Management, as a possible primary treatment to achieve the LDL-C targets across several populations. The upfront use of a moderate-intensity statin plus ezetimibe may improve LDL-C control and lead to the prevention of CVD in real-world settings.
Collapse
Affiliation(s)
- Sha Li
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Hui-Hui Liu
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Jian-Jun Li
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China.
| |
Collapse
|
30
|
Eisman AS, Chen ES, Wu WC, Crowley KM, Aluthge DP, Brown K, Sarkar IN. Learning health system linchpins: information exchange and a common data model. J Am Med Inform Assoc 2025; 32:9-19. [PMID: 39538369 DOI: 10.1093/jamia/ocae277] [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: 05/09/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE To demonstrate the potential for a centrally managed health information exchange standardized to a common data model (HIE-CDM) to facilitate semantic data flow needed to support a learning health system (LHS). MATERIALS AND METHODS The Rhode Island Quality Institute operates the Rhode Island (RI) statewide HIE, which aggregates RI health data for more than half of the state's population from 47 data partners. We standardized HIE data to the Observational Medical Outcomes Partnership (OMOP) CDM. Atherosclerotic cardiovascular disease (ASCVD) risk and primary prevention practices were selected to demonstrate LHS semantic data flow from 2013 to 2023. RESULTS We calculated longitudinal 10-year ASCVD risk on 62,999 individuals. Nearly two-thirds had ASCVD risk factors from more than one data partner. This enabled granular tracking of individual ASCVD risk, primary prevention (ie, statin therapy), and incident disease. The population was on statins for fewer than half of the guideline-recommended days. We also found that individuals receiving care at Federally Qualified Health Centers were more likely to have unfavorable ASCVD risk profiles and more likely to be on statins. CDM transformation reduced data heterogeneity through a unified health record that adheres to defined terminologies per OMOP domain. DISCUSSION We demonstrated the potential for an HIE-CDM to enable observational population health research. We also showed how to leverage existing health information technology infrastructure and health data best practices to break down LHS barriers. CONCLUSION HIE-CDM facilitates knowledge curation and health system intervention development at the individual, health system, and population levels.
Collapse
Affiliation(s)
- Aaron S Eisman
- Center for Biomedical Informatics, Brown University, Providence, RI 02912, United States
- The Warren Alpert Medical School, Brown University, Providence, RI 02912, United States
- Yale School of Medicine, New Haven, CT 06510, United States
| | - Elizabeth S Chen
- Center for Biomedical Informatics, Brown University, Providence, RI 02912, United States
- The Warren Alpert Medical School, Brown University, Providence, RI 02912, United States
- School of Public Health, Brown University, Providence, RI 02912, United States
| | - Wen-Chih Wu
- The Warren Alpert Medical School, Brown University, Providence, RI 02912, United States
- School of Public Health, Brown University, Providence, RI 02912, United States
- Division of Cardiology, VA Providence Health Care, Providence, RI 02912, United States
| | - Karen M Crowley
- Center for Biomedical Informatics, Brown University, Providence, RI 02912, United States
| | - Dilum P Aluthge
- Center for Biomedical Informatics, Brown University, Providence, RI 02912, United States
- The Warren Alpert Medical School, Brown University, Providence, RI 02912, United States
| | - Katherine Brown
- Center for Biomedical Informatics, Brown University, Providence, RI 02912, United States
- The Warren Alpert Medical School, Brown University, Providence, RI 02912, United States
| | - Indra Neil Sarkar
- Center for Biomedical Informatics, Brown University, Providence, RI 02912, United States
- The Warren Alpert Medical School, Brown University, Providence, RI 02912, United States
- School of Public Health, Brown University, Providence, RI 02912, United States
- Rhode Island Quality Institute, Providence, RI 02912, United States
| |
Collapse
|
31
|
Shimizu Y, Kawashiri SY, Yamanashi H, Nakamichi S, Hayashida N, Nagata Y, Maeda T. Beneficial influence of low-density lipoprotein cholesterol on the endothelium in relation to endothelial repair. Environ Health Prev Med 2025; 30:24. [PMID: 40189259 PMCID: PMC11986262 DOI: 10.1265/ehpm.24-00332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/20/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDLc) is regarded as a risk factor for endothelial dysfunction. However, LDLc stimulates the proliferation of hematopoietic stem cells (CD34-positive cells), which contribute to endothelial repair. Therefore, LDLc may have a beneficial influence on the endothelium of individuals with lower endothelial repair activity. METHODS This cross-sectional study included 245 men aged 60-69 years. Endothelial repair activity was categorized by the circulating levels of CD34-positive cells based on median values. The status of endothelium was evaluated using the cardio-ankle vascular index (CAVI). RESULTS Among individuals with low levels of circulating CD34-positive cells, LDL-c levels were significantly inversely correlated with CAVI and positively correlated with circulating CD34-positive cells. No significant correlations were observed among the participants with high levels of circulating CD34-positive cells. Among low levels of CD34-positive cells, the adjusted standardized parameter (β) and p value were -0.24 (p = 0.021) for CAVI and 0.41 (p < 0.001) for CD34-positive cells, whereas among high levels of CD34-positive cells, the corresponding values were 0.03 (p = 0.738) and -0.09 (p = 0.355). CONCLUSION LDLc has a beneficial influence on endothelial health among individuals with low endothelial repair activity, possibly by stimulating the proliferation of hematopoietic stem cells.
Collapse
Affiliation(s)
- Yuji Shimizu
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka, Japan
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hirotomo Yamanashi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Seiko Nakamichi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naomi Hayashida
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Division of Strategic Collaborative Research, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Yasuhiro Nagata
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Islands and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
32
|
Modi M, Ndunda P, Modi K. Premature Coronary Artery Disease Presenting as STEMI in a Teenager. J Investig Med High Impact Case Rep 2025; 13:23247096251313985. [PMID: 39861967 PMCID: PMC11760119 DOI: 10.1177/23247096251313985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/21/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
An 18-year-old teenager with significant atherosclerotic cardiovascular disease (ASCVD) risk factors developed acute chest pain. His electrocardiogram showed inferior ST-segment elevations. Emergent coronary angiogram revealed complete thrombotic occlusion of the right coronary artery. He underwent stenting of the culprit lesion with complete clinical recovery and resolution of his electrocardiographic abnormalities secondary to myocardial infarction.
Collapse
|
33
|
Li H, Zhang H, Zhao X, Huang J, Zhang J, Liu Z, Wen J, Qin S. The role of C-reactive protein and genetic predisposition in the risk of psoriasis: results from a national prospective cohort. BMC Rheumatol 2024; 8:72. [PMID: 39707502 DOI: 10.1186/s41927-024-00450-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Psoriasis is an immune-mediated chronic inflammatory disease associated with multiple factors. To evaluate the extent to which C-reactive protein (CRP) and genetic predisposition affect the incidence of psoriasis. METHODS The cohort study retrieved 420,040 participants without psoriasis at baseline from the UK Biobank. Serum CRP was categorized into two levels: < 2 mg/L (normal) and ≥ 2 mg/L (elevated). The polygenic risk score (PRS) was used to estimate genetic predisposition, and was characterized as low, moderate and high PRS. The possible interaction and joint associations between CRP and PRS were assessed using Cox proportional hazards models. RESULTS Participants with high CRP levels had an increased risk of incident psoriasis compared to those with low CRP levels (HR: 1.26, 95% CI: 1.18-1.34). Participants with high CRP levels and high PRS had the highest risk of incident psoriasis [2.24 (95% CI: 2.01, 2.49)], compared with those had low CRP levels and low PRS. Significant additive and multiplicative interaction were found between CRP and PRS in relation to the incidence of psoriasis. CONCLUSIONS Our results suggest that higher CRP concentration may be associated with higher psoriasis incidence, with a more pronounced association observed in individuals with high PRS for psoriasis. So, clinicians should be aware that the risk of incident psoriasis may increase in general population with high CRP levels and high PRS, so that early investigation and intervention can be initiated.
Collapse
Affiliation(s)
- Huarun Li
- Department of Dermatology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Haobin Zhang
- Institute for Healthcare Artificial Intelligence Application, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiangyue Zhao
- Department of Dermatology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinping Huang
- Department of Dermatology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Junguo Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhaoyan Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Ju Wen
- Department of Dermatology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Si Qin
- Department of Dermatology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
34
|
Cheng J, Zhang Z, Wang Y, He H, Zhao T, Wang X. Effect of Statins on the Prognosis After Thoracic Endovascular Aortic Repair for Patients With Acute Type B Aortic Dissection. J Endovasc Ther 2024:15266028241306356. [PMID: 39698950 DOI: 10.1177/15266028241306356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
OBJECTIVE To analyze the clinical efficacy of long-term statin therapy following thoracic endovascular aortic repair (TEVAR) in patients with acute type B aortic dissection (ATBAD). METHODS We retrospectively analyzed data from 645 patients treated between January 2005 and June 2022, dividing them into Statin Group (n=330) and Non-statin Group (n=315) based on whether they received long-term postoperative statin therapy. Patients were further categorized based on median admission low-density lipoprotein cholesterol (LDL-C) levels into High and Low LDL-C Groups to assess the effect of statins on the prognosis of ATBAD patients after TEVAR. RESULTS The cohort had an average age of 53.44±11.42 years old, and 81.71% were male. Statin therapy significantly reduced occurrences of all-cause death (3.03% vs 8.57%, p=0.002) and aorta-related death (0.91% vs 3.81%, p=0.015), particularly in patients with high admission LDL-C levels. In addition, patients with statin therapy had a lower incidence of aorta-related adverse events (ARAE) (4.24% vs 11.11%, p=0.001). Kaplan-Meier analysis indicated statins reduced 5-year cumulative incidence rates of all-cause death and ARAE (all Log-rank p<0.05). These trends were sustained after adjustment. Multivariate Cox analysis confirmed that statin therapy was associated with reduced risks of all-cause and aorta-related deaths, as well as ARAE. CONCLUSION Long-term statin therapy appears to decrease the risk of all-cause and aorta-related death in ATBAD patients after TEVAR, particularly patients with high admission LDL-C levels. Patients with lower LDL-C levels at admission have a reduction of aorta-related death in the follow-up period. Statin therapy also was associated with a lower incidence of ARAE in follow-up. These findings suggest that statins might be crucial in improving long-term outcomes in this patient population. CLINICAL IMPACT Long-term statin therapy administered to patients with acute type B aortic dissection (ATBAD) following thoracic endovascular aortic repair (TEVAR) demonstrates a substantial reduction in both all-cause and aorta-related mortality. Notably, this therapeutic benefit is most evident in patients presenting with elevated low-density lipoprotein cholesterol (LDL-C) levels at admission. Furthermore, statin therapy is associated with a decreased incidence of aorta-related adverse events during follow-up. These findings underscore the pivotal role of statin therapy in enhancing long-term clinical outcomes for ATBAD patients undergoing TEVAR, thereby contributing to improved patient care and prognosis.
Collapse
Affiliation(s)
- Jiaxin Cheng
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Zhiqiang Zhang
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yasong Wang
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Houlin He
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Tinghao Zhao
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaozeng Wang
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
35
|
Mizuta H, Ishii M, Ikebe S, Otsuka Y, Yamanouchi Y, Nakamura T, Tsujita K. Triglycerides and the Risk of Atherosclerotic Cardiovascular Events Across Different Risk Categories. J Atheroscler Thromb 2024:65334. [PMID: 39675972 DOI: 10.5551/jat.65334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024] Open
Abstract
AIMS To investigate the association between triglyceride levels and major adverse cardiovascular events (MACE) in primary and secondary prevention cohorts. METHODS This retrospective study was conducted with a nationwide health insurance claims database, which included approximately 3.8 million participants with medical checkups between January 2005 and August 2020 in Japan. The participants were classified into primary prevention (n=3,415,522) and secondary prevention (n=29,806) cohorts based on cardiovascular or cerebrovascular disease history. Each participant was categorized as having very low (triglyceride <50 mg/dL), low normal (50-99), high normal (100-149), or hypertriglyceridemia (≥ 150). The primary prevention cohort was further stratified into low-, intermediate-, and high-risk groups according to atherosclerotic cardiovascular diseases risk. Outcome was MACE, including acute myocardial infarction (AMI), unstable angina, ischemic stroke, and cardiac death. RESULTS Over a mean follow-up of 3.25 years, 0.3% and 2.6% MACE occurred in primary and secondary prevention, respectively. Hypertriglyceridemia was associated with high risk of MACE in the primary prevention, but not in the secondary prevention. A significant interaction was observed between prevention categories and the association of TG levels with MACE in those with TG <150 mg/dL and ischemic stroke in those with TG ≥ 150 mg/dL. The population-attributable fraction for hypertriglyceridemia in primary prevention was 4.1% for MACE. In primary prevention, lower risks of AMI were observed in the lower TG category compared to the current threshold. CONCLUSIONS This study suggests distinct triglyceride thresholds for MACE risk in primary and secondary prevention cohorts, requiring further prospective validation for clinical implementation.
Collapse
Affiliation(s)
- Hiroyuki Mizuta
- Tokushukai Isen Clinic
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
- Department of Medical Information Science, Graduate School of Medical Sciences, Kumamoto University
| | - So Ikebe
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
| | - Yasuhiro Otsuka
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
| | | | - Taishi Nakamura
- Department of Medical Information Science, Graduate School of Medical Sciences, Kumamoto University
- Department of Clinical Investigation, Kumamoto University Hospital
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
- Department of Clinical Investigation, Kumamoto University Hospital
| |
Collapse
|
36
|
Okada K, Haze T, Kikuchi S, Kirigaya H, Hanajima Y, Tsutsumi K, Kirigaya J, Nakahashi H, Gohbara M, Kimura Y, Kosuge M, Ebina T, Sugano T, Hibi K. Early, Intensive and Persistent Lipid-Lowering Therapy for Secondary Prevention of Acute Coronary Syndrome. J Atheroscler Thromb 2024; 31:1748-1762. [PMID: 38880605 PMCID: PMC11620828 DOI: 10.5551/jat.64988] [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: 03/23/2024] [Accepted: 05/01/2024] [Indexed: 06/18/2024] Open
Abstract
AIM Early and intensive low-density lipoprotein (LDL-C)-lowering therapy plays important roles in secondary prevention of acute coronary syndrome (ACS), but the treatment period for further clinical benefit remains undefined. This single-center, retrospective study explored LDL-C trajectory after ACS and its associations with subsequent cardiovascular events (CVE). METHODS In 831 patients with ACS, we evaluated LDL-C reduction during the first 2 months post-ACS as an index of early intervention and the area over the curve for LDL-C using 70 mg/dl as the threshold in the next 6 months (AOC-70) as a persistent intensity index. Patients were followed for a median of 3.0 (1.1-5.2) years for CVE, defined as the composite of cardiovascular death, non-fatal myocardial infarction, angina pectoris requiring revascularization, cerebral infarction, and coronary bypass grafting. RESULTS LDL-C decreased from baseline to 2 months post-ACS (107±38 mg/dl to 78±25 mg/dl, p<0.001) through high-intensity statin prescription (91.8%), while achieving rates of LDL-C <70 mg/dl at 2 months remained only 40.2% with no significant changes thereafter. During the follow-up period, CVE occurred in 200 patients. LDL-C reduction during the first 2 months and AOC-70 in the next 6 months were both associated with subsequent CVE risk (sub-HR [hazard ratio] [95% confidence interval]: 1.48 [1.16-1.89] and 1.22 [1.05-1.44]). Furthermore, early intervention followed by persistently intensive LDL-C-lowering therapy resulted in further CVE risk reduction. CONCLUSIONS The present study observed that achieving early and intensive LDL-C reduction within the first two months after ACS and maintaining it for the next six months suppressed subsequent CVE risk, suggesting the importance of early, intensive, and persistent LDL-C-lowering therapy in the secondary prevention of ACS.
Collapse
Affiliation(s)
- Kozo Okada
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
- YCU Center for Novel and Exploratory Clinical Trials (Y-NEXT), Yokohama City University Hospital, Yokohama, Japan
| | - Tatsuya Haze
- Department of Nephrology and Hypertension, Yokohama City University Medical Center, Yokohama, Japan
- YCU Center for Novel and Exploratory Clinical Trials (Y-NEXT), Yokohama City University Hospital, Yokohama, Japan
| | - Shinnosuke Kikuchi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Hidekuni Kirigaya
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Yohei Hanajima
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Katsuhiko Tsutsumi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Jin Kirigaya
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Hidefumi Nakahashi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Masaomi Gohbara
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Yuichiro Kimura
- 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
| | - Teruyasu Sugano
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| |
Collapse
|
37
|
Lolekha P, Khovidhunkit W, Deerochanawong C, Thongtang N, Boonyasirinant T, Rattarasarn C, Chutinet A, Ophascharoensuk V, Somlaw N, Sitthisook S, Suntorntham S, Nitiyanant W, Krittayaphong R. 2024 The Royal College of Physicians of Thailand (RCPT) clinical practice guidelines on management of dyslipidemia for atherosclerotic cardiovascular disease prevention. ASIAN BIOMED 2024; 18:246-267. [PMID: 39697215 PMCID: PMC11650434 DOI: 10.2478/abm-2024-0033] [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] [Indexed: 12/20/2024]
Abstract
Background The Royal College of Physicians of Thailand (RCPT) published a Clinical Practice Guideline on Pharmacologic Therapy of Dyslipidemia for Atherosclerotic Cardiovascular Disease (ASCVD) Prevention in 2016. The availability of newer classes of medications for dyslipidemia, supported by extensive clinical research findings, indicates a significant need for the updating of the existing clinical practice guideline. Objectives To serve as guidelines on the management of dyslipidemia for Thai adults. Methods The RCPT Dyslipidemia Guidelines Committee was established with representatives from selected professional societies to revise the 2016 Guideline by critically reviewing the latest evidence. Meetings were conducted from August to December 2023, culminating in a public hearing that engaged various stakeholders in January 2024. The final Thai version received approval in April 2024, while the English translation was completed in October 2024. Results Lifestyle modifications and statins remain the cornerstone of therapy for dyslipidemia in adults across various clinical settings. Emerging evidence regarding newer classes of lipid-lowering medications indicates that these treatments are effective in lowering LDL-cholesterol levels and reducing atherosclerotic cardiovascular events. This suggests that they may serve as an add-on therapy for individuals who cannot achieve target levels or who are at high risk for future cardiovascular events. The Thai CV Risk Score is recommended due to its specificity for the Thai population. Conclusions The 2024 updated clinical practice guidelines establish a framework, provide recommendations, and serve as a comprehensive resource for the contemporary management of dyslipidemia in adults, with the goal of preventing ASCVD in Thailand.
Collapse
Affiliation(s)
- Praween Lolekha
- Department of Medicine, Thammasat University, Patrhumthani12121, Thailand
| | - Weerapan Khovidhunkit
- Department of Medicine, King Chulalongkorn Memorial Hospital and Chulalongkorn University, Bangkok10330, Thailand
| | | | - Nuntakorn Thongtang
- Department of Medicine, Siriraj Hospital, Mahidol University, Bangkok10700, Thailand
| | | | - Chatchalit Rattarasarn
- Department of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok10400, Thailand
| | - Aurauma Chutinet
- Department of Medicine, King Chulalongkorn Memorial Hospital and Chulalongkorn University, Bangkok10330, Thailand
| | | | - Nicha Somlaw
- Department of Medicine, King Chulalongkorn Memorial Hospital and Chulalongkorn University, Bangkok10330, Thailand
| | - Surapun Sitthisook
- Department of Medicine, King Chulalongkorn Memorial Hospital and Chulalongkorn University, Bangkok10330, Thailand
| | | | - Wannee Nitiyanant
- Department of Medicine, Siriraj Hospital, Mahidol University, Bangkok10700, Thailand
| | - Rungroj Krittayaphong
- Department of Medicine, Siriraj Hospital, Mahidol University, Bangkok10700, Thailand
| |
Collapse
|
38
|
Rose SJ, Hartnett J, Estep ZJ, Ameen D, Karki S, Schuster E, Newman RB, Hsi DH. Measurement of breast artery calcification using an artificial intelligence detection model and its association with major adverse cardiovascular events. PLOS DIGITAL HEALTH 2024; 3:e0000698. [PMID: 39715147 DOI: 10.1371/journal.pdig.0000698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/10/2024] [Indexed: 12/25/2024]
Abstract
Breast artery calcification (BAC) obtained from standard mammographic images is currently under evaluation to stratify risk of major adverse cardiovascular events in women. Measuring BAC using artificial intelligence (AI) technology, we aimed to determine the relationship between BAC and coronary artery calcification (CAC) severity with Major Adverse Cardiac Events (MACE). This retrospective study included women who underwent chest computed tomography (CT) within one year of mammography. T-test assessed the associations between MACE and variables of interest (BAC versus MACE, CAC versus MACE). Risk differences were calculated to capture the difference in observed risk and reference groups. Chi-square tests and/or Fisher's exact tests were performed to assess age and ASCVD risk with MACE and to assess BAC and CAC association with atherosclerotic cardiovascular disease (ASCVD) risk as a secondary outcome. A logistic regression model was conducted to measure the odds ratio between explanatory variables (BAC and CAC) and the outcome variables (MACE). Out of the 99 patients included in the analysis, 49 patients (49.49%) were BAC positive, with 37 patients (37.37%) CAC positive, and 26 patients (26.26%) had MACE. One unit increase in BAC score resulted in a 6% increased odds of having a moderate to high ASCVD risk >7.5% (p = 0.01) and 2% increased odds of having MACE (p = 0.005). The odds of having a moderate-high ASCVD risk score in BAC positive patients was higher (OR = 4.27, 95% CI 1.58-11.56) than CAC positive (OR = 4.05, 95% CI 1.36-12.06) patients. In this study population, the presence of BAC is associated with MACE and useful in corroborating ASCVD risk. Our results provide evidence to support the potential utilization of AI generated BAC measurements from standard of care mammograms in addition to the widely adopted ASCVD and CAC scores, to identify and risk-stratify women who are at increased risk of CVD and may benefit from targeted prevention measures.
Collapse
Affiliation(s)
- Suzanne J Rose
- Department of Research and Discovery, Stamford Hospital, Stamford, Connecticut, United States of America
| | | | - Zachary J Estep
- Sidney Kimmel Medical College at Thomas Jefferson University/Deborah Heart and Lung Center, Browns Mills, New Jersey, United States of America
| | - Daniyal Ameen
- Department of Internal Medicine, Yale New Haven Health/Bridgeport Hospital, Bridgeport, Connecticut, United States of America
| | - Shweta Karki
- Department of Research and Discovery, Stamford Hospital, Stamford, Connecticut, United States of America
| | - Edward Schuster
- Stamford Health Medical Group, Stamford, Connecticut, United States of America
| | - Rebecca B Newman
- Department of Internal Medicine, Stamford Hospital, Stamford, Connecticut, United States of America
| | - David H Hsi
- Heart and Vascular Institute, Stamford Hospital, Stamford, Connecticut, United States of America
| |
Collapse
|
39
|
Jiang JC, Singh K, Nitin R, Davis LK, Wray NR, Shah S. Sex-Specific Association Between Genetic Risk of Psychiatric Disorders and Cardiovascular Diseases. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004685. [PMID: 39611256 PMCID: PMC11651350 DOI: 10.1161/circgen.124.004685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/15/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Though epidemiological studies show increased cardiovascular disease (CVD) risks among individuals with psychiatric disorders, findings on sex differences in comorbidity have been inconsistent. METHODS This genetic epidemiology study examined the sex-specific association between the genetic risk of 3 psychiatric disorders (major depression [MD], schizophrenia, and bipolar disorder), estimated using polygenic scores (PGSs), and risks of 3 CVDs (atrial fibrillation [AF], coronary artery disease [CAD], and heart failure [HF]) in 345 169 European-ancestry individuals (UK Biobank), with analyses replicated in an independent BioVU cohort (n=49 057). Mediation analysis was conducted to determine whether traditional CVD risk factors could explain any observed sex difference. RESULTS In the UK Biobank, a 1-SD increase in PGSMD was significantly associated with the incident risks of all 3 CVDs in females after multiple testing corrections (hazard ratio [HR]AF-female=1.04 [95% CI, 1.02-1.06]; P=1.5×10-4; HRCAD-female=1.07 [95% CI, 1.04-1.11]; P=2.6×10-6; and HRHF-female=1.09 [95% CI, 1.06-1.13]; P=9.7×10-10), but not in males. These female-specific associations remained even in the absence of any psychiatric disorder diagnosis or psychiatric medication use. Although mediation analysis demonstrated that the association between PGSMD and CVDs in females was partly mediated by baseline body mass index, hypercholesterolemia, hypertension, and smoking, these risk factors did not explain the higher risk compared with males. The association between PGSMD and CAD was consistent between females who were premenopausal and postmenopausal at baseline, while the association with AF and HF was only observed in the baseline postmenopausal cohort. No significant association with CVD risks was observed for the PGS of schizophrenia or bipolar disorder. The female-specific positive association of PGSMD with CAD risk was replicated in BioVU. CONCLUSIONS Genetic predisposition to MD confers a greater risk of CVDs in females versus males, even in the absence of any depression diagnosis. This study warrants further investigation into whether genetic predisposition to depression could be useful for improving cardiovascular risk prediction, especially in women.
Collapse
Affiliation(s)
- Jiayue-Clara Jiang
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia (J.-C.J., N.R.W., S.S.)
| | - Kritika Singh
- Division of Genetic Medicine, Department of Medicine (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Genetics Institute (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Rachana Nitin
- Division of Genetic Medicine, Department of Medicine (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Genetics Institute (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Lea K. Davis
- Division of Genetic Medicine, Department of Medicine (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Genetics Institute (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics (L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Department of Psychiatry and Behavioral Sciences (L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Departments of Medicine and Biomedical Informatics (L.K.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Naomi R. Wray
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia (J.-C.J., N.R.W., S.S.)
- Department of Psychiatry, University of Oxford, Warneford Hospital, United Kingdom (N.R.W.)
| | - Sonia Shah
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia (J.-C.J., N.R.W., S.S.)
| |
Collapse
|
40
|
Rajalingamgari P, Khatua B, Summers MJ, Kostenko S, Chang YHH, Elmallahy M, Anand A, Narayana Pillai A, Morsy M, Trivedi S, McFayden B, Jahangir S, Snozek CL, Singh VP. Prospective observational study and mechanistic evidence showing lipolysis of circulating triglycerides worsens hypertriglyceridemic acute pancreatitis. J Clin Invest 2024; 135:e184785. [PMID: 39509346 DOI: 10.1172/jci184785] [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: 07/12/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUNDWhile most hypertriglyceridemia is asymptomatic, hypertriglyceridemia-associated acute pancreatitis (HTG-AP) can be more severe than AP of other etiologies. The reasons underlying this are unclear. We thus examined whether lipolytic generation of nonesterified fatty acids (NEFAs) from circulating triglycerides (TGs) could worsen clinical outcomes.METHODSAdmission serum TGs, NEFA composition, and concentrations were analyzed prospectively for 269 patients with AP. These parameters, demographics, and clinical outcomes were compared between HTG-AP (TGs >500 mg/dL; American Heart Association [AHA] 2018 guidelines) and AP of other etiologies. Serum NEFAs were correlated with serum TG fatty acids (TGFAs) alone and with the product of TGFA serum lipase (NEFAs - TGFAs × lipase). Studies in mice and rats were conducted to understand the role of HTG lipolysis in organ failure and to interpret the NEFA-TGFA correlations.RESULTSPatients with HTG-AP had higher serum NEFA and TG levels and more severe AP (19% vs. 7%; P < 0.03) than did individuals with AP of other etiologies. Correlations of long-chain unsaturated NEFAs with corresponding TGFAs increased with TG concentrations up to 500 mg/dL and declined thereafter. However, NEFA - TGFA × lipase correlations became stronger with TGs above 500 mg/dL. AP and intravenous lipase infusion in rodents caused lipolysis of circulating TGs to NEFAs. This led to multisystem organ failure, which was prevented by pancreatic TG lipase deletion or lipase inhibition.CONCLUSIONSHTG-AP is made severe by the NEFAs generated from intravascular lipolysis of circulating TGs. Strategies that prevent TG lipolysis may be effective in improving clinical outcomes for patients with HTG-AP.FUNDINGNational Institute of Diabetes and Digestive and Kidney Diseases (NIDDK, NIH) (RO1DK092460 and R01DK119646); Department of Defense (PR191945 under W81XWH-20-1-0400); National Institute on Alcohol Abuse and Alcoholism (NIAAA), NIH (R01AA031257).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Vijay P Singh
- Department of Medicine
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Arizona, USA
| |
Collapse
|
41
|
Shah NP, Mulder H, Lydon E, Chiswell K, Hu X, Lampron Z, Cohen L, Patel MR, Taubes S, Song W, Mulukutla SR, Saeed A, Morin DP, Bradley SM, Hernandez AF, Pagidipati NJ. Lipoprotein (a) Testing in Patients With Atherosclerotic Cardiovascular Disease in 5 Large US Health Systems. J Am Heart Assoc 2024; 13:e035610. [PMID: 39494552 PMCID: PMC11935684 DOI: 10.1161/jaha.124.035610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/12/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Lipoprotein (a) is an independent risk factor for atherosclerotic cardiovascular disease. However, lipoprotein (a) testing remains variable and it is unclear what factors influence testing and if testing changes clinical management. METHODS AND RESULTS A retrospective study using electronic medical record data from 5 health systems identified an atherosclerotic cardiovascular disease cohort divided into those with and without a lipoprotein (a) test between 2019 and 2021. Baseline characteristics and lipid-lowering therapy patterns were assessed. Multivariable regression modeling was used to determine factors associated with lipoprotein (a) testing. Among 595 684 patients with atherosclerotic cardiovascular disease, only 2587 (0.4%) were tested for lipoprotein (a). Those who were older or Black individuals were less likely to have lipoprotein (a) testing, while those with familial hypercholesterolemia, ischemic stroke/transient ischemic attack, peripheral artery disease, prior lipid-lowering therapy, or low-density lipoprotein cholesterol ≥130 mg/dL were more likely to be tested. Those with a lipoprotein (a) test, regardless of the lipoprotein (a) value, were more frequently initiated on any statin therapy (30.3% versus 10.6%, P < 0.001), ezetimibe (7.65% versus 0.8%, P < 0.001), or proprotein convertase substilisin/kexin type 9 inhibitor (6.7% versus 0.3%, P < 0.001) compared with those without a test. Those with an elevated lipoprotein (a) level more frequently initiated ezetimibe (11.5% versus 5.9%, P < 0.001) or proprotein convertase substilisin/kexin type 9 inhibitor (10.9% versus 4.8%, P < 0.001). CONCLUSIONS Lipoprotein (a) testing in patients with atherosclerotic cardiovascular disease is infrequent, with evidence of disparities among older or Black individuals. Testing for lipoprotein (a), regardless of level, is associated with greater initiation of any lipid-lowering therapy, while elevated lipoprotein (a) is associated with greater initiation of nonstatin lipid-lowering therapy. There is a critical need for multidisciplinary and inclusive approaches to raise awareness for lipoprotein (a) testing, and its implications on management.
Collapse
Affiliation(s)
- Nishant P. Shah
- Division of CardiologyDuke University HospitalDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| | | | | | | | - Xingdi Hu
- Novartis Pharmaceuticals CorporationEast HanoverNJUSA
| | | | | | - Manesh R. Patel
- Division of CardiologyDuke University HospitalDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| | - Susan Taubes
- Novartis Pharmaceuticals CorporationEast HanoverNJUSA
| | - Wenliang Song
- Division of CardiologyVanderbilt UniversityNashvilleTNUSA
| | | | - Anum Saeed
- Division of CardiologyUniversity of PittsburghPittsburghPAUSA
| | - Daniel P. Morin
- Division of CardiologyOchsner Medical CenterNew OrleansLAUSA
| | | | - Adrian F. Hernandez
- Division of CardiologyDuke University HospitalDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| | - Neha J. Pagidipati
- Division of CardiologyDuke University HospitalDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| |
Collapse
|
42
|
Thompson A, Fleischmann KE, Smilowitz NR, de Las Fuentes L, Mukherjee D, Aggarwal NR, Ahmad FS, Allen RB, Altin SE, Auerbach A, Berger JS, Chow B, Dakik HA, Eisenstein EL, Gerhard-Herman M, Ghadimi K, Kachulis B, Leclerc J, Lee CS, Macaulay TE, Mates G, Merli GJ, Parwani P, Poole JE, Rich MW, Ruetzler K, Stain SC, Sweitzer B, Talbot AW, Vallabhajosyula S, Whittle J, Williams KA. 2024 AHA/ACC/ACS/ASNC/HRS/SCA/SCCT/SCMR/SVM Guideline for Perioperative Cardiovascular Management for Noncardiac Surgery: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation 2024; 150:e351-e442. [PMID: 39316661 DOI: 10.1161/cir.0000000000001285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
AIM The "2024 AHA/ACC/ACS/ASNC/HRS/SCA/SCCT/SCMR/SVM Guideline for Perioperative Cardiovascular Management for Noncardiac Surgery" provides recommendations to guide clinicians in the perioperative cardiovascular evaluation and management of adult patients undergoing noncardiac surgery. METHODS A comprehensive literature search was conducted from August 2022 to March 2023 to identify clinical studies, reviews, and other evidence conducted on human subjects that were published in English from MEDLINE (through PubMed), EMBASE, the Cochrane Library, the Agency for Healthcare Research and Quality, and other selected databases relevant to this guideline. STRUCTURE Recommendations from the "2014 ACC/AHA Guideline on Perioperative Cardiovascular Evaluation and Management of Patients Undergoing Noncardiac Surgery" have been updated with new evidence consolidated to guide clinicians; clinicians should be advised this guideline supersedes the previously published 2014 guideline. In addition, evidence-based management strategies, including pharmacological therapies, perioperative monitoring, and devices, for cardiovascular disease and associated medical conditions, have been developed.
Collapse
Affiliation(s)
| | | | | | - Lisa de Las Fuentes
- Former ACC/AHA Joint Committee on Clinical Practice Guidelines member; current member during the writing effort
| | | | | | | | | | | | | | | | - Benjamin Chow
- Society of Cardiovascular Computed Tomography representative
| | | | | | | | | | | | | | | | | | | | | | - Purvi Parwani
- Society for Cardiovascular Magnetic Resonance representative
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Gazit T, Mann H, Gaber S, Adamenko P, Pariente G, Volsky L, Dolev A, Lyson H, Zimlichman E, Pandit JA, Paz E. A novel, machine-learning model for prediction of short-term ASCVD risk over 90 and 365 days. Front Digit Health 2024; 6:1485508. [PMID: 39552935 PMCID: PMC11564171 DOI: 10.3389/fdgth.2024.1485508] [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: 08/23/2024] [Accepted: 10/11/2024] [Indexed: 11/19/2024] Open
Abstract
Background Current atherosclerotic cardiovascular disease (ASCVD) risk assessment tools like the Pooled Cohort Equations (PCEs) and PREVENT™ scores offer long-term predictions but may not effectively drive behavior change. Short-term risk predictions using mobile health (mHealth) data and electronic health records (EHRs) could enhance clinical decision-making and patient engagement. The aim of this study was to develop a short-term ASCVD risk prediction model for hypertensive individuals using mHealth and EHR data and compare its performance to existing risk assessment tools. Methods This is a retrospective cohort study including 51,127 hypertensive participants aged ≥18 years old who enrolled in the Hello Heart CV risk self-management program between January 2015 and January 2024. A machine learning (ML) model was derived from EHR data and mHealth measurements of blood pressure (BP) and heart rate (HR) collected via at-home BP monitors. Its performance was compared to that of PCE and PREVENT. Results The XgBoost model incorporating 291 features outperformed the PCE and PREVENT scores in discriminating ASCVD risk for both prediction periods. For 90-day prediction, mean C-statistics were 0.81 (XgBoost) vs. 0.74 (PCE) and 0.65 (PREVENT). Similar findings were observed for 365-day prediction. mHealth measurements incrementally enhanced 365-day risk prediction (ROC-AUC 0.82 vs. 0.80 without mHealth). Conclusion An EHR and mHealth-based ML model offers superior short-term ASCVD prediction compared to traditional tools. This approach supports personalized preventive strategies, particularly for populations with incomplete features for PCE or PREVENT. Further research should explore this novel risk prediction framework, and particularly additional mHealth data integration for broader applicability and increased predictive power.
Collapse
Affiliation(s)
- Tomer Gazit
- Hello Heart, Inc., Menlo Park, CA, United States
| | - Hanan Mann
- Hello Heart, Inc., Menlo Park, CA, United States
| | - Shiri Gaber
- Hello Heart, Inc., Menlo Park, CA, United States
| | | | | | - Liron Volsky
- Hello Heart, Inc., Menlo Park, CA, United States
| | - Amir Dolev
- Hello Heart, Inc., Menlo Park, CA, United States
| | - Helena Lyson
- Hello Heart, Inc., Menlo Park, CA, United States
| | | | - Jay A. Pandit
- Scripps Research Translational Institute, La Jolla, CA, United States
| | - Edo Paz
- Hello Heart, Inc., Menlo Park, CA, United States
| |
Collapse
|
44
|
Hilser JR, Spencer NJ, Afshari K, Gilliland FD, Hu H, Deb A, Lusis AJ, Wilson Tang W, Hartiala JA, Hazen SL, Allayee H. COVID-19 Is a Coronary Artery Disease Risk Equivalent and Exhibits a Genetic Interaction With ABO Blood Type. Arterioscler Thromb Vasc Biol 2024; 44:2321-2333. [PMID: 39381876 PMCID: PMC11495539 DOI: 10.1161/atvbaha.124.321001] [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: 03/25/2024] [Accepted: 08/08/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND COVID-19 is associated with acute risk of major adverse cardiac events (MACE), including myocardial infarction, stroke, and mortality (all-cause). However, the duration and underlying determinants of heightened risk of cardiovascular disease and MACE post-COVID-19 are not known. METHODS Data from the UK Biobank was used to identify COVID-19 cases (n=10 005) who were positive for polymerase chain reaction (PCR+)-based tests for SARS-CoV-2 infection (n=8062) or received hospital-based International Classification of Diseases version-10 (ICD-10) codes for COVID-19 (n=1943) between February 1, 2020 and December 31, 2020. Population controls (n=217 730) and propensity score-matched controls (n=38 860) were also drawn from the UK Biobank during the same period. Proportional hazard models were used to evaluate COVID-19 for association with long-term (>1000 days) risk of MACE and as a coronary artery disease risk equivalent. Additional analyses examined whether COVID-19 interacted with genetic determinants to affect the risk of MACE and its components. RESULTS The risk of MACE was elevated in COVID-19 cases at all levels of severity (HR, 2.09 [95% CI, 1.94-2.25]; P<0.0005) and to a greater extent in cases hospitalized for COVID-19 (HR, 3.85 [95% CI, 3.51-4.24]; P<0.0005). Hospitalization for COVID-19 represented a coronary artery disease risk equivalent since incident MACE risk among cases without history of cardiovascular disease was even higher than that observed in patients with cardiovascular disease without COVID-19 (HR, 1.21 [95% CI, 1.08-1.37]; P<0.005). A significant genetic interaction was observed between the ABO locus and hospitalization for COVID-19 (Pinteraction=0.01), with risk of thrombotic events being increased in subjects with non-O blood types (HR, 1.65 [95% CI, 1.29-2.09]; P=4.8×10-5) to a greater extent than subjects with blood type O (HR, 0.96 [95% CI, 0.66-1.39]; P=0.82). CONCLUSIONS Hospitalization for COVID-19 represents a coronary artery disease risk equivalent, with post-acute myocardial infarction and stroke risk particularly heightened in non-O blood types. These results may have important clinical implications and represent, to our knowledge, one of the first examples of a gene-pathogen exposure interaction for thrombotic events.
Collapse
Affiliation(s)
- James R. Hilser
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Biochemistry and Molecular Medicine (J.R.H., N.J.S., K.A., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Neal J. Spencer
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Biochemistry and Molecular Medicine (J.R.H., N.J.S., K.A., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Kimia Afshari
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Biochemistry and Molecular Medicine (J.R.H., N.J.S., K.A., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Frank D. Gilliland
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Howard Hu
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Arjun Deb
- Department of Medicine (A.D., A.J.L.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Aldons J. Lusis
- Department of Medicine (A.D., A.J.L.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Microbiology, Immunology, and Molecular Genetics (A.J.L.), David Geffen School of Medicine of UCLA, CA
- Department of Human Genetics (A.J.L.), David Geffen School of Medicine of UCLA, CA
| | - W.H. Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome and Human Health (W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Jaana A. Hartiala
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome and Human Health (W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Hooman Allayee
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Biochemistry and Molecular Medicine (J.R.H., N.J.S., K.A., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| |
Collapse
|
45
|
de Kermenguy C, Durand A, Tollenaere Q, Le Pabic E, Paillard F, Mahé G. A retrospective analysis on optimal medical therapy for patients with symptomatic lower extremity peripheral artery disease: a French observational study. BMC Cardiovasc Disord 2024; 24:611. [PMID: 39482624 PMCID: PMC11529423 DOI: 10.1186/s12872-024-04289-w] [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: 11/17/2023] [Accepted: 10/23/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE Patients with symptomatic lower extremity artery disease (LEAD) should have an optimal management in terms of lipid goal [i.e. controlled LDL-cholesterol (LDLc)] and medical treatment (triple therapy with an antiplatelet agent, a statin and an angiotensin-converting enzyme inhibitor or a angiotensin-receptor antagonist). Prevalence of LEAD patients with a LDLc < 0.55 g/l is unknown. Aims of this study were to: (i) describe the prevalence of patients with a LDLc < 0.55 g/l, (ii) describe the prevalence of patients with an optimal medical treatment; (iii) compare this management between patients with a vascular surgery history and those without a vascular surgery history; and (iv) evaluate the number of patients eligible for new lipid-lowering therapies according to FOURIER and REDUCE-IT criteria. METHODS In this single-center retrospective study, prevalence is expressed as numbers and percentages. Comparison of the number of well managed patients between LEAD patients with a vascular surgery history and those without was performed. Number of patients who would be eligible for FOURIER and REDUCE-IT studies were calculated. RESULTS Among the LEAD patients included in the analysis (n = 225), only 12.4% (n = 28) had a LDLc < 0.55 g/L. The prevalence of patients who received the optimal medical treatment was 50.7% (n = 114). There was no statistical difference in the prevalence of patients with and without vascular surgery history achieving the LDLc goal (n = 9 (10.6%) vs. n = 19 (13.6%); p = not significant). Ninety-three patients (46.0%) would be eligible for EVOLOCUMAB treatment according to the Fourier study design whereas 17 patients (8.4%) would be eligible for treatment with ICOSAPENT ETHYL according to the REDUCE-IT study design. CONCLUSION A majority of LEAD patients did not reach the LDLc goals. LEAD patients with a vascular surgery history did not experience a better management whereas they had a more consistent follow-up.
Collapse
Affiliation(s)
- Camille de Kermenguy
- Vascular Medicine Unit, CHU Rennes, 2 Rue Henri Le Guilloux, Rennes, 35033, France
- CHU Rennes, Inserm, CIC 1414 (Clinical Investigation Center), Rennes, F-35000, France
| | - Anne Durand
- Vascular Medicine Unit, CHU Rennes, 2 Rue Henri Le Guilloux, Rennes, 35033, France
- CHU Rennes, Inserm, CIC 1414 (Clinical Investigation Center), Rennes, F-35000, France
| | - Quentin Tollenaere
- Vascular Medicine Unit, CHU Rennes, 2 Rue Henri Le Guilloux, Rennes, 35033, France
- CHU Rennes, Inserm, CIC 1414 (Clinical Investigation Center), Rennes, F-35000, France
| | - Estelle Le Pabic
- CHU Rennes, Inserm, CIC 1414 (Clinical Investigation Center), Rennes, F-35000, France
| | | | - Guillaume Mahé
- Vascular Medicine Unit, CHU Rennes, 2 Rue Henri Le Guilloux, Rennes, 35033, France.
- Univ Rennes, M2S - EA 7470, F-35000, Rennes, France.
- CHU Rennes, Inserm, CIC 1414 (Clinical Investigation Center), Rennes, F-35000, France.
- Univ Rennes, Rennes, France.
| |
Collapse
|
46
|
Akiyama T, Ikegami R, Kubota N, Takano T, Yoneyama S, Okubo T, Hoyano M, Ozaki K, Inomata T. Serum Apolipoprotein-A2 Levels Are a Strong Predictor of Future Cardiovascular Events in Patients Undergoing Percutaneous Coronary Intervention. Circ J 2024; 88:1770-1777. [PMID: 38897974 DOI: 10.1253/circj.cj-24-0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
BACKGROUND Because apolipoprotein-A2 (ApoA2), a key component of high-density lipoprotein cholesterol (HDL-C), lacks clear clinical significance, we investigated its impact on cardiovascular events in patients undergoing percutaneous coronary intervention (PCI). METHODS AND RESULTS We examined 638 patients who underwent PCI with a new-generation drug-eluting stent for acute or chronic coronary syndrome and had their apolipoprotein levels measured between 2016 and 2021. The patients were divided into 2 groups based on the median serum ApoA2 values, and the incidence of major adverse cardiovascular events (MACE) was assessed. Of the 638 patients, 563 (88%) received statin treatment, with a median serum LDL-C level of 93 mg/dL. Furthermore, 137 patients (21.5%) experienced MACE, and Kaplan-Meier analysis revealed that the higher ApoA2 group had a significantly lower incidence of MACE than the lower ApoA2 group (30.9% vs. 41.6%). However, the other apolipoproteins, including ApoA1, ApoB, ApoC2, ApoC3, and ApoE, showed no significant differences in MACE. Multivariable Cox hazard analysis indicated that ApoA2 was an independent predictor of MACEs (hazard ratio, 0.666; 95% confidence interval, 0.465-0.954). Furthermore, ApoA2 levels exhibited the strongest inverse association with high-sensitivity C-reactive protein levels (rs=-0.479). CONCLUSIONS Among all the apolipoproteins, the serum ApoA2 level may be the strongest predictor of future cardiovascular events and prognosis in patients undergoing PCI.
Collapse
Affiliation(s)
- Takumi Akiyama
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences
| | - Ryutaro Ikegami
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences
| | - Naoki Kubota
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences
| | - Toshiki Takano
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences
| | - Shintaro Yoneyama
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences
| | - Takeshi Okubo
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences
| | - Makoto Hoyano
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences
| | - Kazuyuki Ozaki
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences
| | - Takayuki Inomata
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences
| |
Collapse
|
47
|
Wang Y, Li H, Zhang Z, Wu F, Liu J, Zhu Z, Xiang H. The association between vitamin E intake and remnant cholesterol, total cholesterol, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol in US adults: a cross-sectional study. Lipids Health Dis 2024; 23:325. [PMID: 39354564 PMCID: PMC11443817 DOI: 10.1186/s12944-024-02313-8] [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: 08/08/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Blood lipid profiles are associated with various nutritional elements and dietary factors. This study aimed to explore the association between total dietary vitamin E intake and remnant cholesterol (RC), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) using data from the National Health and Nutrition Examination Survey (NHANES). METHODS A cross-sectional analysis was conducted using NHANES 2007-2018 data. A total of 8,639 eligible participants (45.58% men and 54.42% women) with an average age of 46.12 ± 16.65 years were included in this study. Weighted multivariate linear regression and subgroup analyses were used to examine the association between vitamin E intake and RC, TC, HDL-C, and LDL-C. Smooth curve fitting was used to explore potential non-linear associations. RESULTS After adjusting for other covariates, multivariate linear regression analysis showed that higher vitamin E intake was negatively associated with plasma RC (β = -0.22, 95% CI: -0.27, -0.16), TC (β = -0.33, 95% CI: -0.51, -0.16), LDL-C (β = -0.25, 95% [confidence interval] CI: -0.40, -0.10) and positively associated with HDL-C (β = 0.13, 95% CI: 0.07, 0.20) in US adults. Subgroup analysis indicated that age may influence the association between vitamin E intake and RC. At the same time, gender may also affect the association between vitamin E intake and HDL-C. CONCLUSION Higher vitamin E intake was negatively associated with plasma RC, TC, LDL-C and positively associated with HDL-C.
Collapse
Affiliation(s)
- Yuxuan Wang
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
- Clinical Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, Shandong, 266000, China
| | - Hao Li
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Zhihao Zhang
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Futong Wu
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Jiarui Liu
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Zhongze Zhu
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Hongfei Xiang
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| |
Collapse
|
48
|
Harada-Shiba M, Davdison MH, Ditmarsch M, Hsieh A, Wuerdeman E, Kling D, Nield A, Dicklin MR, Nakata A, Sueyoshi A, Kuroyanagi S, Kastelein JJ. Obicetrapib as an Adjunct to Stable Statin Therapy in Japanese Subjects: Results from a Randomized Phase 2 Trial. J Atheroscler Thromb 2024; 31:1386-1397. [PMID: 38569868 PMCID: PMC11456355 DOI: 10.5551/jat.64828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 02/15/2024] [Indexed: 04/05/2024] Open
Abstract
AIMS Obicetrapib is a highly selective cholesteryl ester transfer protein (CETP) inhibitor shown to reduce low-density lipoprotein cholesterol (LDL-C) and apolipoprotein B (apoB), when taken as monotherapy and in combination with ezetimibe on a background of statins, in clinical trials predominantly conducted in Northern European/Caucasian participants. We characterized the efficacy, safety, and tolerability of obicetrapib within an Asian-Pacific region population. METHODS This double-blind, randomized, phase 2 trial examined obicetrapib 2.5, 5, and 10 mg/d, compared with placebo, for 8 weeks as an adjunct to stable statin therapy (atorvastatin 10 or 20 mg/d or rosuvastatin 5 or 10 mg/d) in Japanese men and women who had not achieved 2022 Japan Atherosclerosis Society Guidelines and had LDL-C >70 mg/dL or non-high-density lipoprotein cholesterol (non-HDL-C) >100 mg/dL and triglycerides (TG) <400 mg/dL. Endpoints included LDL-C, non-HDL-C, HDL-C, very low-density lipoprotein cholesterol, apolipoproteins, TG, steady state pharmacokinetics (PK) in obicetrapib arms, safety, and tolerability. RESULTS In the 102 randomized subjects (mean age 64.8 y, 71.6% male), obicetrapib significantly lowered median LDL-C, apoB, and non-HDL-C, and raised HDL-C at all doses; responses in the obicetrapib 10 mg group were -45.8%, -29.7%, -37.0%, and +159%, respectively (all p<0.0001 vs. placebo). The PK profile demonstrated near complete elimination of drug by 4 weeks. Obicetrapib was well tolerated and there were no adverse safety signals. CONCLUSIONS All doses of obicetrapib taken as an adjunct to stable statin therapy significantly lowered atherogenic lipoprotein lipid parameters, showed near complete elimination of drug by 4 weeks, and were safe and well tolerated in a Japanese population, similar to previous studies of obicetrapib conducted in predominantly Caucasian participants.
Collapse
Affiliation(s)
- Mariko Harada-Shiba
- Cardiovascular Center, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | | | | | | | | | | | - Annie Nield
- NewAmsterdam Pharma B.V. Naarden, Netherlands
| | | | - Akitaka Nakata
- Department of Cardiology, Sanai Hospital, Saitama, Japan
| | - Atsushi Sueyoshi
- Department of Diabetes Internal Medicine, Uji-Tokushukai Medical Center, Kyoto, Japan
| | - Satoshi Kuroyanagi
- Department of Cardiovascular Surgery, Kishiwada Tokushukai Hospital, Osaka, Japan
| | | |
Collapse
|
49
|
Sheng F, Wang AY, Miyawaki K, Tsuchiya T, Osada N, Miller R, Fu Z, Okamura T. Real-World Clinical Profile of Patients Prescribed Evolocumab in Japan. Circ J 2024; 88:1629-1636. [PMID: 38432948 DOI: 10.1253/circj.cj-23-0814] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
BACKGROUND Real-world utilization data for evolocumab, the first proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor to be introduced in Japan in 2016, to date are limited. This study aimed to clarify the current real-world patient user profiles of evolocumab based on large-scale health claims data. METHODS AND RESULTS This retrospective database study examined patients from a health administrative database (MDV database) who initiated evolocumab between April 2016 (baseline) and November 2021. Characteristics and clinical profiles of this patient population are described. In all, 4,022 patients were included in the final analysis. Most evolocumab prescriptions occurred in the outpatient setting (3,170; 78.82%), and 940 patients (23.37%) had a recent diagnosis of familial hypercholesterolemia. Common recent atherosclerotic cardiovascular disease events at baseline included myocardial infarction (1,633; 40.60%), unstable angina (561; 13.95%), and ischemic stroke (408; 10.14%). Comorbidity diseases included hypertension (2,504; 62.26%), heart failure (1,750; 43.51%), diabetes (1,199; 29.81%), and chronic kidney disease (297; 7.38%). Among the lipid-lowering regimens concomitant with evolocumab, ezetimibe+statin was used most frequently (1,281; 31.85%), followed by no concomitant lipid-lowering regimen (1,190; 29.59%), statin (950; 23.62%), and ezetimibe (601; 14.94%). The median evolocumab treatment duration for all patients was 260 days (interquartile range 57-575 days). CONCLUSIONS This study provides real-world insights into evolocumab utilization in Japan for optimizing patient care and adherence to guideline-based therapies to better address hypercholesterolemia in Japan.
Collapse
Affiliation(s)
| | - Alex Y Wang
- Center for Observational Research, Amgen Australia
| | | | | | | | | | | | - Tomonori Okamura
- Department of Preventive Medicine and Public Health, Keio University School of Medicine
| |
Collapse
|
50
|
Liu D, Zhang J, Zhang X, Jiang F, Wu Y, Yang B, Li X, Fan X, Li H, Sun Y, Gou R, Wang X. The efficacy and safety of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors combined with statins in patients with hypercholesterolemia: a network meta-analysis. Front Cardiovasc Med 2024; 11:1454918. [PMID: 39386388 PMCID: PMC11461350 DOI: 10.3389/fcvm.2024.1454918] [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: 07/05/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024] Open
Abstract
Background In recent years, the position of PCSK9 inhibitors as adjuvant therapy to statins in guidelines has further improved. However, there remained a dearth of direct comparative studies among different PCSK9 inhibitors. Therefore, this study aimed to conduct a network meta-analysis to evaluate the efficacy and safety of different PCSK9 inhibitors combined with statins. Methods A comprehensive literature search was conducted from the study's inception to 12 November 2023, encompassing multiple online databases including PubMed, Embase, Cochrane Central, Web of Science, and ClinicalTrials.gov to obtain relevant randomized controlled trials. Frequentist network meta-analysis was employed to compare the efficacy and safety of different PCSK9 inhibitors. The efficacy endpoints were low-density lipoprotein cholesterol (LDL-C), apolipoprotein B (ApoB), and lipoprotein (a) (Lp(a)). The safety endpoints were any adverse events (AE), severe adverse events (SAE), AE leading to treatment discontinuation, and injection-site reaction. Results Compared with placebo and ezetimibe, all PCSK9 inhibitors demonstrated significant reductions in LDL-C levels. Notably, evolocumab exhibited the most pronounced effect with a treatment difference of -63.67% (-68.47% to -58.87%) compared with placebo. Regarding dosage selection for evolocumab, the regimen of 140 mg Q2W (-69.13%, -74.55% to -63.72%) was superior to 420 mg QM (-61.51%, -65.97% to -57.05%). Based on rankings and P-scores analysis, tafolecimab 150 mg Q2W demonstrated superior efficacy in reducing ApoB levels (-61.70%, -84.38% to -39.02%) and Lp(a) levels (-43%, 30%, -68%, 81% to -17%, 79%). Furthermore, the safety profile of PCSK9 inhibitors was favorable with no increase in the incidence of AE, SAE, or AE leading to treatment discontinuation; however, alirocumab, inclisiran, and tafolecimab may potentially entail a potential risk associated with injection-site reactions. Conclusion Compared with placebo and ezetimibe, PCSK9 inhibitors can significantly reduce LDL-C, ApoB, and Lp(a) when combined with statins to treat hypercholesterolemia. Furthermore, PCSK9 inhibitors and ezetimibe exhibit similar safety profiles. Systematic Review Registration [PROSPERO], identifier [CRD42023490506].
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xinyu Wang
- Clinical Pharmacy Office, Baoji Central Hospital, Baoji, Shaanxi, China
| |
Collapse
|