51
|
Terasaka K, Gohbara M, Abe T, Yoshii T, Hanajima Y, Kirigaya J, Horii M, Kikuchi S, Nakahashi H, Matsushita K, Minamimoto Y, Okada K, Matsuzawa Y, Iwahashi N, Kosuge M, Sugano T, Ebina T, Hibi K. Association between evolocumab use and slow progression of aortic valve stenosis. Heart Vessels 2024; 39:725-734. [PMID: 38499696 DOI: 10.1007/s00380-024-02386-6] [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: 10/31/2023] [Accepted: 02/28/2024] [Indexed: 03/20/2024]
Abstract
No medications have been reported to inhibit the progression of aortic valve stenosis (AS). The present study aimed to investigate whether evolocumab use is related to the slow progression of AS evaluated by serial echocardiography. This was a retrospective observational study from 2017 to 2022 at Yokohama City University Medical Center. Patients aged ≥ 18 with moderate AS were included. Exclusion criteria were (1) mild AS; (2) severe AS defined by maximum aortic valve (AV) velocity ≥ 4.0 m/s; and/or (3) no data of annual follow-up echocardiography. The primary endpoint was the association between evolocumab use and annual changes in the maximum AV-velocity or peak AV-pressure gradient (PG). A total of 57 patients were enrolled: 9 patients treated with evolocumab (evolocumab group), and the other 48 patients assigned to a control group. During a median follow-up of 33 months, the cumulative incidence of AS events (a composite of all-cause death, AV intervention, or unplanned hospitalization for heart failure) was 11% in the evolocumab group and 58% in the control group (P = 0.012). Annual change of maximum AV-velocity or peak AV-PG from the baseline to the next year was 0.02 (- 0.18 to 0.22) m/s per year or 0.60 (- 4.20 to 6.44) mmHg per year in the evolocumab group, whereas it was 0.29 (0.04-0.59) m/s per year or 7.61 (1.46-16.48) mmHg per year in the control group (both P < 0.05). Evolocumab use was associated with slow progression of AS and a low incidence of AS events in patients with moderate AS.
Collapse
Affiliation(s)
- Kengo Terasaka
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Advanced Critical Care and Emergency Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Masaomi Gohbara
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
| | - Takeru Abe
- Advanced Critical Care and Emergency Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Tomohiro Yoshii
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Yohei Hanajima
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Jin Kirigaya
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Advanced Critical Care and Emergency Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Mutsuo Horii
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Department of Laboratory Medicine and Clinical Investigation, Yokohama City University Medical Center, Yokohama, Japan
| | - Shinnosuke Kikuchi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Hidefumi Nakahashi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Kensuke Matsushita
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Yugo Minamimoto
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Advanced Critical Care and Emergency Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Kozo Okada
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Yasushi Matsuzawa
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Noriaki Iwahashi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Department of Cardiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masami Kosuge
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Teruyasu Sugano
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Toshiaki Ebina
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Department of Laboratory Medicine and Clinical Investigation, Yokohama City University Medical Center, Yokohama, Japan
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Department of Cardiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
52
|
Ramakrishnan KK, Vikram MA, Sam A, Muralidharan Y, Natarajan P. The Role of Plain CT in Assessing and Estimating Normal Values of Pericardial Fat Pad Thickness and Their Correlation With Patient's Age, Gender, Body Weight, and Body Mass Index. Cureus 2024; 16:e66271. [PMID: 39238697 PMCID: PMC11375986 DOI: 10.7759/cureus.66271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/06/2024] [Indexed: 09/07/2024] Open
Abstract
Introduction The pericardial fat pad, located anteriorly to the heart between the pericardium and myocardium, has garnered significant interest in cardiovascular research due to its potential role in the pathophysiology of various cardiac conditions. Despite its proximity to the myocardium, it is distinct from the epicardial fat depot found between the myocardium and the visceral layer of the pericardium. Studies have shown that excess pericardial fat is associated with an increased risk of heart failure and other cardiovascular diseases. Non-contrast computed tomography (CT) is a reliable, non-invasive method for assessing pericardial fat pad thickness, offering less radiation exposure compared to other imaging modalities. Establishing standardized measurements for pericardial fat pad thickness is essential, particularly for the South Indian population, which may exhibit unique genetic, dietary, and lifestyle influences on these measurements. Materials and methods A cross-sectional study was conducted on 300 participants from South India, stratified into three age groups: 18-35, 36-50, and 51-70 years, with body weights ranging from 45 kg to 120 kg. Participants were recruited from outpatient departments and community outreach programs, ensuring equal representation from each age group. Non-contrast CT imaging was performed using a Siemens Somatom goTop 128 Slice CT scanner to measure pericardial fat pad thickness and correlate it with age, gender, body weight, and body mass index. Exclusion criteria included diagnosed cardiac or pericardial diseases, prior chest surgery or trauma, pregnancy, and contraindications to CT scans. Ethical approval was obtained, and informed consent was collected from all participants. Data analysis was performed using SPSS software, employing descriptive statistics, ANOVA, t-tests, and Pearson's correlation. Results The study included 300 participants, with an equal gender distribution of 150 males and 150 females. Pericardial fat pad thickness increased with age, averaging 4.2 mm in the 18-35 age group, 5.1 mm in the 36-50 age group, and 6.4 mm in the 51-70 age group. Males exhibited a higher average thickness (5.6 mm) compared to females (5.0 mm). Body weight also showed a positive correlation with pericardial fat pad thickness, with mean values increasing from 4.5 mm in the 45-60 kg range to 6.7 mm in the 106-120 kg range. Statistical analysis confirmed significant differences in pericardial fat pad thickness across age groups, genders, and weight categories, emphasizing the importance of these factors in assessing cardiovascular risk. Conclusion This study provides a benchmark for pericardial fat pad thickness in the Kancheepuram Population of South India, highlighting its correlation with age, gender, body weight, and body mass index. The findings underscore the significance of non-invasive CT imaging in evaluating cardiovascular risk factors. Further research should focus on longitudinal studies and advanced imaging techniques to enhance the diagnostic accuracy and clinical relevance of pericardial fat pad measurements. The established reference values can aid clinicians in identifying individuals at higher risk for cardiovascular diseases, facilitating early intervention and management.
Collapse
Affiliation(s)
- Karthik Krishna Ramakrishnan
- Radiodiagnosis, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, IND
| | - Michael Antony Vikram
- Radiodiagnosis, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, IND
| | - Ajina Sam
- Radiodiagnosis, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, IND
| | - Yuvaraj Muralidharan
- Radiodiagnosis, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, IND
| | - Paarthipan Natarajan
- Radiodiagnosis, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, IND
| |
Collapse
|
53
|
Wen H, Huang R, Xu X, Xiong Z, Liu M, Guo Y, Zhuang X, Liao X. Prognostic significance of aortic valve calcification in relation to coronary artery calcification for cardiovascular diseases. Eur J Prev Cardiol 2024; 31:1173-1182. [PMID: 38394450 DOI: 10.1093/eurjpc/zwae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024]
Abstract
AIMS Both coronary artery calcification (CAC) and aortic valve calcification (AVC) are strongly associated with cardiovascular diseases (CVDs), but data about the prognostic significance of multiple cardiovascular calcifications are limited. We aim to investigate the interaction relationship between AVC and CAC for major events. METHODS AND RESULTS We included 6695 participants from the Multi-Ethnic Study of Atherosclerosis at baseline and divided them into four groups: (i) no AVC or CAC; (ii) only AVC; (iii) only CAC; and (iv) with CAC and CAC. The Cox regression model and the Kaplan-Meier method were used to analyse CVD outcomes. We evaluated the interaction between AVC and CAC and their added predictive value based on the pooled cohort equations (PCEs). Subgroup analyses were also explored. Among 6695 participants (mean age 62.2 ± 10.2 years, 47.2% male), after follow-up, 943 cases (14.1%) of CVD and 1274 cases (19.0%) of all-cause death occurred. For participants with both AVC and CAC, the risk of CVD significantly increased [hazard ratio = 3.43 (2.69-4.37), P < 0.001], even higher than the sum of the ones with only AVC and only CAC. This trend remained the same for all-cause death and among subgroup analyses. The addictive interaction was statistically significant (P < 0.001). When AVC and CAC were added, the predictive value of PCEs increased. CONCLUSION Our results indicated a synergistic interaction between valve calcification and coronary calcification in CVDs. Management for both AVC and CAC may bring health co-benefits in preventing poor outcomes.
Collapse
Affiliation(s)
- Han Wen
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Rihua Huang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Xinghao Xu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Zhenyu Xiong
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Menghui Liu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Yue Guo
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Xiaodong Zhuang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Xinxue Liao
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| |
Collapse
|
54
|
Delgado-Lista J, Mostaza JM, Arrobas-Velilla T, Blanco-Vaca F, Masana L, Pedro-Botet J, Perez-Martinez P, Civeira F, Cuende-Melero JI, Gomez-Barrado JJ, Lahoz C, Pintó X, Suarez-Tembra M, Lopez-Miranda J, Guijarro C. Consensus on lipoprotein(a) of the Spanish Society of Arteriosclerosis. Literature review and recommendations for clinical practice. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:243-266. [PMID: 38599943 DOI: 10.1016/j.arteri.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
The irruption of lipoprotein(a) (Lp(a)) in the study of cardiovascular risk factors is perhaps, together with the discovery and use of proprotein convertase subtilisin/kexin type 9 (iPCSK9) inhibitor drugs, the greatest novelty in the field for decades. Lp(a) concentration (especially very high levels) has an undeniable association with certain cardiovascular complications, such as atherosclerotic vascular disease (AVD) and aortic stenosis. However, there are several current limitations to both establishing epidemiological associations and specific pharmacological treatment. Firstly, the measurement of Lp(a) is highly dependent on the test used, mainly because of the characteristics of the molecule. Secondly, Lp(a) concentration is more than 80% genetically determined, so that, unlike other cardiovascular risk factors, it cannot be regulated by lifestyle changes. Finally, although there are many promising clinical trials with specific drugs to reduce Lp(a), currently only iPCSK9 (limited for use because of its cost) significantly reduces Lp(a). However, and in line with other scientific societies, the SEA considers that, with the aim of increasing knowledge about the contribution of Lp(a) to cardiovascular risk, it is relevant to produce a document containing the current status of the subject, recommendations for the control of global cardiovascular risk in people with elevated Lp(a) and recommendations on the therapeutic approach to patients with elevated Lp(a).
Collapse
Affiliation(s)
- Javier Delgado-Lista
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España.
| | - Jose M Mostaza
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario La Paz, Madrid, España
| | - Teresa Arrobas-Velilla
- Sociedad Española de Medicina de Laboratorio (SEQCML), Laboratorio de Bioquímica Clínica, Hospital Universitario Virgen Macarena, Sevilla, España
| | - Francisco Blanco-Vaca
- Departamento de Bioquímica Clínica, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB Sant Pau), Barcelona; Departamento de Bioquímica y Biología Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, España
| | - Luis Masana
- Unidad de Medicina Vascular y Metabolismo, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Tarragona, España
| | - Juan Pedro-Botet
- Unidad de Lípidos y Riesgo Vascular, Servicio de Endocrinología y Nutrición, Hospital del Mar, Barcelona; Departamento de Medicina, Universidad Autónoma de Barcelona, Barcelona, España
| | - Pablo Perez-Martinez
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España
| | - Fernando Civeira
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Servicio de Medicina Interna, Hospital Universitario Miguel Servet, IIS Aragón, Universidad de Zaragoza, Zaragoza; CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, España
| | - Jose I Cuende-Melero
- Consulta de Riesgo Vascular, Servicio de Medicina Interna, Complejo Asistencial Universitario de Palencia, Palencia; Departamento de Medicina, Dermatología y Toxicología, Facultad de Medicina, Universidad de Valladolid, Valladolid, España
| | - Jose J Gomez-Barrado
- Unidad de Cuidados Cardiológicos Agudos y Riesgo Cardiovascular, Servicio de Cardiología, Hospital Universitario San Pedro de Alcántara, Cáceres, España
| | - Carlos Lahoz
- Unidad de Lípidos y Arteriosclerosis, Servicio de Medicina Interna, Hospital La Paz-Carlos III, Madrid, España
| | - Xavier Pintó
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell-Universidad de Barcelona-CiberObn, España
| | - Manuel Suarez-Tembra
- Unidad de Lípidos y RCV, Servicio de Medicina Interna, Hospital San Rafael, A Coruña, España
| | - Jose Lopez-Miranda
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España.
| | - Carlos Guijarro
- Unidad de Medicina Interna, Hospital Universitario Fundación Alcorcón, Universidad Rey Juan Carlos, Alcorcón, Madrid, España
| |
Collapse
|
55
|
Chen C, Dong X, Zhang W, Chang X, Gao W. Dialogue between mitochondria and endoplasmic reticulum-potential therapeutic targets for age-related cardiovascular diseases. Front Pharmacol 2024; 15:1389202. [PMID: 38939842 PMCID: PMC11208709 DOI: 10.3389/fphar.2024.1389202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/30/2024] [Indexed: 06/29/2024] Open
Abstract
Mitochondria-associated endoplasmic reticulum membranes (MAMs) act as physical membrane contact sites facilitating material exchange and signal transmission between mitochondria and endoplasmic reticulum (ER), thereby regulating processes such as Ca2+/lipid transport, mitochondrial dynamics, autophagy, ER stress, inflammation, and apoptosis, among other pathological mechanisms. Emerging evidence underscores the pivotal role of MAMs in cardiovascular diseases (CVDs), particularly in aging-related pathologies. Aging significantly influences the structure and function of the heart and the arterial system, possibly due to the accumulation of reactive oxygen species (ROS) resulting from reduced antioxidant capacity and the age-related decline in organelle function, including mitochondria. Therefore, this paper begins by describing the composition, structure, and function of MAMs, followed by an exploration of the degenerative changes in MAMs and the cardiovascular system during aging. Subsequently, it discusses the regulatory pathways and approaches targeting MAMs in aging-related CVDs, to provide novel treatment strategies for managing CVDs in aging populations.
Collapse
Affiliation(s)
- Chen Chen
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xueyan Dong
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wang Zhang
- Shandong Provincial Mental Health Center, Jinan, China
| | - Xing Chang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wulin Gao
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
56
|
Nicholls SJ. Therapeutic Potential of Lipoprotein(a) Inhibitors. Drugs 2024; 84:637-643. [PMID: 38849700 PMCID: PMC11196316 DOI: 10.1007/s40265-024-02046-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2024] [Indexed: 06/09/2024]
Abstract
Increasing evidence has implicated lipoprotein(a) [Lp(a)] in the causality of atherosclerosis and calcific aortic stenosis. This has stimulated immense interest in developing novel approaches to integrating Lp(a) into the setting of cardiovascular prevention. Current guidelines advocate universal measurement of Lp(a) levels, with the potential to influence cardiovascular risk assessment and triage of higher-risk patients to use of more intensive preventive therapies. In parallel, considerable activity has been undertaken to develop novel therapeutics with the potential to achieve selective and substantial reductions in Lp(a) levels. Early studies of antisense oligonucleotides (e.g., mipomersen, pelacarsen), RNA interference (e.g., olpasiran, zerlasiran, lepodisiran) and small molecule inhibitors (e.g., muvalaplin) have demonstrated effective Lp(a) lowering and good tolerability. These agents are moving forward in clinical development, in order to determine whether Lp(a) lowering reduces cardiovascular risk. The results of these studies have the potential to transform our approach to the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Stephen J Nicholls
- Victorian Heart Institute, Monash University, 631 Blackburn Road, Clayton, Melbourne, VIC, 3168, Australia.
| |
Collapse
|
57
|
Marrero N, Jha K, Razavi AC, Boakye E, Anchouche K, Dzaye O, Budoff MJ, Tsai MY, Shah SJ, Rotter JI, Guo X, Yao J, Blumenthal RS, Thanassoulis G, Post WS, Blaha MJ, Whelton SP. Identifying People at High Risk for Severe Aortic Stenosis: Aortic Valve Calcium Versus Lipoprotein(a) and Low-Density Lipoprotein Cholesterol. Circ Cardiovasc Imaging 2024; 17:e016372. [PMID: 38889215 PMCID: PMC11423705 DOI: 10.1161/circimaging.123.016372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/11/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Aortic valve calcification (AVC), Lp(a) [lipoprotein(a)], and low-density lipoprotein cholesterol (LDL-C) are associated with severe aortic stenosis (AS). We aimed to determine which of these risk factors were most strongly associated with the risk of incident severe AS. METHODS A total of 6792 participants from the MESA study (Multi-Ethnic Study of Atherosclerosis) had computed tomography-quantified AVC, Lp(a), and LDL-C values at MESA visit 1 (2000-2002). We calculated the absolute event rate of incident adjudicated severe AS per 1000 person-years and performed multivariable adjusted Cox proportional hazards regression. RESULTS The mean age was 62 years old, and 47% were women. Over a median 16.7-year follow-up, the rate of incident severe AS increased exponentially with higher AVC, regardless of Lp(a) or LDL-C values. Participants with AVC=0 had a very low rate of severe AS even with elevated Lp(a) ≥50 mg/dL (<0.1/1000 person-years) or LDL-C ≥130 mg/dL (0.1/1000 person-years). AVC >0 was strongly associated with severe AS when Lp(a) <50 mg/dL hazard ratio (HR) of 33.8 (95% CI, 16.4-70.0) or ≥50 mg/dL HR of 61.5 (95% CI, 7.7-494.2) and when LDL-C <130 mg/dL HR of 31.1 (95% CI, 14.4-67.1) or ≥130 mg/dL HR of 50.2 (95% CI, 13.2-191.9). CONCLUSIONS AVC better identifies people at high risk for severe AS compared with Lp(a) or LDL-C, and people with AVC=0 have a very low long-term rate of severe AS regardless of Lp(a) or LDL-C level. These results suggest AVC should be the preferred prognostic risk marker to identify patients at high risk for severe AS, which may help inform participant selection for future trials testing novel strategies to prevent severe AS.
Collapse
Affiliation(s)
| | - Kunal Jha
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
- University of Louisville, Division of Cardiology, KY (K.J.)
| | - Alexander C Razavi
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
- Center for Heart Disease Prevention, Emory School of Medicine, Atlanta, GA (A.C.R.)
| | - Ellen Boakye
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| | - Khalil Anchouche
- Preventive and Genomic Cardiology, Department of Medicine, McGill University, and the McGill University Health Center Research Institute, Montréal, Québec, Canada (K.A., G.T.)
| | - Omar Dzaye
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| | - Matthew J Budoff
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (M.Y.T.)
| | - Sanjiv J Shah
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.)
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences (J.I.R., X.G., J.Y.), The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences (J.I.R., X.G., J.Y.), The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Jie Yao
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences (J.I.R., X.G., J.Y.), The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Roger S Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| | - George Thanassoulis
- Preventive and Genomic Cardiology, Department of Medicine, McGill University, and the McGill University Health Center Research Institute, Montréal, Québec, Canada (K.A., G.T.)
| | - Wendy S Post
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| | - Michael J Blaha
- Department of Medicine (M.J.B.), The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Seamus P Whelton
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| |
Collapse
|
58
|
Kaur G, Abdelrahman K, Berman AN, Biery DW, Shiyovich A, Huck D, Garshick M, Blankstein R, Weber B. Lipoprotein(a): Emerging insights and therapeutics. Am J Prev Cardiol 2024; 18:100641. [PMID: 38646022 PMCID: PMC11033089 DOI: 10.1016/j.ajpc.2024.100641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/08/2024] [Accepted: 02/24/2024] [Indexed: 04/23/2024] Open
Abstract
The strong association between lipoprotein (a) [Lp(a)] and atherosclerotic cardiovascular disease has led to considerations of Lp(a) being a potential target for mitigating residual cardiovascular risk. While approximately 20 % of the population has an Lp(a) level greater than 50 mg/dL, there are no currently available pharmacological lipid-lowering therapies that have demonstrated substantial reduction in Lp(a). Novel therapies to lower Lp(a) include antisense oligonucleotides and small-interfering ribonucleic acid molecules and have shown promising results in phase 2 trials. Phase 3 trials are currently underway and will test the causal relationship between Lp(a) and ASCVD and whether lowering Lp(a) reduces cardiovascular outcomes. In this review, we summarize emerging insights related to Lp(a)'s role as a risk-enhancing factor for ASCVD, association with calcific aortic stenosis, effects of existing therapies on Lp(a) levels, and variations amongst patient populations. The evolving therapeutic landscape of emerging therapeutics is further discussed.
Collapse
Affiliation(s)
- Gurleen Kaur
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Adam N. Berman
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - David W. Biery
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Albert Einstein College of Medicine, New York, NY, USA
| | - Arthur Shiyovich
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel Huck
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Ron Blankstein
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Brittany Weber
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
59
|
Mohammadyari P, Vieceli Dalla Sega F, Fortini F, Minghini G, Rizzo P, Cimaglia P, Mikus E, Tremoli E, Campo G, Calore E, Schifano SF, Zambelli C. Deep-learning survival analysis for patients with calcific aortic valve disease undergoing valve replacement. Sci Rep 2024; 14:10902. [PMID: 38740898 DOI: 10.1038/s41598-024-61685-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
Calcification of the aortic valve (CAVDS) is a major cause of aortic stenosis (AS) leading to loss of valve function which requires the substitution by surgical aortic valve replacement (SAVR) or transcatheter aortic valve intervention (TAVI). These procedures are associated with high post-intervention mortality, then the corresponding risk assessment is relevant from a clinical standpoint. This study compares the traditional Cox Proportional Hazard (CPH) against Machine Learning (ML) based methods, such as Deep Learning Survival (DeepSurv) and Random Survival Forest (RSF), to identify variables able to estimate the risk of death one year after the intervention, in patients undergoing either to SAVR or TAVI. We found that with all three approaches the combination of six variables, named albumin, age, BMI, glucose, hypertension, and clonal hemopoiesis of indeterminate potential (CHIP), allows for predicting mortality with a c-index of approximately 80 % . Importantly, we found that the ML models have a better prediction capability, making them as effective for statistical analysis in medicine as most state-of-the-art approaches, with the additional advantage that they may expose non-linear relationships. This study aims to improve the early identification of patients at higher risk of death, who could then benefit from a more appropriate therapeutic intervention.
Collapse
Affiliation(s)
| | | | | | - Giada Minghini
- Department of Environmental and Prevention Sciences, Università di Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy.
- Department of Translational Medicine, Università di Ferrara, Ferrara, Italy.
- Laboratory for Technologies of Advanced Therapies (LTTA), Ferrara, Italy.
| | - Paolo Cimaglia
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Elisa Mikus
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Gianluca Campo
- Department of Translational Medicine, Università di Ferrara, Ferrara, Italy
- Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Enrico Calore
- Istituto Nazionale di Fisica Nucleare (INFN), Ferrara, Italy
| | - Sebastiano Fabio Schifano
- Istituto Nazionale di Fisica Nucleare (INFN), Ferrara, Italy.
- Department of Environmental and Prevention Sciences, Università di Ferrara, Ferrara, Italy.
| | | |
Collapse
|
60
|
Khan TG, Cunha JB, Raut C, Burroughs M, Goonewardena SN, Smrcka AV, Speliotes EK, Emmer BT. Functional interrogation of cellular Lp(a) uptake by genome-scale CRISPR screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.593568. [PMID: 38766193 PMCID: PMC11100788 DOI: 10.1101/2024.05.11.593568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
An elevated level of lipoprotein(a), or Lp(a), in the bloodstream has been causally linked to the development of atherosclerotic cardiovascular disease and calcific aortic valve stenosis. Steady state levels of circulating lipoproteins are modulated by their rate of clearance, but the identity of the Lp(a) uptake receptor(s) has been controversial. In this study, we performed a genome-scale CRISPR screen to functionally interrogate all potential Lp(a) uptake regulators in HuH7 cells. Strikingly, the top positive and negative regulators of Lp(a) uptake in our screen were LDLR and MYLIP, encoding the LDL receptor and its ubiquitin ligase IDOL, respectively. We also found a significant correlation for other genes with established roles in LDLR regulation. No other gene products, including those previously proposed as Lp(a) receptors, exhibited a significant effect on Lp(a) uptake in our screen. We validated the functional influence of LDLR expression on HuH7 Lp(a) uptake, confirmed in vitro binding between the LDLR extracellular domain and purified Lp(a), and detected an association between loss-of-function LDLR variants and increased circulating Lp(a) levels in the UK Biobank cohort. Together, our findings support a central role for the LDL receptor in mediating Lp(a) uptake by hepatocytes.
Collapse
Affiliation(s)
- Taslima G. Khan
- Program in Chemical Biology, University of Michigan, Ann Arbor MI
| | - Juliana Bragazzi Cunha
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor MI
| | - Chinmay Raut
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor MI
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor MI
| | | | - Sascha N. Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor MI
- Frankel Cardiovascular Center, University of Michigan, Ann Arbor MI
| | - Alan V. Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor MI
| | - Elizabeth K. Speliotes
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor MI
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor MI
| | - Brian T. Emmer
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor MI
- Frankel Cardiovascular Center, University of Michigan, Ann Arbor MI
| |
Collapse
|
61
|
Kamstrup PR, Neely RDG, Nissen S, Landmesser U, Haghikia A, Costa-Scharplatz M, Abbas C, Nordestgaard BG. Lipoprotein(a) and cardiovascular disease: sifting the evidence to guide future research. Eur J Prev Cardiol 2024; 31:903-914. [PMID: 38253342 DOI: 10.1093/eurjpc/zwae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024]
Abstract
Lipoprotein(a) (Lp(a)) is a genetically determined causal risk factor for cardiovascular disease including coronary heart disease, peripheral arterial disease, ischaemic stroke, and calcific aortic valve stenosis. Clinical trials of specific and potent Lp(a)-lowering drugs are currently underway. However, in clinical practice, widespread assessment of Lp(a) is still lacking despite several guideline recommendations to measure Lp(a) at least once in a lifetime in all adults to identify those at high or very high risk due to elevated levels. The present review provides an overview of key findings from observational and genetic Lp(a) studies, highlights the main challenges in observational Lp(a) studies, and proposes a minimum set of requirements to enhance the quality and harmonize the collection of Lp(a)-related data. Adherence to the recommendations set forth in the present manuscript is intended to enhance the quality of future observational Lp(a) studies, to better define thresholds for increased risk, and to better inform clinical trial design. The recommendations can also potentially assist in the interpretation and generalization of clinical trial findings, to improve care of patients with elevated Lp(a) and optimize treatment and prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Pia R Kamstrup
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Borgmester Ib Juuls Vej 73, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - R Dermot G Neely
- Academic Health Science Network North East and North Cumbria (AHSN), Room 2.13, Biomedical Research Building, The Campus for Ageing and Vitality, Nun's Moor Road, Newcastle, NE4 5PL
| | - Steven Nissen
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, Berlin 10178, Germany
| | - Arash Haghikia
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, Berlin 10178, Germany
| | | | - Cheryl Abbas
- Novartis Pharmaceuticals Corporation, US Medical, East Hanover, NJ, USA
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Borgmester Ib Juuls Vej 73, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
62
|
Bess C, Mehta A, Joshi PH. All we need to know about lipoprotein(a). Prog Cardiovasc Dis 2024; 84:27-33. [PMID: 38759878 DOI: 10.1016/j.pcad.2024.05.007] [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: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Lipoprotein(a) [Lp(a)], a genetically determined macromolecular complex, is independently and causally associated with atherosclerotic cardiovascular disease (ASCVD) and calcific aortic stenosis via proposed proinflammatory, prothrombotic, and proatherogenic mechanisms. While Lp(a) measurement standardization issues are being resolved, several guidelines now support testing Lp(a) at least once in each adult's lifetime for ASCVD risk prediction which can foster implementation of more aggressive primary or secondary prevention therapies. Currently, there are several emerging targeted Lp(a) lowering therapies in active clinical investigation for safety and cardiovascular benefit among both primary and secondary prevention populations. First degree relatives of patients with high Lp(a) should be encouraged to undergo cascade screening. Primary prevention patients with high Lp(a) should consider obtaining a coronary calcium score for further risk estimation and to guide further ASCVD risk factor management including consideration of low dose aspirin therapy. Secondary prevention patients with high Lp(a) levels should consider adding PCSK9 inhibition to statin therapy.
Collapse
Affiliation(s)
- Courtney Bess
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern, Dallas, TX, United States of America; Parkland Health and Hospital System, Dallas, TX, United States of America
| | - Anurag Mehta
- VCU Health Pauley Health Center, Richmond, VA, United States of America
| | - Parag H Joshi
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern, Dallas, TX, United States of America; Parkland Health and Hospital System, Dallas, TX, United States of America.
| |
Collapse
|
63
|
Kim M, Kim JJ, Lee ST, Shim Y, Lee H, Bae S, Son NH, Shin S, Jung IH. Association Between Aortic Valve Sclerosis and Clonal Hematopoiesis of Indeterminate Potential. Ann Lab Med 2024; 44:279-288. [PMID: 38205526 PMCID: PMC10813825 DOI: 10.3343/alm.2023.0268] [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: 07/11/2023] [Revised: 10/06/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024] Open
Abstract
Background The mechanism and medical treatment target for degenerative aortic valve disease, including aortic stenosis, is not well studied. In this study, we investigated the effect of clonal hematopoiesis of indeterminate potential (CHIP) on the development of aortic valve sclerosis (AVS), a calcified aortic valve without significant stenosis. Methods Participants with AVS (valves ≥2 mm thick, high echogenicity, and a peak transaortic velocity of <2.5 m/sec) and an age- and sex-matched control group were enrolled. Twenty-four CHIP genes with common variants in cardiovascular disease were used to generate a next-generation sequencing panel. The primary endpoint was the CHIP detection rate between the AVS and control groups. Inverse-probability treatment weighting (IPTW) analysis was performed to adjust for differences in baseline characteristics. Results From April 2020 to April 2022, 187 participants (125 with AVS and 62 controls) were enrolled; the mean age was 72.6±8.5 yrs, and 54.5% were male. An average of 1.3 CHIP variants was observed. CHIP detection, defined by a variant allele frequency (VAF) of ≥0.5%, was similar between the groups. However, the AVS group had larger CHIP clones: 49 (39.2%) participants had a VAF of ≥1% (vs. 13 [21.0%] in the control group; P=0.020), and 25 (20.0%) had a VAF of ≥2% (vs. 4 [6.5%]; P=0.028). AVS is independently associated with a VAF of ≥1% (adjusted odds ratio: 2.44, 95% confidence interval: 1.11-5.36; P=0.027). This trend was concordant and clearer in the IPTW cohort. Conclusions Participants with AVS more commonly had larger CHIP clones than age- and sex-matched controls. Further studies are warranted to identify causality between AVS and CHIP.
Collapse
Affiliation(s)
- Minkwan Kim
- Division of Cardiology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine and Cardiovascular Center, Yongin, Korea
| | - Jin Ju Kim
- Department of Laboratory Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yeeun Shim
- Department of Laboratory Medicine, Graduate School of Medical Sciences, Brain Korea 21 PLUS Project, Yonsei University College of Medicine, Seoul, Korea
| | - Hyeonah Lee
- Department of Laboratory Medicine, Graduate School of Medical Sciences, Brain Korea 21 PLUS Project, Yonsei University College of Medicine, Seoul, Korea
| | - SungA Bae
- Division of Cardiology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine and Cardiovascular Center, Yongin, Korea
| | - Nak-Hoon Son
- Department of Statistics, Keimyung University, Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - In Hyun Jung
- Division of Cardiology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine and Cardiovascular Center, Yongin, Korea
| |
Collapse
|
64
|
Boudoulas KD, Marmagkiolis K, Iliescu C, Boudoulas H. Editorial: Calcific aortic stenosis: A complex entity with multiple coexisting disorders. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2024; 62:18-20. [PMID: 38195344 DOI: 10.1016/j.carrev.2023.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024]
Affiliation(s)
| | - Konstantinos Marmagkiolis
- Tampa Heart, Tampa, FL, USA; Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Cezar Iliescu
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
65
|
Yu M, Bouatia-Naji N. Insights into the Inherited Basis of Valvular Heart Disease. Curr Cardiol Rep 2024; 26:381-392. [PMID: 38581562 DOI: 10.1007/s11886-024-02041-6] [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] [Accepted: 03/07/2024] [Indexed: 04/08/2024]
Abstract
PURPOSE OF REVIEW: Increases in the availability of genetic data and advances in the tools and methods for their analyses have enabled well-powered genetic association studies that have significantly enhanced our understanding of the genetic factors underlying both rare and common valve diseases. Valvular heart diseases, such as congenital valve malformations and degenerative valve lesions, increase the risk of heart failure, arrhythmias, and sudden death. In this review, we provide an updated overview of our current understanding of the genetic mechanisms underlying valvular heart diseases. With a focus on discoveries from the past 5 years, we describe recent insights into genetic risk and underlying biological pathways. RECENT FINDINGS: Recently acquired knowledge around valvular heart disease genetics has provided important insights into novel mechanisms related to disease pathogenesis. Newly identified risk loci associated valvular heart disease mainly regulate the composition of the extracellular matrix, accelerate the endothelial-to-mesenchymal transition, contribute to cilia formation processes, and play roles in lipid metabolism. Large-scale genomic analyses have identified numerous risk loci, genes, and biological pathways associated with degenerative valve disease and congenital valve malformations. Shared risk genes suggest common mechanistic pathways for various valve pathologies. More recent studies have combined cardiac magnetic resonance imaging and machine learning to offer a novel approach for exploring genotype-phenotype relationships regarding valve disease. Progress in the field holds promise for targeted prevention, particularly through the application of polygenic risk scores, and innovative therapies based on the biological mechanisms for predominant forms of valvular heart diseases.
Collapse
Affiliation(s)
- Mengyao Yu
- Shanghai Pudong Hospital, Human Phenome Institute, Fudan University Pudong Medical Center, Zhangjiang Fudan International Innovation Center, Fundan University, 825 Zhangheng Road, Pudong District, Shanghai, 201203, China.
| | | |
Collapse
|
66
|
Praz F, Beyersdorf F, Haugaa K, Prendergast B. Valvular heart disease: from mechanisms to management. Lancet 2024; 403:1576-1589. [PMID: 38554728 DOI: 10.1016/s0140-6736(23)02755-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/16/2023] [Accepted: 12/06/2023] [Indexed: 04/02/2024]
Abstract
Valvular heart disease is common and its prevalence is rapidly increasing worldwide. Effective medical therapies are insufficient and treatment was historically limited to the surgical techniques of valve repair or replacement, resulting in systematic underprovision of care to older patients and those with substantial comorbidities, frailty, or left ventricular dysfunction. Advances in imaging and surgical techniques over the past 20 years have transformed the management of valvular heart disease. Better understanding of the mechanisms and causes of disease and an increasingly extensive and robust evidence base provide a platform for the delivery of individualised treatment by multidisciplinary heart teams working within networks of diagnostic facilities and specialist heart valve centres. In this Series paper, we aim to provide an overview of the current and future management of valvular heart disease and propose treatment approaches based on an understanding of the underlying pathophysiology and the application of multidisciplinary treatment strategies to individual patients.
Collapse
Affiliation(s)
- Fabien Praz
- University Hospital Bern Inselspital, University of Bern, Bern, Switzerland.
| | - Friedhelm Beyersdorf
- Department of Cardiovascular Surgery, Heart Center, University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kristina Haugaa
- Department of Cardiology, Oslo University Hospital, Rikshospitalet and University of Oslo, Oslo, Norway
| | - Bernard Prendergast
- Heart Vascular and Thoracic Institute, Cleveland Clinic London, London, UK; Department of Cardiology, St Thomas' Hospital, London, UK
| |
Collapse
|
67
|
Small AM, Melloni GEM, Kamanu FK, Bergmark BA, Bonaca MP, O’Donoghue ML, Giugliano RP, Scirica BM, Bhatt D, Antman EM, Raz I, Wiviott SD, Truong B, Wilson PWF, Cho K, O’Donnell CJ, Braunwald E, Lubitz SA, Ellinor P, Peloso GM, Ruff CT, Sabatine MS, Natarajan P, Marston NA. Novel Polygenic Risk Score and Established Clinical Risk Factors for Risk Estimation of Aortic Stenosis. JAMA Cardiol 2024; 9:357-366. [PMID: 38416462 PMCID: PMC10902779 DOI: 10.1001/jamacardio.2024.0011] [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: 09/06/2023] [Accepted: 12/28/2023] [Indexed: 02/29/2024]
Abstract
Importance Polygenic risk scores (PRSs) have proven to be as strong as or stronger than established clinical risk factors for many cardiovascular phenotypes. Whether this is true for aortic stenosis remains unknown. Objective To develop a novel aortic stenosis PRS and compare its aortic stenosis risk estimation to established clinical risk factors. Design, Setting, and Participants This was a longitudinal cohort study using data from the Million Veteran Program (MVP; 2011-2020), UK Biobank (2006-2010), and 6 Thrombolysis in Myocardial Infarction (TIMI) trials, including DECLARE-TIMI 58 (2013-2018), FOURIER (TIMI 59; 2013-2017), PEGASUS-TIMI 54 (2010-2014), SAVOR-TIMI 53 (2010-2013), SOLID-TIMI 52 (2009-2014), and ENGAGE AF-TIMI 48 (2008-2013), which were a mix of population-based and randomized clinical trials. Individuals from UK Biobank and the MVP meeting a previously validated case/control definition for aortic stenosis were included. All individuals from TIMI trials were included unless they had a documented preexisting aortic valve replacement. Analysis took place from January 2022 to December 2023. Exposures PRS for aortic stenosis (developed using data from MVP and validated in UK Biobank) and other previously validated cardiovascular PRSs, defined either as a continuous variable or as low (bottom 20%), intermediate, and high (top 20%), and clinical risk factors. Main Outcomes Aortic stenosis (defined using International Classification of Diseases or Current Procedural Terminology codes in UK Biobank and MVP or safety event data in the TIMI trials). Results The median (IQR) age in MVP was 67 (57-73) years, and 135 140 of 147 104 participants (92%) were male. The median (IQR) age in the TIMI trials was 66 (54-78) years, and 45 524 of 59 866 participants (71%) were male. The best aortic stenosis PRS incorporated 5 170 041 single-nucleotide variants and was associated with aortic stenosis in both the MVP testing sample (odds ratio, 1.41; 95% CI, 1.37-1.45 per 1 SD PRS; P = 4.6 × 10-116) and TIMI trials (hazard ratio, 1.44; 95% CI, 1.27-1.62 per 1 SD PRS; P = 3.2 × 10-9). Among genetic and clinical risk factors, the aortic stenosis PRS performed comparably to most risk factors besides age, and within a given age range, the combination of clinical and genetic risk factors was additive, providing a 3- to 4-fold increased gradient of risk of aortic stenosis. However, the addition of the aortic stenosis PRS to a model including clinical risk factors only improved risk discrimination of aortic stenosis by 0.01 to 0.02 (C index in MVP: 0.78 with clinical risk factors, 0.79 with risk factors and aortic stenosis PRS; C index in TIMI: 0.71 with clinical risk factors, 0.73 with risk factors and aortic stenosis PRS). Conclusions This study developed and validated 1 of the first aortic stenosis PRSs. While aortic stenosis genetic risk was independent from clinical risk factors and performed comparably to all other risk factors besides age, genetic risk resulted in only a small improvement in overall aortic stenosis risk discrimination beyond age and clinical risk factors. This work sets the stage for further development of an aortic stenosis PRS.
Collapse
Affiliation(s)
- Aeron M. Small
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Cardiology, Boston Veterans Affairs Healthcare System, West Roxbury, Massachusetts
| | - Giorgio E. M. Melloni
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Frederick K. Kamanu
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Brian A. Bergmark
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marc P. Bonaca
- Department of Medicine, Cardiology and Vascular Medicine, University of Colorado School of Medicine, Aurora
| | - Michelle L. O’Donoghue
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Robert P. Giugliano
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Benjamin M. Scirica
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Deepak Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Elliott M. Antman
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Itamar Raz
- Department of Endocrinology and Metabolism, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Stephen D. Wiviott
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Buu Truong
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Peter W. F. Wilson
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Kelly Cho
- Veterans Affairs Healthcare System, Boston, Massachusetts
- Division of Aging, Mass General Brigham and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Christopher J. O’Donnell
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Cardiology, Boston Veterans Affairs Healthcare System, West Roxbury, Massachusetts
| | - Eugene Braunwald
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Steve A. Lubitz
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - Patrick Ellinor
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Gina M. Peloso
- Veterans Affairs Healthcare System, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Christian T. Ruff
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marc S. Sabatine
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Pradeep Natarajan
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
| | - Nicholas A. Marston
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
68
|
Wulff AB, Nordestgaard BG, Langsted A. Novel Therapies for Lipoprotein(a): Update in Cardiovascular Risk Estimation and Treatment. Curr Atheroscler Rep 2024; 26:111-118. [PMID: 38311667 DOI: 10.1007/s11883-024-01192-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2024] [Indexed: 02/06/2024]
Abstract
PURPOSE OF REVIEW Lipoprotein(a) is an important causal risk factor for cardiovascular disease but currently no available medication effectively reduces lipoprotein(a). This review discusses recent findings regarding lipoprotein(a) as a causal risk factor and therapeutic target in cardiovascular disease, it reviews current clinical recommendations, and summarizes new lipoprotein(a) lowering drugs. RECENT FINDINGS Epidemiological and genetic studies have established lipoprotein(a) as a causal risk factor for cardiovascular disease and mortality. Guidelines worldwide now recommend lipoprotein(a) to be measured once in a lifetime, to offer patients with high lipoprotein(a) lifestyle advise and initiate other cardiovascular medications. Clinical trials including antisense oligonucleotides, small interfering RNAs, and an oral lipoprotein(a) inhibitor have shown great effect on lowering lipoprotein(a) with reductions up to 106%, without any major adverse effects. Recent clinical phase 1 and 2 trials show encouraging results and ongoing phase 3 trials will hopefully result in the introduction of specific lipoprotein(a) lowering drugs to lower the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Anders Berg Wulff
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Frederiksberg, Denmark
| | - Anne Langsted
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen Ø, Denmark.
| |
Collapse
|
69
|
Gagnon E, Arsenault BJ. Drug target Mendelian randomization supports apolipoprotein C3-lowering for lipoprotein-lipid levels reductions and cardiovascular diseases prevention. Atherosclerosis 2024; 391:117501. [PMID: 38547584 DOI: 10.1016/j.atherosclerosis.2024.117501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND AND AIMS Inhibitors of apolipoprotein C-III (apoC3) are currently approved for the reduction of triglyceride levels in patients with Familial Chylomicronemia Syndrome. We used drug target Mendelian randomization (MR) to assess the effect of genetically predicted decrease in apoC3 blood protein levels on cardiometabolic traits and diseases. METHODS We quantified lifelong reductions in apoC3 blood levels by selecting all genome wide significant and independent (r2<0.1) single nucleotide polymorphisms (SNPs) in the APOC3 gene region ±1 Mb, from three genome-wide association studies (GWAS) of apoC3 blood protein levels (deCODE, n = 35,378, Fenland, n = 10,708 and ARIC, n = 7213). We included the largest GWASes on 18 cardiometabolic traits and 9 cardiometabolic diseases as study outcomes. RESULTS A one standard deviation lowering in apoC3 blood protein levels was associated with lower triglycerides, apolipoprotein B, low-density lipoprotein cholesterol, alanine aminotransferase, and glomerular filtration rate as well as higher high-density lipoprotein cholesterol levels. ApoC3 lowering was also associated with lower risk of acute pancreatitis (odds ratio [OR] = 0.91 95% CI = 0.82 to 1.00), aortic stenosis (OR = 0.82 95% CI = 0.73 to 0.93), and coronary artery disease (OR = 0.86 95% CI = 0.80 to 0.93), and was associated with increased parental lifespan (0.06 95% CI = 0.03-0.09 years). These results were concordant across robust MR methods, the three protein datasets and upon adjustment for APOA1, APOA4 and APOA5 using a multivariable MR framework. CONCLUSIONS These results provide evidence that apoC3 lowering could result in widespread benefits for cardiometabolic health and encourage the launch of trials on apoC3 inhibition for coronary artery disease prevention.
Collapse
Affiliation(s)
- Eloi Gagnon
- Centre de Recherche de L'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | - Benoit J Arsenault
- Centre de Recherche de L'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada; Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
70
|
Aikawa E, Blaser MC, Singh SA, Levine RA, Yacoub MH. Challenges and Opportunities in Valvular Heart Disease: From Molecular Mechanisms to the Community. Arterioscler Thromb Vasc Biol 2024; 44:763-767. [PMID: 38536897 PMCID: PMC10977651 DOI: 10.1161/atvbaha.123.319563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Affiliation(s)
- Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert A. Levine
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
71
|
Hilleman DE, Vacek JL, Backes JM. Elevated Lp(a): Guidance for Identifying and Managing Patients. South Med J 2024; 117:208-213. [PMID: 38569611 DOI: 10.14423/smj.0000000000001675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Lipoprotein(a) (Lp(a)) is a unique low-density lipoprotein-like lipoprotein that is considered an independent and causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and calcific aortic valve stenosis. The Lp(a) molecule also contains apolipoprotein A and apolipoprotein B, which collectively promote atherosclerosis, thrombosis, and inflammation. Lp(a) is highly genetic and minimally responsive to nonpharmacological measures. Lp(a) serum levels ≥125 nmol/L are associated with increased ASCVD risk, but this threshold has not been accepted universally. Elevated Lp(a) is the most common genetic dyslipidemia affecting approximately 20% of the general population. Certain currently available lipid-lowering drugs, including the proprotein convertase subtilisin/kexin type 9 therapies, produce moderate reductions in Lp(a); however, none are indicated for the treatment of elevated Lp(a). There are currently four investigational RNA-based therapeutic agents that reduce Lp(a) by 70% to 100%. Two of these agents are being evaluated for ASCVD risk reduction in adequately powered outcomes trials, with results expected in 2 to 3 years. Until such therapies become available and demonstrate favorable clinical outcomes, strategies for elevated Lp(a) primarily involve early and intensive ASCVD risk factor management.
Collapse
Affiliation(s)
- Daniel E Hilleman
- From the Creighton University School of Pharmacy and Health Professions, Omaha, Nebraska
| | - James L Vacek
- Department of Cardiovascular Medicine, University of Kansas Health System, Kansas City
| | - James M Backes
- the Atherosclerosis and LDL-Apheresis Center, University of Kansas Medical Center, KU School of Pharmacy, Lawrence
| |
Collapse
|
72
|
Laffin LJ, Nissen SE. Lp(a) - an overlooked risk factor. Trends Cardiovasc Med 2024; 34:193-199. [PMID: 36681362 DOI: 10.1016/j.tcm.2023.01.003] [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: 10/05/2022] [Revised: 12/09/2022] [Accepted: 01/14/2023] [Indexed: 01/20/2023]
Abstract
Lipoprotein(a) (Lp(a)) is an increasingly discussed and studied risk factor for atherosclerotic cardiovascular disease and aortic valve stenosis. Many genetic and epidemiological studies support the important causal role that Lp(a) plays in the incidence of cardiovascular disease. Although dependent upon the threshold and unit of measurement of Lp(a), most estimates suggest between 20 and 30% of the world's population have elevated serum levels of Lp(a). Lp(a) levels are predominantly mediated by genetics and are not significantly modified by lifestyle interventions. Efforts are ongoing to develop effective pharmacotherapies to lower Lp(a) and to determine if lowering Lp(a) with these medications ultimately decreases the incidence of adverse cardiovascular events. In this review, the genetics and pathophysiological properties of Lp(a) will be discussed as well as the epidemiological data demonstrating its impact on the incidence of cardiovascular disease. Recommendations for screening and how to currently approach patients with elevated Lp(a) are also noted. Finally, the spectrum of pharmacotherapies under development for Lp(a) lowering is detailed.
Collapse
|
73
|
Akahori H. New Classification to Predict Postoperative Prognosis After Transcatheter Aortic Valve Implantation. Circ J 2024; 88:460-461. [PMID: 36928056 DOI: 10.1253/circj.cj-23-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Affiliation(s)
- Hirokuni Akahori
- Department of Cardiovascular and Renal Medicine, Hyogo Medical University
| |
Collapse
|
74
|
Thériault S, Li Z, Abner E, Luan J, Manikpurage HD, Houessou U, Zamani P, Briend M, Boudreau DK, Gaudreault N, Frenette L, Argaud D, Dahmene M, Dagenais F, Clavel MA, Pibarot P, Arsenault BJ, Boekholdt SM, Wareham NJ, Esko T, Mathieu P, Bossé Y. Integrative genomic analyses identify candidate causal genes for calcific aortic valve stenosis involving tissue-specific regulation. Nat Commun 2024; 15:2407. [PMID: 38494474 PMCID: PMC10944835 DOI: 10.1038/s41467-024-46639-4] [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: 06/28/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
There is currently no medical therapy to prevent calcific aortic valve stenosis (CAVS). Multi-omics approaches could lead to the identification of novel molecular targets. Here, we perform a genome-wide association study (GWAS) meta-analysis including 14,819 cases among 941,863 participants of European ancestry. We report 32 genomic loci, among which 20 are novel. RNA sequencing of 500 human aortic valves highlights an enrichment in expression regulation at these loci and prioritizes candidate causal genes. Homozygous genotype for a risk variant near TWIST1, a gene involved in endothelial-mesenchymal transition, has a profound impact on aortic valve transcriptomics. We identify five genes outside of GWAS loci by combining a transcriptome-wide association study, colocalization, and Mendelian randomization analyses. Using cross-phenotype and phenome-wide approaches, we highlight the role of circulating lipoproteins, blood pressure and inflammation in the disease process. Our findings pave the way for the development of novel therapies for CAVS.
Collapse
Affiliation(s)
- Sébastien Thériault
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada.
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, QC, Canada.
| | - Zhonglin Li
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Hasanga D Manikpurage
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Ursula Houessou
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Pardis Zamani
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Mewen Briend
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Dominique K Boudreau
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Nathalie Gaudreault
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Lily Frenette
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Déborah Argaud
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Manel Dahmene
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - François Dagenais
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Surgery, Université Laval, Quebec City, QC, Canada
| | - Marie-Annick Clavel
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Philippe Pibarot
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Benoit J Arsenault
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - S Matthijs Boekholdt
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Patrick Mathieu
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Surgery, Université Laval, Quebec City, QC, Canada
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
75
|
Yoon D, Choi B, Kim JE, Kim EY, Chung SH, Min HJ, Sung Y, Chang EJ, Song JK. Autotaxin inhibition attenuates the aortic valve calcification by suppressing inflammation-driven fibro-calcific remodeling of valvular interstitial cells. BMC Med 2024; 22:122. [PMID: 38486246 PMCID: PMC10941471 DOI: 10.1186/s12916-024-03342-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Patients with fibro-calcific aortic valve disease (FCAVD) have lipid depositions in their aortic valve that engender a proinflammatory impetus toward fibrosis and calcification and ultimately valve leaflet stenosis. Although the lipoprotein(a)-autotaxin (ATX)-lysophosphatidic acid axis has been suggested as a potential therapeutic target to prevent the development of FCAVD, supportive evidence using ATX inhibitors is lacking. We here evaluated the therapeutic potency of an ATX inhibitor to attenuate valvular calcification in the FCAVD animal models. METHODS ATX level and activity in healthy participants and patients with FCAVD were analyzed using a bioinformatics approach using the Gene Expression Omnibus datasets, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, and western blotting. To evaluate the efficacy of ATX inhibitor, interleukin-1 receptor antagonist-deficient (Il1rn-/-) mice and cholesterol-enriched diet-induced rabbits were used as the FCAVD models, and primary human valvular interstitial cells (VICs) from patients with calcification were employed. RESULTS The global gene expression profiles of the aortic valve tissue of patients with severe FCAVD demonstrated that ATX gene expression was significantly upregulated and correlated with lipid retention (r = 0.96) or fibro-calcific remodeling-related genes (r = 0.77) in comparison to age-matched non-FCAVD controls. Orally available ATX inhibitor, BBT-877, markedly ameliorated the osteogenic differentiation and further mineralization of primary human VICs in vitro. Additionally, ATX inhibition significantly attenuated fibrosis-related factors' production, with a detectable reduction of osteogenesis-related factors, in human VICs. Mechanistically, ATX inhibitor prohibited fibrotic changes in human VICs via both canonical and non-canonical TGF-β signaling, and subsequent induction of CTGF, a key factor in tissue fibrosis. In the in vivo FCAVD model system, ATX inhibitor exposure markedly reduced calcific lesion formation in interleukin-1 receptor antagonist-deficient mice (Il1rn-/-, P = 0.0210). This inhibition ameliorated the rate of change in the aortic valve area (P = 0.0287) and mean pressure gradient (P = 0.0249) in the FCAVD rabbit model. Moreover, transaortic maximal velocity (Vmax) was diminished with ATX inhibitor administration (mean Vmax = 1.082) compared to vehicle control (mean Vmax = 1.508, P = 0.0221). Importantly, ATX inhibitor administration suppressed the effects of a high-cholesterol diet and vitamin D2-driven fibrosis, in association with a reduction in macrophage infiltration and calcific deposition, in the aortic valves of this rabbit model. CONCLUSIONS ATX inhibition attenuates the development of FCAVD while protecting against fibrosis and calcification in VICs, suggesting the potential of using ATX inhibitors to treat FCAVD.
Collapse
Affiliation(s)
- Dohee Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Bongkun Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Ji-Eun Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Young Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Soo-Hyun Chung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hyo-Jin Min
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yoolim Sung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Jae-Kwan Song
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
76
|
Li Q, Yin K, Ma HP, Liu HH, Li S, Luo X, Hu R, Zhang WW, Lv ZS, Niu XL, Gu MH, Li CL, Liu YS, Liu YJ, Li HB, Li N, Li C, Gu WW, Li JJ. Application of improved GalNAc conjugation in development of cost-effective siRNA therapies targeting cardiovascular diseases. Mol Ther 2024; 32:637-645. [PMID: 38204163 PMCID: PMC10928129 DOI: 10.1016/j.ymthe.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024] Open
Abstract
N-Acetylgalactosamine (GalNAc)-conjugated small interfering RNA (siRNA) therapies have received approval for treating both orphan and prevalent diseases. To improve in vivo efficacy and streamline the chemical synthesis process for efficient and cost-effective manufacturing, we conducted this study to identify better designs of GalNAc-siRNA conjugates for therapeutic development. Here, we present data on redesigned GalNAc-based ligands conjugated with siRNAs against angiopoietin-like 3 (ANGPTL3) and lipoprotein (a) (Lp(a)), two target molecules with the potential to address large unmet medical needs in atherosclerotic cardiovascular diseases. By attaching a novel pyran-derived scaffold to serial monovalent GalNAc units before solid-phase oligonucleotide synthesis, we achieved increased GalNAc-siRNA production efficiency with fewer synthesis steps compared to the standard triantennary GalNAc construct L96. The improved GalNAc-siRNA conjugates demonstrated equivalent or superior in vivo efficacy compared to triantennary GalNAc-conjugated siRNAs.
Collapse
Affiliation(s)
- Qian Li
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Ke Yin
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Hai-Ping Ma
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Hui-Hui Liu
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Heart Failure Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sha Li
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Luo
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Rong Hu
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | | | | | | | - Mei-Hua Gu
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Cheng-Lu Li
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | | | | | - Hai-Bo Li
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Nancy Li
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Chong Li
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | | | - Jian-Jun Li
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
77
|
Berman AN, Biery DW, Besser SA, Singh A, Shiyovich A, Weber BN, Huck DM, Divakaran S, Hainer J, Kaur G, Blaha MJ, Cannon CP, Plutzky J, Januzzi JL, Booth JN, López JAG, Kent ST, Nasir K, Di Carli MF, Bhatt DL, Blankstein R. Lipoprotein(a) and Major Adverse Cardiovascular Events in Patients With or Without Baseline Atherosclerotic Cardiovascular Disease. J Am Coll Cardiol 2024; 83:873-886. [PMID: 38418000 DOI: 10.1016/j.jacc.2023.12.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/14/2023] [Accepted: 12/22/2023] [Indexed: 03/01/2024]
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is associated with an increased risk of atherosclerotic cardiovascular disease (ASCVD). However, whether the optimal Lp(a) threshold for risk assessment should differ based on baseline ASCVD status is unknown. OBJECTIVES The purpose of this study was to assess the association between Lp(a) and major adverse cardiovascular events (MACE) among patients with and without baseline ASCVD. METHODS We studied a retrospective cohort of patients with Lp(a) measured at 2 medical centers in Boston, Massachusetts, from 2000 to 2019. To assess the association of Lp(a) with incident MACE (nonfatal myocardial infarction [MI], nonfatal stroke, coronary revascularization, or cardiovascular mortality), Lp(a) percentile groups were generated with the reference group set at the first to 50th Lp(a) percentiles. Cox proportional hazards modeling was used to assess the association of Lp(a) percentile group with MACE. RESULTS Overall, 16,419 individuals were analyzed with a median follow-up of 11.9 years. Among the 10,181 (62%) patients with baseline ASCVD, individuals in the 71st to 90th percentile group had a 21% increased hazard of MACE (adjusted HR: 1.21; P < 0.001), which was similar to that of individuals in the 91st to 100th group (adjusted HR: 1.26; P < 0.001). Among the 6,238 individuals without established ASCVD, there was a continuously higher hazard of MACE with increasing Lp(a), and individuals in the 91st to 100th Lp(a) percentile group had the highest relative risk with an adjusted HR of 1.93 (P < 0.001). CONCLUSIONS In a large, contemporary U.S. cohort, elevated Lp(a) is independently associated with long-term MACE among individuals with and without baseline ASCVD. Our results suggest that the threshold for risk assessment may be different in primary vs secondary prevention cohorts.
Collapse
Affiliation(s)
- Adam N Berman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/adambermanMD
| | - David W Biery
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephanie A Besser
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Avinainder Singh
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Arthur Shiyovich
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brittany N Weber
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel M Huck
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sanjay Divakaran
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jon Hainer
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gurleen Kaur
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael J Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, Maryland, USA
| | - Christopher P Cannon
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jorge Plutzky
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | - John N Booth
- Center for Observational Research, Amgen Inc, Thousand Oaks, California, USA
| | | | - Shia T Kent
- Center for Observational Research, Amgen Inc, Thousand Oaks, California, USA
| | - Khurram Nasir
- Department of Cardiovascular Medicine, Division of Cardiovascular Prevention and Wellness, Houston Methodist DeBakey Heart and Vascular Center, Houston, Texas, USA
| | - Marcelo F Di Carli
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai Health System, New York, New York, USA. https://twitter.com/DLBHATTMD
| | - Ron Blankstein
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
78
|
Karp A, Jacobs M, Barris B, Labkowsky A, Frishman WH. Lipoprotein(a): A Review of Risk Factors, Measurements, and Novel Treatment Modalities. Cardiol Rev 2024:00045415-990000000-00218. [PMID: 38415744 DOI: 10.1097/crd.0000000000000667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
The study of lipoprotein(a) [Lp(a)] has long been a source of interest as a possible independent risk factor for atherosclerotic cardiovascular disease (ASCVD). The results of large sample observational studies, genome-wide association studies, and Mendelian randomization studies have been strong indicators supporting the link between ASCVD and Lp(a) despite early studies, with less sensitive assays, failing to show a connection. The recommendations for the indications and frequency of testing Lp(a) levels vary between US, Canadian, and European organizations due to the uncertain role of Lp(a) in ASCVD. The innovation of recent therapies, such as antisense oligonucleotides and small interfering RNA, designed to specifically target and reduce Lp(a) levels by targeting mRNA translation have once more thrust LP(a) into the spotlight of inquiry. These emerging modalities serve the dual purpose of definitively elucidating the connection between elevated Lp(a) levels and atherosclerotic cardiovascular risk, as well as the possibility of providing clinicians with the tools necessary to manage elevated Lp(a) levels in vulnerable populations. This review seeks to examine the mechanisms of atherogenicity of Lp(a) and explore the most current pharmacologic therapies currently in development.
Collapse
Affiliation(s)
- Avrohom Karp
- From the Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| | - Menachem Jacobs
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Ben Barris
- From the Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| | - Alexander Labkowsky
- From the Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| | - William H Frishman
- From the Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| |
Collapse
|
79
|
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, Commodore-Mensah Y, Currie ME, Elkind MSV, Evenson KR, Generoso G, Heard DG, Hiremath S, Johansen MC, Kalani R, Kazi DS, Ko D, Liu J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Perman SM, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Tsao CW, Urbut SM, Van Spall HGC, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2024; 149:e347-e913. [PMID: 38264914 DOI: 10.1161/cir.0000000000001209] [Citation(s) in RCA: 699] [Impact Index Per Article: 699.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2024 AHA Statistical Update is the product of a full year's worth of effort in 2023 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. The AHA strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional global data, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
80
|
Museedi AS, Le Jemtel TH. Mitral Annular Calcification-Related Valvular Disease: A Challenging Entity. J Clin Med 2024; 13:896. [PMID: 38337590 PMCID: PMC10856114 DOI: 10.3390/jcm13030896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Mitral valve annular calcification-related valvular disease is increasingly common due to the rising prevalence of age-related mitral annular calcifications. Mitral annular calcification alters the structure and function of the mitral valve annulus, which in turn causes mitral valve regurgitation, stenosis, or both. As it frequently coexists with comorbid conditions and overlapping symptoms, mitral annular calcification-related valvular disease poses significant diagnostic and therapeutic challenges. For instance, left ventricular diastolic dysfunction hinders the assessment of mitral valvular disease. Detection of mitral annular calcifications and assessment of related mitral valve disease hinge on two-dimensional echocardiography. Comprehensive assessment of mitral annular calcifications and related mitral valve disease may require multidetector computed tomography and three-dimensional echocardiography. Invasive hemodynamic testing with exercise helps identify the cause of symptoms in patients with comorbid conditions, and transcatheter interventions have emerged as a viable therapeutic option for older patients. After an outline of the normal mitral annulus, we examine how mitral annular calcifications lead to mitral valve disease and how to accurately assess mitral regurgitation and stenosis. Lastly, we review surgical and transcatheter approaches to the management of mitral annular calcification-related mitral valve regurgitation, stenosis, or both.
Collapse
Affiliation(s)
| | - Thierry H. Le Jemtel
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, Tulane University Heart and Vascular Institute, New Orleans, LA 70112, USA;
| |
Collapse
|
81
|
Notenboom ML, Van Hoof L, Schuermans A, Takkenberg JJM, Rega FR, Taverne YJHJ. Aortic Valve Embryology, Mechanobiology, and Second Messenger Pathways: Implications for Clinical Practice. J Cardiovasc Dev Dis 2024; 11:49. [PMID: 38392263 PMCID: PMC10888685 DOI: 10.3390/jcdd11020049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
During the Renaissance, Leonardo Da Vinci was the first person to successfully detail the anatomy of the aortic root and its adjacent structures. Ever since, novel insights into morphology, function, and their interplay have accumulated, resulting in advanced knowledge on the complex functional characteristics of the aortic valve (AV) and root. This has shifted our vision from the AV as being a static structure towards that of a dynamic interconnected apparatus within the aortic root as a functional unit, exhibiting a complex interplay with adjacent structures via both humoral and mechanical stimuli. This paradigm shift has stimulated surgical treatment strategies of valvular disease that seek to recapitulate healthy AV function, whereby AV disease can no longer be seen as an isolated morphological pathology which needs to be replaced. As prostheses still cannot reproduce the complexity of human nature, treatment of diseased AVs, whether stenotic or insufficient, has tremendously evolved, with a similar shift towards treatments options that are more hemodynamically centered, such as the Ross procedure and valve-conserving surgery. Native AV and root components allow for an efficient Venturi effect over the valve to allow for optimal opening during the cardiac cycle, while also alleviating the left ventricle. Next to that, several receptors are present on native AV leaflets, enabling messenger pathways based on their interaction with blood and other shear-stress-related stimuli. Many of these physiological and hemodynamical processes are under-acknowledged but may hold important clues for innovative treatment strategies, or as potential novel targets for therapeutic agents that halt or reverse the process of valve degeneration. A structured overview of these pathways and their implications for cardiothoracic surgeons and cardiologists is lacking. As such, we provide an overview on embryology, hemodynamics, and messenger pathways of the healthy and diseased AV and its implications for clinical practice, by relating this knowledge to current treatment alternatives and clinical decision making.
Collapse
Affiliation(s)
- Maximiliaan L. Notenboom
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| | - Lucas Van Hoof
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Art Schuermans
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Johanna J. M. Takkenberg
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| | - Filip R. Rega
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Yannick J. H. J. Taverne
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| |
Collapse
|
82
|
Sakaeyama Y, Kondo K, Terazono S, Fuchinoue Y, Kubota S, Mikai M, Abe M, Sugo N, Nagao T, Nemoto M. Local and systemic factors associated with quantitative stiffness of carotid plaque. Acta Neurochir (Wien) 2024; 166:54. [PMID: 38289409 DOI: 10.1007/s00701-024-05952-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/06/2024] [Indexed: 02/01/2024]
Abstract
PURPOSE Plaque stiffness in carotid artery stenosis is a clinically important factor involved in the development of stroke and surgical complications. The purpose of this study was to clarify which local and systemic factors are associated with the quantitatively measured stiffness of plaque. METHODS The subjects were 104 consecutive patients who underwent carotid endarterectomy at our institution. To measure quantitative stiffness of plaque, we used an industrial hard meter in the operating room within 1 h after removal of plaque. Local factors related to carotid plaque hardness were evaluated, including maximum intima-media thickness (max IMT), degree of stenosis using the European Carotid Surgery Trial (ECST), presence of ulceration or calcification, and echo brightness on preoperative carotid ultrasound. The degree of stenosis was also evaluated using the North American Symptomatic Carotid Endarterectomy Trial method in digital subtraction angiography. Age, sex, and presence or absence of hypertension, diabetes, and dyslipidemia (low-density lipoprotein cholesterol and triglyceride [TG] levels) served as systemic factors and were compared with the quantitative stiffness of carotid plaque. RESULTS In multivariate analysis, ECST stenosis degree, calcification, and IMT max as local factors affected plaque stiffness. As a systemic factor, plaque stiffness was statistically significantly negatively correlated with TG values in multivariate analysis (p < 0.05). CONCLUSION The quantitative stiffness of the plaque was negatively correlated with TG levels as a systemic factor in addition to local factors. This might suggest that reducing high TG levels is associated with plaque stabilization.
Collapse
Affiliation(s)
- Yuki Sakaeyama
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, 6-11-1, Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Kosuke Kondo
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, 6-11-1, Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Sayaka Terazono
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, 6-11-1, Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Yutaka Fuchinoue
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, 6-11-1, Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | | | - Masataka Mikai
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, 6-11-1, Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | | | - Nobuo Sugo
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, 6-11-1, Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan.
| | - Takaaki Nagao
- Department of Neurosurgery (Sakura), School of Medicine, Faculty of Medicine, Toho University, Sakura-City, Chiba, Japan
| | - Masaaki Nemoto
- Department of Neurosurgery (Sakura), School of Medicine, Faculty of Medicine, Toho University, Sakura-City, Chiba, Japan
| |
Collapse
|
83
|
Wachtell K. Understanding the driving mechanisms of bioprosthetic valve degeneration. Heart 2024; 110:223-224. [PMID: 37788891 DOI: 10.1136/heartjnl-2023-323210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/05/2023] Open
Affiliation(s)
- Kristian Wachtell
- Department of Cardiology, Cornell University Joan and Sanford I Weill Medical College, New York, New York, USA
| |
Collapse
|
84
|
Farina JM, Chao CJ, Pereyra M, Roarke M, Said EF, Barry T, Alsidawi S, Sell-Dottin K, Sweeney JP, Fortuin DF, Ayoub C, Lester SJ, Oh JK, Arsanjani R, Marcotte F. Role of lipoprotein(a) concentrations in bioprosthetic aortic valve degeneration. Heart 2024; 110:299-305. [PMID: 37643771 DOI: 10.1136/heartjnl-2023-322987] [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: 05/18/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023] Open
Abstract
OBJECTIVES Lipoprotein(a) (Lp(a)) is associated with an increased incidence of native aortic stenosis, which shares similar pathological mechanisms with bioprosthetic aortic valve (bAV) degeneration. However, evidence regarding the role of Lp(a) concentrations in bAV degeneration is lacking. This study aims to evaluate the association between Lp(a) concentrations and bAV degeneration. METHODS In this retrospective multicentre study, patients who underwent a bAV replacement between 1 January 2010 and 31 December 2020 and had a Lp(a) measurement were included. Echocardiography follow-up was performed to determine the presence of bioprosthetic valve degeneration, which was defined as an increase >10 mm Hg in mean gradient from baseline with concomitant decrease in effective orifice area and Doppler Velocity Index, or new moderate/severe prosthetic regurgitation. Levels of Lp(a) were compared between patients with and without degeneration and Cox regression analysis was performed to investigate the association between Lp(a) levels and bioprosthetic valve degeneration. RESULTS In total, 210 cases were included (mean age 74.1±9.4 years, 72.4% males). Median time between baseline and follow-up echocardiography was 4.4 (IQR 3.7) years. Bioprostheses degeneration was observed in 33 (15.7%) patients at follow-up. Median serum levels of Lp(a) were significantly higher in patients affected by degeneration versus non-affected cases: 50.0 (IQR 72.0) vs 15.6 (IQR 48.6) mg/dL, p=0.002. In the regression analysis, high Lp(a) levels (≥30 mg/dL) were associated with degeneration both in a univariable analysis (HR 3.6, 95% CI 1.7 to 7.6, p=0.001) and multivariable analysis adjusted by other risk factors for bioprostheses degeneration (HR 4.4, 95% CI 1.9 to 10.4, p=0.001). CONCLUSIONS High serum Lp(a) is associated with bAV degeneration. Prospective studies are needed to confirm these findings and to investigate whether lowering Lp(a) levels could slow bioprostheses degradation.
Collapse
Affiliation(s)
- Juan M Farina
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Chieh-Ju Chao
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Michael Roarke
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Ebram F Said
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Timothy Barry
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Said Alsidawi
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Kristen Sell-Dottin
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - John P Sweeney
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - David F Fortuin
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Steven J Lester
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Jae K Oh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Francois Marcotte
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| |
Collapse
|
85
|
Tasdighi E, Adhikari R, Almaadawy O, Leucker TM, Blaha MJ. LP(a): Structure, Genetics, Associated Cardiovascular Risk, and Emerging Therapeutics. Annu Rev Pharmacol Toxicol 2024; 64:135-157. [PMID: 37506332 DOI: 10.1146/annurev-pharmtox-031023-100609] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Lipoprotein(a) [Lp(a)] is a molecule bound to apolipoprotein(a) with some similarity to low-density lipoprotein cholesterol (LDL-C), which has been found to be a risk factor for cardiovascular disease (CVD). Lp(a) appears to induce inflammation, atherogenesis, and thrombosis. Approximately 20% of the world's population has increased Lp(a) levels, determined predominantly by genetics. Current clinical practices for the management of dyslipidemia are ineffective in lowering Lp(a) levels. Evolving RNA-based therapeutics, such as the antisense oligonucleotide pelacarsen and small interfering RNA olpasiran, have shown promising results in reducing Lp(a) levels. Phase III pivotal cardiovascular outcome trials [Lp(a)HORIZON and OCEAN(a)] are ongoing to evaluate their efficacy in secondary prevention of major cardiovascular events in patients with elevated Lp(a). The future of cardiovascular residual risk reduction may transition to a personalized approach where further lowering of either LDL-C, triglycerides, or Lp(a) is selected after high-intensity statin therapy based on the individual risk profile and preferences of each patient.
Collapse
Affiliation(s)
- Erfan Tasdighi
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rishav Adhikari
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Omar Almaadawy
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore, Maryland, USA
| | - Thorsten M Leucker
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael J Blaha
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
86
|
Li ZH, Hao QY, Zeng YH, Guo JB, Li SC, Gao JW, Yang PZ. Remnant cholesterol and the risk of aortic valve calcium progression: insights from the MESA study. Cardiovasc Diabetol 2024; 23:20. [PMID: 38195550 PMCID: PMC10777602 DOI: 10.1186/s12933-023-02081-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/28/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Remnant cholesterol (RC) is implicated in the risk of cardiovascular disease. However, comprehensive population-based studies elucidating its association with aortic valve calcium (AVC) progression are limited, rendering its precise role in AVC ambiguous. METHODS From the Multi-Ethnic Study of Atherosclerosis database, we included 5597 individuals (61.8 ± 10.1 years and 47.5% men) without atherosclerotic cardiovascular disease at baseline for analysis. RC was calculated as total cholesterol minus high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C), as estimated by the Martin/Hopkins equation. Using the adjusted Cox regression analyses, we examined the relationships between RC levels and AVC progression. Furthermore, we conducted discordance analyses to evaluate the relative AVC risk in RC versus LDL-C discordant/concordant groups. RESULTS During a median follow-up of 2.4 ± 0.9 years, 568 (10.1%) participants exhibited AVC progression. After adjusting for traditional cardiovascular risk factors, the HRs (95% CIs) for AVC progression comparing the second, third, and fourth quartiles of RC levels with the first quartile were 1.195 (0.925-1.545), 1.322 (1.028-1.701) and 1.546 (1.188-2.012), respectively. Notably, the discordant high RC/low LDL-C group demonstrated a significantly elevated risk of AVC progression compared to the concordant low RC/LDL-C group based on their medians (HR, 1.528 [95% CI 1.201-1.943]). This pattern persisted when clinical LDL-C threshold was set at 100 and 130 mg/dL. The association was consistently observed across various sensitivity analyses. CONCLUSIONS In atherosclerotic cardiovascular disease-free individuals, elevated RC is identified as a residual risk for AVC progression, independent of traditional cardiovascular risk factors. The causal relationship of RC to AVC and the potential for targeted RC reduction in primary prevention require deeper exploration.
Collapse
Affiliation(s)
- Ze-Hua Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Qing-Yun Hao
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yu-Hong Zeng
- Medical Apparatus and Equipment Deployment, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Bin Guo
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Shi-Chao Li
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Wei Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Ping-Zhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
87
|
Mukherjee D, Nissen SE. Lipoprotein (a) as a Biomarker for Cardiovascular Diseases and Potential New Therapies to Mitigate Risk. Curr Vasc Pharmacol 2024; 22:171-179. [PMID: 38141196 DOI: 10.2174/0115701611267835231210054909] [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: 08/04/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is a molecule that induces inflammation of the blood vessels, atherogenesis, valvular calcification, and thrombosis. METHODS We review the available evidence that suggests that high Lp(a) levels are associated with a persisting risk for atherosclerotic cardiovascular diseases despite optimization of established risk factors, including low-density lipoprotein cholesterol (LDL-C) levels. OBSERVATIONS Approximately a quarter of the world population have Lp(a) levels of >50 mg/dL (125 nmol/L), a level associated with elevated cardiovascular risk. Lifestyle modification, statins, and ezetimibe do not effectively lower Lp(a) levels, while proprotein convertase subtilisin/kexin type 9 (PCSK-9) inhibitors and niacin only lower Lp(a) levels modestly. We describe clinical studies suggesting that gene silencing therapeutics, such as small interfering RNA (siRNA) and antisense oligonucleotide targeting Lp(a), offer a targeted approach with the potential for safe and robust Lp(a)- lowering with only a few doses (3-4) per year. Prospective randomized phase 3 studies are ongoing to validate safety, effectiveness in improving hard clinical outcomes, and tolerability to assess these therapies. CONCLUSION Several emerging treatments with robust Lp(a)-lowering effects may significantly lower atherosclerotic cardiovascular risk.
Collapse
Affiliation(s)
- Debabrata Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center at El Paso, Texas, USA
| | - Steven E Nissen
- Heart, Vascular, and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
88
|
Abstract
Importance Mendelian randomization (MR) is a statistical approach that has become increasingly popular in the field of cardiovascular disease research. It offers a way to infer potentially causal relationships between risk factors and outcomes using observational data, which is particularly important in cases where randomized clinical trials are not feasible or ethical. With the growing availability of large genetic data sets, MR has become a powerful and accessible tool for studying the risk factors for cardiovascular disease. Observations MR uses genetic variation associated with modifiable exposures or risk factors to mitigate biases that affect traditional observational study designs. The approach uses genetic variants that are randomly assigned at conception as proxies for exposure to a risk factor, mimicking a randomized clinical trial. By comparing the outcomes of individuals with different genetic variants, researchers may draw causal inferences about the effects of specific risk factors on cardiovascular disease, provided assumptions are met that address (1) the association between each genetic variant and risk factor and (2) the association of the genetic variants with confounders and (3) that the association between each genetic variant and the outcome only occurs through the risk factor. Like other observational designs, MR has limitations, which include weak instruments that are not strongly associated with the exposure of interest, linkage disequilibrium where genetic instruments influence the outcome via correlated rather than direct effects, overestimated genetic associations, and selection and survival biases. In addition, many genetic databases and MR studies primarily include populations genetically similar to European reference populations; improved diversity of participants in these databases and studies is critically needed. Conclusions and Relevance This review provides an overview of MR methodology, including assumptions, strengths, and limitations. Several important applications of MR in cardiovascular disease research are highlighted, including the identification of drug targets, evaluation of potential cardiovascular risk factors, as well as emerging methodology. Overall, while MR alone can never prove a causal relationship beyond reasonable doubt, MR offers a rigorous approach for investigating possible causal relationships in observational data and has the potential to transform our understanding of the etiology and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| | - Stephen Burgess
- Medical Research Council Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
89
|
Whelton SP, Jha K, Dardari Z, Razavi AC, Boakye E, Dzaye O, Verghese D, Shah S, Budoff MJ, Matsushita K, Carr JJ, Vasan RS, Blumenthal RS, Anchouche K, Thanassoulis G, Guo X, Rotter JI, McClelland RL, Post WS, Blaha MJ. Prevalence of Aortic Valve Calcium and the Long-Term Risk of Incident Severe Aortic Stenosis. JACC Cardiovasc Imaging 2024; 17:31-42. [PMID: 37178073 PMCID: PMC10902718 DOI: 10.1016/j.jcmg.2023.02.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Aortic valve calcification (AVC) is a principal mechanism underlying aortic stenosis (AS). OBJECTIVES This study sought to determine the prevalence of AVC and its association with the long-term risk for severe AS. METHODS Noncontrast cardiac computed tomography was performed among 6,814 participants free of known cardiovascular disease at MESA (Multi-Ethnic Study of Atherosclerosis) visit 1. AVC was quantified using the Agatston method, and normative age-, sex-, and race/ethnicity-specific AVC percentiles were derived. The adjudication of severe AS was performed via chart review of all hospital visits and supplemented with visit 6 echocardiographic data. The association between AVC and long-term incident severe AS was evaluated using multivariable Cox HRs. RESULTS AVC was present in 913 participants (13.4%). The probability of AVC >0 and AVC scores increased with age and were generally highest among men and White participants. In general, the probability of AVC >0 among women was equivalent to men of the same race/ethnicity who were approximately 10 years younger. Incident adjudicated severe AS occurred in 84 participants over a median follow-up of 16.7 years. Higher AVC scores were exponentially associated with the absolute risk and relative risk of severe AS with adjusted HRs of 12.9 (95% CI: 5.6-29.7), 76.4 (95% CI: 34.3-170.2), and 380.9 (95% CI: 169.7-855.0) for AVC groups 1 to 99, 100 to 299, and ≥300 compared with AVC = 0. CONCLUSIONS The probability of AVC >0 varied significantly by age, sex, and race/ethnicity. The risk of severe AS was exponentially higher with higher AVC scores, whereas AVC = 0 was associated with an extremely low long-term risk of severe AS. The measurement of AVC provides clinically relevant information to assess an individual's long-term risk for severe AS.
Collapse
Affiliation(s)
- Seamus P Whelton
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.
| | - Kunal Jha
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Zeina Dardari
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | - Ellen Boakye
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Omar Dzaye
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Dhiran Verghese
- Department of Medicine, Harbor University of California, Los Angeles Medical Center, Los Angeles, California, USA
| | - Sanjiv Shah
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Matthew J Budoff
- Department of Medicine, Harbor University of California, Los Angeles Medical Center, Los Angeles, California, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - J Jeffery Carr
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ramachandran S Vasan
- University of Texas School of Public Health San Antonio, San Antonio, Texas, USA
| | - Roger S Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Khalil Anchouche
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | | | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor University of California, Los Angeles Medical Center, Torrance, California, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor University of California, Los Angeles Medical Center, Torrance, California, USA
| | - Robyn L McClelland
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Wendy S Post
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Michael J Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
90
|
Thomas PE, Vedel-Krogh S, Nordestgaard BG. Measuring lipoprotein(a) for cardiovascular disease prevention - in whom and when? Curr Opin Cardiol 2024; 39:39-48. [PMID: 38078600 DOI: 10.1097/hco.0000000000001104] [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: 12/18/2023]
Abstract
PURPOSE OF REVIEW The aim of this study is to summarize major cardiovascular guideline recommendations on lipoprotein(a) and highlighting recent findings that emphasize how measuring lipoprotein(a) once in all adults is meaningful regardless of age, sex, comorbidities, or ethnicity. RECENT FINDINGS Many international guidelines now recommend once in a lifetime measurement of lipoprotein(a) in all adult individuals to facilitate accurate risk prediction. Lipoprotein(a)-lowering therapy to reduce cardiovascular disease is on the horizon, with results from the first phase 3 trial expected in 2025. SUMMARY Elevated lipoprotein(a) is an independent causal risk factor for atherosclerotic cardiovascular disease and aortic valve stenosis and measuring lipoprotein(a) once in all individuals regardless of age, sex, comorbidities, or ethnicity is meaningful to aid in risk stratification.
Collapse
Affiliation(s)
- Peter E Thomas
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Signe Vedel-Krogh
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
91
|
Sarzani R, Spannella F, Di Pentima C, Giulietti F, Landolfo M, Allevi M. Molecular Therapies in Cardiovascular Diseases: Small Interfering RNA in Atherosclerosis, Heart Failure, and Hypertension. Int J Mol Sci 2023; 25:328. [PMID: 38203499 PMCID: PMC10778861 DOI: 10.3390/ijms25010328] [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: 11/16/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Small interfering RNA (siRNA) represents a novel, fascinating therapeutic strategy that allows for selective reduction in the production of a specific protein through RNA interference. In the cardiovascular (CV) field, several siRNAs have been developed in the last decade. Inclisiran has been shown to significantly reduce low-density lipoprotein cholesterol (LDL-C) circulating levels with a reassuring safety profile, also in older patients, by hampering proprotein convertase subtilisin/kexin type 9 (PCSK9) production. Olpasiran, directed against apolipoprotein(a) mRNA, prevents the assembly of lipoprotein(a) [Lp(a)] particles, a lipoprotein linked to an increased risk of ischemic CV disease and heart valve damage. Patisiran, binding transthyretin (TTR) mRNA, has demonstrated an ability to improve heart failure and polyneuropathy in patients with TTR amyloidosis, even in older patients with wild-type form. Zilebesiran, designed to reduce angiotensinogen secretion, significantly decreases systolic and diastolic blood pressure (BP). Thanks to their effectiveness, safety, and tolerability profile, and with a very low number of administrations in a year, thus overcoming adherence issues, these novel drugs are the leaders of a new era in molecular therapies for CV diseases.
Collapse
Affiliation(s)
- Riccardo Sarzani
- Internal Medicine and Geriatrics, ESH “Hypertension Excellence Centre”, SISA LIPIGEN Centre, IRCCS INRCA, 60127 Ancona, Italy; (R.S.); (M.L.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, 60126 Ancona, Italy
| | - Francesco Spannella
- Internal Medicine and Geriatrics, ESH “Hypertension Excellence Centre”, SISA LIPIGEN Centre, IRCCS INRCA, 60127 Ancona, Italy; (R.S.); (M.L.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, 60126 Ancona, Italy
| | - Chiara Di Pentima
- Internal Medicine and Geriatrics, ESH “Hypertension Excellence Centre”, SISA LIPIGEN Centre, IRCCS INRCA, 60127 Ancona, Italy; (R.S.); (M.L.)
| | - Federico Giulietti
- Internal Medicine and Geriatrics, ESH “Hypertension Excellence Centre”, SISA LIPIGEN Centre, IRCCS INRCA, 60127 Ancona, Italy; (R.S.); (M.L.)
| | - Matteo Landolfo
- Internal Medicine and Geriatrics, ESH “Hypertension Excellence Centre”, SISA LIPIGEN Centre, IRCCS INRCA, 60127 Ancona, Italy; (R.S.); (M.L.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, 60126 Ancona, Italy
| | - Massimiliano Allevi
- Internal Medicine and Geriatrics, ESH “Hypertension Excellence Centre”, SISA LIPIGEN Centre, IRCCS INRCA, 60127 Ancona, Italy; (R.S.); (M.L.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, 60126 Ancona, Italy
| |
Collapse
|
92
|
Jasti M, Islam S, Steele N, Ivy K, Maimo W, Isiadinso I. Lp(a) and risk of cardiovascular disease - A review of existing evidence and emerging concepts. J Natl Med Assoc 2023:S0027-9684(23)00141-4. [PMID: 38143155 DOI: 10.1016/j.jnma.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death among adults in the United States. There has been significant advancement in the diagnosis and treatment of atherosclerotic cardiovascular disease (ASCVD) and its underlying risk factors. In certain populations, there remains a significant residual risk despite adequate lowering of low-density lipoprotein cholesterol (LDL-C) and control of traditional risk factors. This has led to an interest in research to identify additional risk factors that contribute to atherosclerotic cardiovascular disease. Elevated lipoprotein (a) [Lp(a)] has been identified as an independent risk factor contributing to an increased risk for CVD. There are also ethnic and racial disparities in Lp(a) inheritance that need to be understood. This paper reviews the current literature on lipoprotein a, proposed mechanisms of actions for cardiovascular disease, recommendations for testing, and the current and emerging therapies for lowering Lp(a).
Collapse
Affiliation(s)
- Manasa Jasti
- Division of Cardiology, University of Tennessee Health Science Center/Ascension Saint Thomas, Nashville, TN, United States
| | - Sabrina Islam
- Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Nathan Steele
- Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Kendra Ivy
- Department of Internal Medicine, Morehouse School of Medicine, Atlanta, GA, United States
| | - Willibroad Maimo
- Division of Cardiology, Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Ijeoma Isiadinso
- Division of Cardiology, Department of Medicine, Center for Heart Disease Prevention, Emory University School of Medicine, Atlanta, GA, United States.
| |
Collapse
|
93
|
Chen S, Li Z, Li H, Zeng X, Yuan H, Li Y. Novel lipid biomarkers and ratios as risk predictors for premature coronary artery disease: A retrospective analysis of 2952 patients. J Clin Hypertens (Greenwich) 2023; 25:1172-1184. [PMID: 37986641 PMCID: PMC10710552 DOI: 10.1111/jch.14751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023]
Abstract
This study examined the associations between emerging lipid biomarkers (small dense low-density lipoprotein cholesterol [sdLDL-C), lipoprotein(a) [Lp(a)], and free fatty acids [FFA]), two ratios (sdLDL-C/LDL-C and the triglyceride-glucose [TyG) index), and the Gensini score (GS) in patients with premature coronary artery disease (PCAD) in relation to the extent of coronary stenosis. The authors evaluated a cohort of 2952 individuals undergoing coronary angiography (CAG), encompassing those with PCAD (n = 1749), late-onset coronary artery disease (LCAD; n = 328), and non-coronary artery disease (non-CAD; n = 575). Noteworthy differences were observed in the levels of the novel lipid biomarkers and ratio indexes among the PCAD, LCAD, and non-CAD groups (p < .05). Multiple logistic regression analyses pinpointed Lp(a) (OR = 2.62, 95% CI 1.22-5.63, p = .014) and the TyG index (OR = 2.53, 95% CI 1.08-5.93, p = .033) as independent risk factors for PCAD. Furthermore, these biomarkers and ratio indexes discerned substantial distinctions among PCAD patients with varying GS (p < .05). Consequently, these markers can proficiently anticipate the gravity of coronary artery stenosis (GS > 40) in PCAD patients, as evidenced by the ROC analysis. In conclusion, sdLDL-C, Lp(a), FFA, and the sdLDL-C/LDL-C and TyG indexes have considerable potential as risk and diagnostic markers for coronary artery stenosis in individuals afflicted with PCAD.
Collapse
Affiliation(s)
- Si Chen
- Department of Clinical LaboratoryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- State Key Laboratory of ComplexSevere and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Clinical LaboratoryBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Zhan Li
- Department of Clinical LaboratoryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- State Key Laboratory of ComplexSevere and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Haolong Li
- Department of Clinical LaboratoryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- State Key Laboratory of ComplexSevere and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoli Zeng
- Department of Clinical LaboratoryBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Hui Yuan
- Department of Clinical LaboratoryBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yongzhe Li
- Department of Clinical LaboratoryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- State Key Laboratory of ComplexSevere and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
94
|
Roeters van Lennep JE, Tokgözoğlu LS, Badimon L, Dumanski SM, Gulati M, Hess CN, Holven KB, Kavousi M, Kayıkçıoğlu M, Lutgens E, Michos ED, Prescott E, Stock JK, Tybjaerg-Hansen A, Wermer MJH, Benn M. Women, lipids, and atherosclerotic cardiovascular disease: a call to action from the European Atherosclerosis Society. Eur Heart J 2023; 44:4157-4173. [PMID: 37611089 PMCID: PMC10576616 DOI: 10.1093/eurheartj/ehad472] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/25/2023] Open
Abstract
Cardiovascular disease is the leading cause of death in women and men globally, with most due to atherosclerotic cardiovascular disease (ASCVD). Despite progress during the last 30 years, ASCVD mortality is now increasing, with the fastest relative increase in middle-aged women. Missed or delayed diagnosis and undertreatment do not fully explain this burden of disease. Sex-specific factors, such as hypertensive disorders of pregnancy, premature menopause (especially primary ovarian insufficiency), and polycystic ovary syndrome are also relevant, with good evidence that these are associated with greater cardiovascular risk. This position statement from the European Atherosclerosis Society focuses on these factors, as well as sex-specific effects on lipids, including lipoprotein(a), over the life course in women which impact ASCVD risk. Women are also disproportionately impacted (in relative terms) by diabetes, chronic kidney disease, and auto-immune inflammatory disease. All these effects are compounded by sociocultural components related to gender. This panel stresses the need to identify and treat modifiable cardiovascular risk factors earlier in women, especially for those at risk due to sex-specific conditions, to reduce the unacceptably high burden of ASCVD in women.
Collapse
Affiliation(s)
- Jeanine E Roeters van Lennep
- Department of Internal Medicine, Cardiovascular Institute, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Lale S Tokgözoğlu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Lina Badimon
- Cardiovascular Science Program-ICCC, IR-Hospital de la Santa Creu I Santa Pau, Ciber CV, Autonomous University of Barcelona, Barcelona, Spain
| | - Sandra M Dumanski
- Department of Medicine, Cumming School of Medicine, University of Calgary, Libin Cardiovascular Institute, and O’Brien Institute for Public Health, Calgary, Canada
| | - Martha Gulati
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Connie N Hess
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora and CPC Clinical Research Aurora, CO, USA
| | - Kirsten B Holven
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, and National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Meral Kayıkçıoğlu
- Department of Cardiology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Esther Lutgens
- Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, USA
| | - Erin D Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eva Prescott
- Department of Cardiology, Bispebjerg University Hospital, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
| | - Jane K Stock
- European Atherosclerosis Society, Mässans Gata 10, SE-412 51 Gothenburg, Sweden
| | - Anne Tybjaerg-Hansen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte Hospital, and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marieke J H Wermer
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology at University Medical Center Groningen, Groningen, The Netherlands
| | - Marianne Benn
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte Hospital, and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
95
|
Di Costanzo A, Indolfi C, Franzone A, Esposito G, Spaccarotella CAM. Lp(a) in the Pathogenesis of Aortic Stenosis and Approach to Therapy with Antisense Oligonucleotides or Short Interfering RNA. Int J Mol Sci 2023; 24:14939. [PMID: 37834387 PMCID: PMC10573862 DOI: 10.3390/ijms241914939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
To date, no medical therapy can slow the progression of aortic stenosis. Fibrocalcific stenosis is the most frequent form in the general population and affects about 6% of the elderly population. Over the years, diagnosis has evolved thanks to echocardiography and computed tomography assessments. The application of artificial intelligence to electrocardiography could further implement early diagnosis. Patients with severe aortic stenosis, especially symptomatic patients, have valve repair as their only therapeutic option by surgical or percutaneous technique (TAVI). The discovery that the pathogenetic mechanism of aortic stenosis is similar to the atherosclerosis process has made it possible to evaluate the hypothesis of medical therapy for aortic stenosis. Several drugs have been tested to reduce low-density lipoprotein (LDL) and lipoprotein(a) (Lp(a)) levels, inflammation, and calcification. The Proprotein Convertase Subtilisin/Kexin type 9 inhibitors (PCSK9-i) could decrease the progression of aortic stenosis and the requirement for valve implantation. Great interest is related to circulating Lp(a) levels as causally linked to degenerative aortic stenosis. New therapies with ASO (antisense oligonucleotides) and siRNA (small interfering RNA) are currently being tested. Olpasiran and pelacarsen reduce circulating Lp(a) levels by 85-90%. Phase 3 studies are underway to evaluate the effect of these drugs on cardiovascular events (cardiovascular death, non-fatal myocardial injury, and non-fatal stroke) in patients with elevated Lp(a) and CVD (cardiovascular diseases). For instance, if a reduction in Lp(a) levels is associated with aortic stenosis prevention or progression, further prospective clinical trials are warranted to confirm this observation in this high-risk population.
Collapse
Affiliation(s)
- Assunta Di Costanzo
- Division of Cardiology, Cardiovascular Research Center, University Magna Graecia Catanzaro, 88100 Catanzaro, Italy;
| | - Ciro Indolfi
- Division of Cardiology, Cardiovascular Research Center, University Magna Graecia Catanzaro, 88100 Catanzaro, Italy;
| | - Anna Franzone
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.F.); (G.E.); (C.A.M.S.)
| | - Giovanni Esposito
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.F.); (G.E.); (C.A.M.S.)
| | - Carmen Anna Maria Spaccarotella
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.F.); (G.E.); (C.A.M.S.)
| |
Collapse
|
96
|
Thanassoulis G. Lipoprotein(a): Solving the Puzzle, Deciphering the Riddle, Unraveling the Enigma. Can J Cardiol 2023; 39:1325-1327. [PMID: 37479083 DOI: 10.1016/j.cjca.2023.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 07/02/2023] [Indexed: 07/23/2023] Open
Affiliation(s)
- George Thanassoulis
- Department of Medicine, McGill University, McGill University Health Center, Montreal, Quebec, Canada.
| |
Collapse
|
97
|
Ma GS, Chiou TT, Wilkinson MJ. Is Lipoprotein(a) Clinically Actionable with Today's Evidence? The Answer is Yes. Curr Cardiol Rep 2023; 25:1175-1187. [PMID: 37632608 PMCID: PMC10651710 DOI: 10.1007/s11886-023-01937-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] [Accepted: 08/05/2023] [Indexed: 08/28/2023]
Abstract
PURPOSE OF REVIEW Lipoprotein(a) is an independent risk factor for cardiovascular disease. We review the ongoing shifts in consensus guidelines for the testing and management of Lp(a) and provide insight into whether current evidence suggests that awareness and testing of Lp(a) is clinically actionable. RECENT FINDINGS GWAS and Mendelian randomization studies have established causal links between elevated Lp(a) and forms of CVD, including CAD and calcific aortic valve disease. Testing of Lp(a) identifies patients with similar risk to that of heterozygous FH, enhances risk stratification in patients with borderline/intermediate risk as determined through traditional factors, and facilitates the assessment of inherited CVD risk through cascade screening in patients with known family history of elevated Lp(a). Reductions in Lp(a) through non-targeted therapies including PCSK9 inhibition and lipoprotein apheresis have demonstrated reductions in ASCVD risk that are likely attributable to lowering Lp(a). Targeted therapies to potently lower Lp(a) are in clinical development. Lp(a) is actionable, and can be used to identify high risk patients for primary prevention and their family members through cascade screening, and to guide intensification of therapy in primary and secondary prevention of ASCVD.
Collapse
Affiliation(s)
- Gary S Ma
- Division of Cardiovascular Medicine, Department of Medicine, Cardiovascular Institute, UC San Diego Health, Sulpizio Cardiovascular Center, University of California San Diego, 9434 Medical Center Dr, MC 7241, La Jolla, CA, 92037, San Diego, USA
| | - Tommy T Chiou
- Division of Cardiovascular Medicine, Department of Medicine, Cardiovascular Institute, UC San Diego Health, Sulpizio Cardiovascular Center, University of California San Diego, 9434 Medical Center Dr, MC 7241, La Jolla, CA, 92037, San Diego, USA
| | - Michael J Wilkinson
- Division of Cardiovascular Medicine, Department of Medicine, Cardiovascular Institute, UC San Diego Health, Sulpizio Cardiovascular Center, University of California San Diego, 9434 Medical Center Dr, MC 7241, La Jolla, CA, 92037, San Diego, USA.
| |
Collapse
|
98
|
Tselepis AD. Treatment of Lp(a): Is It the Future or Are We Ready Today? Curr Atheroscler Rep 2023; 25:679-689. [PMID: 37668953 PMCID: PMC10564831 DOI: 10.1007/s11883-023-01141-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 09/06/2023]
Abstract
PURPOSE OF REVIEW The goal of this review is to present the pharmacodynamic effectiveness as well as the clinical efficacy and safety of investigational antisense oligonucleotides (ASOs) and small interference RNAs (siRNAs) drugs that specifically target lipoprotein(a) (Lp(a)). The review will discuss whether the existing lipid-lowering therapies are adequate to treat high Lp(a) levels or whether it is necessary to use the emerging new therapeutic approaches which are based on the current RNA technologies. RECENT FINDINGS Lipoprotein(a) (Lp(a)) is a causal risk factor for atherosclerotic cardiovascular disease (ASCVD), independent of other conventional risk factors. High Lp(a) levels are also independently associated with an increased risk of aortic stenosis progression rate. Plasma Lp(a) levels are primarily genetically determined by variation in the LPA gene coding for apo(a). All secondary prevention trials have demonstrated that the existing hypolipidemic therapies are not adequate to reduce Lp(a) levels to such an extent that could lead to a substantial reduction of ASCVD risk. This has led to the development of new drugs that target the mRNA transcript of LPA and efficiently inhibit Lp(a) synthesis leading to potent Lp(a) reduction. These new drugs are the ASO pelacarsen and the siRNAs olpasiran and SLN360. Recent pharmacodynamic studies showed that all these drugs potently reduce Lp(a) up to 98%, in a dose-dependent manner. Ongoing clinical trials will determine the Lp(a)-lowering efficacy, tolerability, and safety of these drugs as well as their potential effectiveness in reducing the ASCVD risk attributed to high plasma Lp(a) levels. We are not ready today to significantly reduce plasma Lp(a). Emerging therapies potently decrease Lp(a) and ongoing clinical trials will determine their effectiveness in reducing ASCVD risk in subjects with high Lp(a) levels.
Collapse
Affiliation(s)
- Alexandros D Tselepis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110, Ioannina, Greece.
| |
Collapse
|
99
|
Chiesa G, Zenti MG, Baragetti A, Barbagallo CM, Borghi C, Colivicchi F, Maggioni AP, Noto D, Pirro M, Rivellese AA, Sampietro T, Sbrana F, Arca M, Averna M, Catapano AL. Consensus document on Lipoprotein(a) from the Italian Society for the Study of Atherosclerosis (SISA). Nutr Metab Cardiovasc Dis 2023; 33:1866-1877. [PMID: 37586921 DOI: 10.1016/j.numecd.2023.07.019] [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: 06/14/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/18/2023]
Abstract
AIMS In view of the consolidating evidence on the causal role of Lp(a) in cardiovascular disease, the Italian Society for the Study of Atherosclerosis (SISA) has assembled a consensus on Lp(a) genetics and epidemiology, together with recommendations for its measurement and current and emerging therapeutic approaches to reduce its plasma levels. Data on the Italian population are also provided. DATA SYNTHESIS Lp(a) is constituted by one apo(a) molecule and a lipoprotein closely resembling to a low-density lipoprotein (LDL). Its similarity with an LDL, together with its ability to carry oxidized phospholipids are considered the two main features making Lp(a) harmful for cardiovascular health. Plasma Lp(a) concentrations vary over about 1000 folds in humans and are genetically determined, thus they are quite stable in any individual. Mendelian Randomization studies have suggested a causal role of Lp(a) in atherosclerotic cardiovascular disease (ASCVD) and aortic valve stenosis and observational studies indicate a linear direct correlation between cardiovascular disease and Lp(a) plasma levels. Lp(a) measurement is strongly recommended once in a patient's lifetime, particularly in FH subjects, but also as part of the initial lipid screening to assess cardiovascular risk. The apo(a) size polymorphism represents a challenge for Lp(a) measurement in plasma, but new strategies are overcoming these difficulties. A reduction of Lp(a) levels can be currently attained only by plasma apheresis and, moderately, with PCSK9 inhibitor treatment. CONCLUSIONS Awaiting the approval of selective Lp(a)-lowering drugs, an intensive management of the other risk factors for individuals with elevated Lp(a) levels is strongly recommended.
Collapse
Affiliation(s)
- Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy.
| | - Maria Grazia Zenti
- Section of Diabetes and Metabolism, Pederzoli Hospital, Peschiera Del Garda, Verona, Italy.
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Carlo M Barbagallo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Claudio Borghi
- Department of Cardiovascular Medicine, IRCCS AOU S. Orsola, Bologna, Italy
| | - Furio Colivicchi
- Division of Clinical Cardiology, San Filippo Neri Hospital, Rome, Italy
| | - Aldo P Maggioni
- ANMCO Research Center, Heart Care Foundation, Firenze, Italy
| | - Davide Noto
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine and Surgery, University of Perugia, Italy
| | - Angela A Rivellese
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Tiziana Sampietro
- Lipoapheresis Unit, Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Francesco Sbrana
- Lipoapheresis Unit, Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Marcello Arca
- Department of Translational and Precision Medicine (DTPM), Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Maurizio Averna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy; Institute of Biophysics, National Council of Researches, Palermo, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| |
Collapse
|
100
|
Vinci P, Di Girolamo FG, Panizon E, Tosoni LM, Cerrato C, Pellicori F, Altamura N, Pirulli A, Zaccari M, Biasinutto C, Roni C, Fiotti N, Schincariol P, Mangogna A, Biolo G. Lipoprotein(a) as a Risk Factor for Cardiovascular Diseases: Pathophysiology and Treatment Perspectives. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6721. [PMID: 37754581 PMCID: PMC10531345 DOI: 10.3390/ijerph20186721] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023]
Abstract
Cardiovascular disease (CVD) is still a leading cause of morbidity and mortality, despite all the progress achieved as regards to both prevention and treatment. Having high levels of lipoprotein(a) [Lp(a)] is a risk factor for cardiovascular disease that operates independently. It can increase the risk of developing cardiovascular disease even when LDL cholesterol (LDL-C) levels are within the recommended range, which is referred to as residual cardiovascular risk. Lp(a) is an LDL-like particle present in human plasma, in which a large plasminogen-like glycoprotein, apolipoprotein(a) [Apo(a)], is covalently bound to Apo B100 via one disulfide bridge. Apo(a) contains one plasminogen-like kringle V structure, a variable number of plasminogen-like kringle IV structures (types 1-10), and one inactive protease region. There is a large inter-individual variation of plasma concentrations of Lp(a), mainly ascribable to genetic variants in the Lp(a) gene: in the general po-pulation, Lp(a) levels can range from <1 mg/dL to >1000 mg/dL. Concentrations also vary between different ethnicities. Lp(a) has been established as one of the risk factors that play an important role in the development of atherosclerotic plaque. Indeed, high concentrations of Lp(a) have been related to a greater risk of ischemic CVD, aortic valve stenosis, and heart failure. The threshold value has been set at 50 mg/dL, but the risk may increase already at levels above 30 mg/dL. Although there is a well-established and strong link between high Lp(a) levels and coronary as well as cerebrovascular disease, the evidence regarding incident peripheral arterial disease and carotid atherosclerosis is not as conclusive. Because lifestyle changes and standard lipid-lowering treatments, such as statins, niacin, and cholesteryl ester transfer protein inhibitors, are not highly effective in reducing Lp(a) levels, there is increased interest in developing new drugs that can address this issue. PCSK9 inhibitors seem to be capable of reducing Lp(a) levels by 25-30%. Mipomersen decreases Lp(a) levels by 25-40%, but its use is burdened with important side effects. At the current time, the most effective and tolerated treatment for patients with a high Lp(a) plasma level is apheresis, while antisense oligonucleotides, small interfering RNAs, and microRNAs, which reduce Lp(a) levels by targeting RNA molecules and regulating gene expression as well as protein production levels, are the most widely explored and promising perspectives. The aim of this review is to provide an update on the current state of the art with regard to Lp(a) pathophysiological mechanisms, focusing on the most effective strategies for lowering Lp(a), including new emerging alternative therapies. The purpose of this manuscript is to improve the management of hyperlipoproteinemia(a) in order to achieve better control of the residual cardiovascular risk, which remains unacceptably high.
Collapse
Affiliation(s)
- Pierandrea Vinci
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Filippo Giorgio Di Girolamo
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Emiliano Panizon
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Letizia Maria Tosoni
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Carla Cerrato
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Federica Pellicori
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Nicola Altamura
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Alessia Pirulli
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Michele Zaccari
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Chiara Biasinutto
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Chiara Roni
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Nicola Fiotti
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Paolo Schincariol
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, I.R.C.C.S “Burlo Garofolo”, 34137 Trieste, Italy;
| | - Gianni Biolo
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| |
Collapse
|