1
|
Jha M, McCarthy IR, Gelfand EV. Lipoprotein(a) - From Biomarker to Therapy: A Review for the Clinician. Am J Cardiol 2025; 245:42-53. [PMID: 40057218 DOI: 10.1016/j.amjcard.2025.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/15/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Cardiovascular disease (CVD) remains the predominant cause of morbidity and mortality globally. Amid rising CVD rates, Lipoprotein(a) [Lp(a)] has been recognized as a critical biomarker identifying individuals at an increased risk of atherosclerotic cardiovascular disease (ASCVD) and calcific aortic valve stenosis (AS), independent of traditional risk factors. Lp(a) is a lipoprotein variant similar to LDL but includes apolipoprotein(a), which influences its pathogenic potential. Elevated Lp(a) levels are genetically determined and have been implicated in promoting vascular inflammation, atherogenesis, enhanced calcification, and thrombosis. Emerging antisense oligonucleotide (ASO)- and small interfering ribonucleic acids (siRNAs)- based therapies have been shown to lower Lp(a) concentrations, with ongoing trials underway to determine whether they reduce the risk of CVD. While guidelines on screening and management continue to evolve, the advent of specific Lp(a)-lowering therapies may transform CVD prevention and treatment. This review aims to consolidate the current knowledge on Lp(a) from its biological functions to its implications for clinical practice, focusing on its role as a biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Mawra Jha
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Inbar R McCarthy
- Department of Cardiovascular Medicine, Lahey Hospital & Medical Center, University of Massachusetts Chan Medical School, Burlington, Massachusetts
| | - Eli V Gelfand
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
2
|
Kaindl L, Hotz JF, Ferrari J. Cutting-edge stroke prevention strategies. eNeurologicalSci 2025; 39:100561. [PMID: 40070447 PMCID: PMC11893301 DOI: 10.1016/j.ensci.2025.100561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
Stroke represents a major public health problem, ranking as a leading cause of death and long-term disability worldwide (1-3). The goal of prevention strategies is to avoid the occurrence of stroke by educating both healthcare professionals and patients about the reduction of risk factors. Despite increasing improvements in acute revascularization therapies, preventive measures remain more effective in decreasing the burden of stroke, as evidenced by the fact that over 75 % of strokes are first events (2). This paper attempts to provide a comprehensive approach by outlining risk factors and new therapeutic approaches. Understanding these is essential for healthcare providers and the general public to develop effective prevention strategies and ultimately reduce the overall incidence of stroke.
Collapse
Affiliation(s)
- Lisa Kaindl
- Dept. of Neurology, St. John's of God Hospital, Vienna, Austria
| | - Julian Frederic Hotz
- Dept. of Neurology, St. John's of God Hospital, Vienna, Austria
- Dept. of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Austria
| | - Julia Ferrari
- Dept. of Neurology, St. John's of God Hospital, Vienna, Austria
| |
Collapse
|
3
|
Chandra AA, Luciano PD, Swett K, Kaplan R, Talavera GA, Lamar M, Tarraf W, Marquez F, Joshi PH, Gallo L, Sotres-Alvarez D, Gianola M, Daviglus ML, Labovitz DL, Gonzalez H, DeCarli C, Rodriguez CJ. Association of Lp(a) With Stroke and Cerebral Injury on MRI: Insights From the HCHS/SOL (Hispanic Community Health Study/Study of Latinos) and Investigation of Neurocognitive Aging MRI (SOL-INCA MRI). Stroke 2025; 56:1492-1504. [PMID: 40166810 PMCID: PMC12101938 DOI: 10.1161/strokeaha.124.048439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/24/2025] [Accepted: 03/07/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Lp(a) (lipoprotein[a]) is a risk factor for cardiovascular disease; however, its association with cerebrovascular disease is not as well established. METHODS Data from a population-based cohort of Hispanics/Latinos included 16 333 individuals with baseline Lp(a) levels (nmol/L) and self-reported prevalent stroke or transient ischemic attack (TIA). A subset of 2642 individuals with brain magnetic resonance imaging was also included. Linear and multivariate logistic regression assessed the association of Lp(a) with (1) self-reported stroke or TIA, (2) cerebral injury defined as self-reported stroke or TIA or evidence of a stroke on brain magnetic resonance imaging, (3) white matter hyperintensity volume, and (4) silent brain infarcts. Sampling weights were utilized given the HCHS/SOL (Hispanic Community Health Study/Study of Latinos) complex sample design. RESULTS Mean age±SE was 41.1±0.3 years, 52.0% women, and median interquartile range (Q1, Q3) Lp(a) level of 19.7 (7.3-60.6) nmol/L; brain magnetic resonance imaging subset mean age±SE was 49.9±0.4 years, 56.4% women, and median (interquartile range) Lp(a) level of 21.7 (8.1-62.9) nmol/L. Each unit increase in log-transformed Lp(a) was associated with higher odds of self-reported stroke or TIA (odds ratio, 1.13 [95% CI, 1.01-1.27]; P=0.03). Lp(a) levels in the highest quintile (>77 nmol/L) were significantly associated with higher odds of prevalent stroke or TIA compared with Lp(a) <6 nmol/L (first quintile: odds ratio, 1.74 [95% CI, 1.09-2.77]; P=0.02). The highest proportion of cerebral injury was noted in Q5, while the lowest proportion was noted in Q2. When comparing Lp(a) >77 nmol/L with Lp(a) of 6 to <13 nmol/L (second quintile), a significant association was found between Lp(a) and cerebral injury that persisted after fully adjusted models (odds ratio, 2.03 [95% CI, 1.05-3.93]; P=0.03). Each unit increase in log-Lp(a) was associated with a 0.10 increase in log-white matter hyperintensity (β, 0.10; P=0.005). No significant association was found between Lp(a) and silent brain infarcts. CONCLUSIONS Lp(a) is independently and significantly associated with prevalent stroke/TIA, and white matter hyperintensity, in a large diverse population of Hispanics/Latinos.
Collapse
Affiliation(s)
| | | | | | | | | | - Melissa Lamar
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Barkas F, Brandts J, De Bacquer D, Jennings C, De Backer GG, Kotseva K, Ryden L, Lip GY, Santos RD, Libby P, Erlund I, Ganly S, Vihervaara T, Adamska A, Abreu A, Almahmeed W, Ambari AM, Ge J, Hasan-Ali H, Huo Y, Jankowski P, Jimenez RM, Li Y, Mahmood Zuhdi AS, Makubi A, Mbakwem AC, Mbau L, Navarro Estrada JL, Ogah OS, Ogola EN, Quintero–Baiz A, Sani MU, Sosa Liprandi MI, Tan JWC, Urina Triana MA, Yeo TJ, Wood D, McEvoy JW, Ray KK, McEvoy J, Wood D, Adamska A, De Backer G, De Bacquer D, Erlund I, Ganly S, Jennings C, Kotseva K, Lip G, Ray K, Rydén L, Abreu A, Almahmeed W, Ambari A, Ge J, Hasan-Ali H, Huo Y, Jankowski P, Jimenez R, Li Y, Mahmood Zuhdi A, Makubi A, Mbakwem A, Mbau L, Navarro Estrada J, Ogah O, Ogola E, Quintero-Baiz A, Sani M, Sosa Liprandi M, Tan J, Urina Triana M, Yeo T, Wood D, Adamska A, Jennings C, Kotseva K, Ganly S, Zekic A, Rydén L, Salako D, Toulouse C, Chhabra G, Spragg R, Sullivan M, De Bacquer D, Erlund I, Jarvensivu E, Vihervaara T, Deschle H, Estrada JLN, Liprandi MIS, Bellia S, Villarreal R, Elfman M, Perea J, Lombardi F, Masson W, Castaño B, Gamarra A, et alBarkas F, Brandts J, De Bacquer D, Jennings C, De Backer GG, Kotseva K, Ryden L, Lip GY, Santos RD, Libby P, Erlund I, Ganly S, Vihervaara T, Adamska A, Abreu A, Almahmeed W, Ambari AM, Ge J, Hasan-Ali H, Huo Y, Jankowski P, Jimenez RM, Li Y, Mahmood Zuhdi AS, Makubi A, Mbakwem AC, Mbau L, Navarro Estrada JL, Ogah OS, Ogola EN, Quintero–Baiz A, Sani MU, Sosa Liprandi MI, Tan JWC, Urina Triana MA, Yeo TJ, Wood D, McEvoy JW, Ray KK, McEvoy J, Wood D, Adamska A, De Backer G, De Bacquer D, Erlund I, Ganly S, Jennings C, Kotseva K, Lip G, Ray K, Rydén L, Abreu A, Almahmeed W, Ambari A, Ge J, Hasan-Ali H, Huo Y, Jankowski P, Jimenez R, Li Y, Mahmood Zuhdi A, Makubi A, Mbakwem A, Mbau L, Navarro Estrada J, Ogah O, Ogola E, Quintero-Baiz A, Sani M, Sosa Liprandi M, Tan J, Urina Triana M, Yeo T, Wood D, Adamska A, Jennings C, Kotseva K, Ganly S, Zekic A, Rydén L, Salako D, Toulouse C, Chhabra G, Spragg R, Sullivan M, De Bacquer D, Erlund I, Jarvensivu E, Vihervaara T, Deschle H, Estrada JLN, Liprandi MIS, Bellia S, Villarreal R, Elfman M, Perea J, Lombardi F, Masson W, Castaño B, Gamarra A, Llamedo M, Santander P, Barco A, Flores V, Leyes C, Rodríguez F, Marturano M, Alustiza W, Duronto E, Procopio G, Higa C, Corigliano O, Sanchez M, del Cristo Mendoza Beltrán F, Luna P, Urina Triana M, Quintero-Baiz A, Renowitsky C, Pulgar M, Lievano J, Hernandez A, Cordoba G, Duque M, Duque L, Mendoza F, Rojas J, Jaramillo N, Zuluaga S, Restrepo A, Munos J, Buitrago A, Ramirez H, Figueroa F, Gonzales C, Cadena A, Bermudez S, Gomez L, Firdaus I, Dwiputra B, Arityanti D, Susilowati E, Hamdani R, Syaoqi M, Ridwan M, Fitra ND M, Ridwan M, Hartopo A, Arso I, Anggraeni V, Raynaldo A, Hasan H, Siregar Y, Tjahjono C, Sihotang F, Erickatulistiawan G, Tiksnadi B, Febrianora M, Tarsidin N, Fong A, Ahmad WAW, Zuhdi ASM, Zuhdi ASM, Ahmad WAW, Hadi M, Kassim ZA, Shaharudin N, Mohamad Sithik M, Moussa IM, Fong A, Ong T, Foo D, Jong R, Lee C, Kim H, Mahadevan G, Abdul Kader M, Aziz NA, Liew H, Jam E, Regaibalan D, Bugarin O, Leus A, Baniqued A, Arellano J, Mendoza L, Jimenez R, Mendoza R, Reyes M, Locnen S, Luna D, Tiongco R, Alad P, Besa J, Tirador L, Estoce E, Sian A, Roque R, Montejo A, Discípulo A, Lastimosa H, Ybanez E, Bernan A, Coronel R, Samson M, Tamayo T, Tan H, Tan J, Yeo T, Dalakoti M, Fadzillah NFM, Lim S, Low T, Ong J, Ting J, Wang L, Wong C, Ho J, Chee F, Cheng C, Koh C, Yip D, Huang Z, Chin C, Chua K, Fam J, Idu M, Keh Y, Lim C, Rizwan M, Tan J, Tsang C, Wong N, Yap J, Tan M, Tan J, Goh L, Tan K, Wong Y, Yee Y, Yuslane M, Weng Y, Wong C, Imran S, Lim Z, Lu T, Su J, Hoon V, Ho Y, Quek S, Shawki I, Hasan-Ali H, Abdelmegid M, Marghany A, Mosad E, Shafik E, Ibrahim A, Ghaleb R, Kholef E, Shamandy B, Moustafa T, Sobeih M, Mortada M, Elbarbary M, Elawady M, Al Ali J, Almahmeed W, Hashmani S, Manla Y, Green C, Garrod R, Nour S, Alhammadi AF, Mohamed Y, Isaac R, Bazargani N, Almulla A, Praveen J, Al Ali J, Bashir OAO, Faraj A, Hashmath S, Hamadi RL, Ahmad R, Prabakaran M, JI X, Ge J, Huo Y, Li Y, Ge J, Wang C, Zhao G, Mi S, Peng Y, Qi Z, Li Y, Bao L, Gao W, Huo Y, Zheng B, Fang J, Gu X, Su L, Wang Q, Tong S, Chen X, Fu Y, Li Y, Shi J, Jing J, Sheng L, He M, Wang S, Pan W, Kong Y, Cong H, Zhang Y, Chen J, Shi B, Zhao R, Zhang W, Ye J, Hou C, Huang H, Yang L, Chang L, Yang Z, Gao C, Zhang J, Zhang Y, Samia B, Ogola E, Mbau L, Mwazo K, Bajaber A, Nduati B, Mkilo J, Mwasha M, Sood M, Barasa F, Biwott P, Gitura B, Kinuthia P, Ogah O, Mbakwem A, Sani M, Orimolade O, Omoruyi E, Makinde O, Ojo V, Otesanya F, Amadi C, Ogedegbe S, Ejim E, Odom A, Udora N, Maduka C, Onyemachi J, Edafe E, Dodiyi-Manuel S, Bamigbowu O, Ossai G, Umuerri E, Oghojamoni-Ogefere P, Oyovwevotu M, Ojji D, Ajanya O, Alo-Joseph C, Daniel O, Ibrahim A, Ngada E, Ripiye N, Taiwo F, Ubah I, Ogunmodede J, Adeniyi S, Olugbola O, Sani M, Alfa I, Abdussalam T, Abdullahi U, Isezuo S, Zagga M, Umar H, Okocha H, Aloja N, Buba F, Talle M, Ahmadu N, Galtimari I, Tukur A, Zakariyya Z, Ayoola Y, Oyekunle R, Adamu S, Mvungi R, Makubi A, Kisenge P, Msigwa S, Meda J, Magitta N, Chandika A, Mitkowski P, Jankowski P, Kamiński K, Łapińska M, Sobkowicz B, Knapp M, Mickiewicz K, Dobrzycki S, Hirnle T, Charkiewicz-Szeremeta K, Bychowski J, Guzowska – Suchowolec A, Kubica A, Michalski P, Kosobucka-Ozdoba A, Pietrzykowski Ł, Rzepka-Cholasińska A, Ratajczak J, Siedlaczek M, Skonieczny G, Kostrzewa P, Gąsior Z, Maciejowski Ł, Matla-Hajzyk M, Hapeta-Zeman B, Szóstak-Janiak K, Kosior D, Kulak P, Rajska E, Kosior D, Rak A, Jankowski P, Koczwarska-Maciejek D, Wolfshaut-Wolak R, Gonçalves L, Pereira H, Abreu A, Carvalho M, Amador A, Calvão J, Costa C, Moreira H, Palma P, Pinto R, Proença T, Rocha M, Moreira N, Almeida J, Batista G, Borges-Rosa J, Campos G, Fernandes C, Guimarães J, Martinho S, Santos T, Silva A, Simões M, Morais J, Carvalho M, Gonçalves C, Martins A, Vazão A, Abreu A, Alves da Silva P, Brito J, Pires ML, Lima Martins A, Pinto F, Pinto R, Aguiar C, Amador R, Bello R, Lima R, Pais J, Almeida A, Caria M, Carrington M, Conde D, Francisco C, Kongo K, Patricio L, Peralta M, Rebola E, Rocha R, Silva F, Trinca M. Global Variation in Lipoprotein(a) Levels Among Patients With Coronary Heart Disease. J Am Coll Cardiol 2025; 85:2028-2042. [DOI: 10.1016/j.jacc.2025.04.010] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/04/2025]
|
5
|
Cruz-Bautista I, Flores-Jurado Y, Roa-Álvarez G, Salas-Aldana M, Elías-Lopez DB, Hernández-Franco RF, Rosales-Uvera S, Vargas-Vázquez A, Valdez-Echeverría R, Luna Del Villar Velasco S, Muñoz-Hernández L, Mehta R, Morales-Esponda M, Aguilar-Panduro M, Chan-Puga G, Mota AS, Aguilar-Salinas CA. Lipoprotein (a) levels and clinical decision-making: data from a Mexican cohort at a tertiary medical institution. Lipids Health Dis 2025; 24:192. [PMID: 40420307 DOI: 10.1186/s12944-025-02610-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Approximately 20% of the global population has a Lp(a) concentrations above 50 mg/dL (> 125nmol/L), yet many remain unaware of the associated cardiovascular risks. In Mexico, routine measurement of Lp(a) is uncommon. This study aimed to investigate the frequency of Lp(a) testing, and the clinical actions taken by physicians upon detecting elevated Lp(a) concentrations in patients at a tertiary medical institution. METHODS Using an algorithm-based screening system, we reviewed the clinical and biochemical data of patients with Lp(a) measurements from 2019 to 2024. Data were retrieved from the laboratory information system and electronic health records. Complementary assessment data were obtained from the radiology and cardiology departments. RESULTS Of the 150,083 individuals evaluated at the institution, only 830 (0.5%) underwent Lp(a) testing, with testing rates increasing from 0.037% in 2019 to 0.24% in 2023. Elevated Lp(a) concentrations (> 50 mg/dL) were found in 21% of patients, and 2.2% had concentrations > 180 mg/dL. Patients with elevated Lp(a) had significantly higher rates of atherosclerotic cardiovascular disease (ASCVD) (p < 0.001) and familial hypercholesterolemia (p < 0.004) than those with lower Lp(a) levels. Interestingly, diabetes prevalence was higher in those with Lp(a) < 4 mg/dL (51.5% vs. 33.4%, p < 0.001). Despite the cardiovascular risk, only 26% of patients with elevated Lp(a) levels received interventions to modify risk factors. CONCLUSIONS Lp(a) testing was infrequent in a tertiary medical setting. Clinical interventions to modify cardiovascular risk factors were insufficient among patients with elevated Lp(a). These findings highlight the need for greater awareness among healthcare providers and the development of comprehensive screening and management algorithms to mitigate Lp(a) -related cardiovascular risk.
Collapse
Affiliation(s)
- Ivette Cruz-Bautista
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
- Departamento de Endocrinología y Metabolismo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Yuscely Flores-Jurado
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Guillermo Roa-Álvarez
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, México City, Mexico
| | - Mariana Salas-Aldana
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Daniel Benjamin Elías-Lopez
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
- Departamento de Endocrinología y Metabolismo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Ricardo Federico Hernández-Franco
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Sandra Rosales-Uvera
- Departamento de Radiología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Arsenio Vargas-Vázquez
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Raymundo Valdez-Echeverría
- Departamento de Laboratorio Central, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Sonia Luna Del Villar Velasco
- Departamento de Laboratorio Central, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Liliana Muñoz-Hernández
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
- CONAHCYT, Cuernavaca, Mexico
| | - Roopa Mehta
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
- Departamento de Endocrinología y Metabolismo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Mario Morales-Esponda
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Misael Aguilar-Panduro
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Guillermo Chan-Puga
- Departamento de Endocrinología y Metabolismo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Adrián Soto Mota
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, México City, Mexico
| | - Carlos Alberto Aguilar-Salinas
- Unidad de Investigación en Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico.
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, México City, Mexico.
| |
Collapse
|
6
|
van Bruggen FH, Diamond DM. Is Targeting LDL-C Levels Below 70 mg/dL Beneficial for Cardiovascular and Overall Health? A Critical Examination of the Evidence. J Clin Med 2025; 14:3569. [PMID: 40429563 PMCID: PMC12112069 DOI: 10.3390/jcm14103569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Revised: 05/17/2025] [Accepted: 05/18/2025] [Indexed: 05/29/2025] Open
Abstract
Over the past two decades, cardiovascular disease (CVD) prevention guidelines have progressively lowered LDL-C targets to <70 mg/dL for high-risk individuals based on the assumption of a linear relationship between LDL-C levels and CVD risk. However, the available evidence challenges this premise. Multiple studies demonstrate a weak or inconsistent association between LDL-C levels and atherosclerosis progression at the individual patient-level. Systematic reviews supporting the linearity assumption have notable limitations, including extrapolation beyond observed LDL-C ranges and potential ecological fallacy, as meta-regression analyses rely on study-level data, while patient-level data within the same trials often show no association between LDL-C reduction and CVD outcomes. Moreover, randomized controlled trials explicitly designed to assess LDL-C targets have yielded inconclusive and biased results. LDL-C itself is a heterogeneous marker, with particle size and composition influencing its atherogenicity. The cardiovascular benefits of lipid-lowering therapies may arise in part from pleiotropic effects unrelated to LDL-C lowering. Additionally, several studies indicate that higher LDL-C levels are paradoxically associated with longevity in elderly populations that is equal to or even greater than that of the general population. Collectively, this body of evidence raises questions about the validity of current LDL-C targets < 70 mg/dL in high-risk patients.
Collapse
Affiliation(s)
- Folkert H. van Bruggen
- Department of Primary and Long-Term Care, University Medical Centre Groningen, University of Groningen, P.O. Box 196, 9700 AD Groningen, The Netherlands
| | - David M. Diamond
- Department of Psychology, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
7
|
Poredoš P, Mangaroska AS, Poredoš P. Atherosclerotic plaque stabilization and regression. VASA 2025. [PMID: 40356553 DOI: 10.1024/0301-1526/a001202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Atherosclerotic plaques represent a typical deterioration of arterial wall in atherosclerotic process and are the source of cardiovascular events. Plaque progression and composition represent a major risk for cardiovascular events. Therefore, recently many studies have assessed changes in plaque characteristics and their response to various treatment modalities. In the last two decades, improvement in plaque imaging modalities that can assess plaque volumes and composition enable to follow plaque characteristics in a non-invasive way. Clinical trials utilizing arterial imaging modalities have shown that reducing LDL cholesterol to low levels can reduce atherosclerotic plaque burden and favourably modify plaque composition. These outcomes have been achieved with statin therapy and newer lipid-lowering strategies such as protein convertase subtilisin/kexin type 9 inhibitors. Also, some anti-inflammatory drugs and other anti-atherosclerotic medications can lead to significant reduction in plaque burden. However, the data assessing association of plaque regression to reduction of cardiovascular events are limited. Therefore, the aim of this narrative review is to elucidate the possibilities and the role of plaque assessment and if it might offer the potential to guide personalized management of patients at risk for cardiovascular events in the future.
Collapse
Affiliation(s)
- Pavel Poredoš
- Department of Vascular Disease, University Medical Centre Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Slovenia
| | | | - Peter Poredoš
- Faculty of Medicine, University of Ljubljana, Slovenia
- Department of Anaesthesiology and Surgical Intensive Care, University Medical Centre Ljubljana, Slovenia
| |
Collapse
|
8
|
Alebna PL, Ambrosio M, Martin M, Martey S, Spitz JA, Sharma G, Van Tassell B, Dixon DL, Hundley WG, Salloum FN, Mehta A. Association of Lipoprotein(a) with cardiovascular events among individuals with autoimmune conditions. Atherosclerosis 2025; 406:119244. [PMID: 40398292 DOI: 10.1016/j.atherosclerosis.2025.119244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Autoimmune conditions are associated with systemic inflammation, which elevates the risk of major adverse cardiovascular events (MACE). Systemic inflammation can increase plasma lipoprotein(a) [Lp(a)] levels by activating the interleukin-6 response element within the LPA gene promoter region. However, the association between elevated plasma Lp(a) and MACE risk in individuals with autoimmune conditions remains unclear. METHODS We analyzed data from 353,035 UK Biobank participants without cardiovascular disease, including 11,229 individuals with autoimmune conditions such as systemic lupus erythematosus, rheumatoid arthritis, psoriasis, multiple sclerosis, and others. Cox proportional hazards regression models and Kaplan-Meier survival curves assessed the association between elevated Lp(a) (≥125 nmol/L), autoimmune status, and time to MACE (myocardial infarction, stroke, cardiovascular death). RESULTS Over a median follow-up of 14 years, 19,091 MACEs occurred. Autoimmune conditions (HR, 1.30; 95 % CI, 1.20-1.41; P < 0.001) and elevated Lp(a) (HR, 1.24; 95 % CI, 1.18-1.30; P < 0.001) were independently associated with increased MACE risk. The interaction between autoimmune conditions and elevated Lp(a) was not significant (p = 0.40). Compared to participants with neither risk factor, those with both autoimmune conditions and elevated Lp(a) had the highest MACE risk (HR, 1.77; 95 % CI, 1.46-2.15; P < 0.001). Elevated MACE risk was also observed in individuals with only elevated Lp(a) (HR, 1.23; 95 % CI, 1.17-1.29; P < 0.001) or only autoimmune conditions (HR, 1.28; 95 % CI, 1.18-1.40; P < 0.001). CONCLUSION Autoimmune disease status and elevated Lp(a) level have an independent and additive joint association with MACE risk. This may have implications for Lp(a) lowering therapy use in high-risk primary prevention populations.
Collapse
Affiliation(s)
- Pamela L Alebna
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Mathew Ambrosio
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Maxwell Martin
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Sarah Martey
- School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jared A Spitz
- Inova Schar Heart and Vascular, Inova Health, Fairfax, VA, USA
| | - Garima Sharma
- Inova Schar Heart and Vascular, Inova Health, Fairfax, VA, USA
| | - Benjamin Van Tassell
- Department of Pharmacotherapy & Outcomes Science, VCU School of Pharmacy, Richmond, VA, USA
| | - Dave L Dixon
- Department of Pharmacotherapy & Outcomes Science, VCU School of Pharmacy, Richmond, VA, USA
| | - W Gregory Hundley
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Fadi N Salloum
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Anurag Mehta
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
9
|
Amaritei O, Mierlan OL, Gutu C, Gurau G. Lipoprotein(a): Assessing the Current Knowledge and Gaps in Screening and Treatment-A Narrative Review. J Cardiovasc Dev Dis 2025; 12:169. [PMID: 40422940 DOI: 10.3390/jcdd12050169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/11/2025] [Accepted: 04/23/2025] [Indexed: 05/28/2025] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) has long been screened using the traditional lipid profile, mainly focusing on LDL cholesterol. However, despite growing evidence supporting lipoprotein(a) [Lp(a)] as an independent risk factor involved in atherosclerosis, its clinical use remains limited. This review examines the reasons behind the limited use of Lp(a) screening in clinical practice, assessing its role in cardiovascular risk, comparing it to traditional lipid markers and evaluating current assessment methods. It also explores existing and emerging treatments, including gene-silencing therapies, for managing elevated Lp(a) levels. One in four clinicians does not routinely check Lp(a) levels, which proves a lack of awareness amongst them. The reasons for that are implied to be that the cost is too high and that available treatments are scarce. The traditional lipid profile, including LDL, high-density lipoprotein (HDL) and triglycerides, continues to be the gold standard for CV risk assessment. One limitation of using Lp(a) in clinical practice is the significant variability in apo(a) sizes, which results from the presence of multiple isoforms determined by the number of kringle domains. This structural diversity poses challenges in standardizing measurement methods, affecting the accuracy and comparability of results. While statins have a minimal impact on Lp(a), PCSK9-i lowers its levels by 20-25%, although this class is not prescribed primarily for this reason. Lastly, gene-silencing therapies, which achieve the greatest reduction in Lp(a) levels, are still in phase III trials, and there is still a need to examine whether this reduction translates into CV benefits. These limitations should not discourage further research, because ASCVD's complexity requires a more tailored approach. Current lipid-lowering therapy still fails in a minority of cases, as evidenced by new-onset cardiovascular events in patients with well-controlled LDL levels. There is a need for future interventional studies to assess whether a reduction in Lp(a) by PCSK9-i really translates into CV benefits, independent of LDL.
Collapse
Affiliation(s)
- Octavian Amaritei
- Faculty of Medicine and Pharmacy, "Dunărea de Jos" University of Galați, 800008 Galați, Romania
- "Sf. Andrei" Clinical Emergency County Hospital, 800578 Galați, Romania
| | - Oana Laura Mierlan
- Faculty of Medicine and Pharmacy, "Dunărea de Jos" University of Galați, 800008 Galați, Romania
- "Sf. Ioan" Emergency Clinical Pediatric Hospital, 800487 Galați, Romania
| | - Cristian Gutu
- Faculty of Medicine and Pharmacy, "Dunărea de Jos" University of Galați, 800008 Galați, Romania
- "Dr. Aristide Serfioti" Emergency Military Hospital, 800150 Galați, Romania
- Center for Research and Technology Transfer in the Medico-Pharmaceutical Field, "Dunărea de Jos" University of Galați, 800008 Galați, Romania
| | - Gabriela Gurau
- Faculty of Medicine and Pharmacy, "Dunărea de Jos" University of Galați, 800008 Galați, Romania
- "Sf. Ioan" Emergency Clinical Pediatric Hospital, 800487 Galați, Romania
- Center for Research and Technology Transfer in the Medico-Pharmaceutical Field, "Dunărea de Jos" University of Galați, 800008 Galați, Romania
| |
Collapse
|
10
|
Hoogeveen RC, Diffenderfer MR, Lim E, Liu CT, Ikezaki H, Guan W, Tsai MY, Ballantyne CM. Lipoprotein(a) and Risk of Incident Atherosclerotic Cardiovascular Disease: Impact of High-Sensitivity C-Reactive Protein and Risk Variability Among Human Clinical Subgroups. Nutrients 2025; 17:1324. [PMID: 40284189 PMCID: PMC12030245 DOI: 10.3390/nu17081324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Elevated lipoprotein(a) [Lp(a)] is associated with increased incidence of atherosclerotic cardiovascular disease (ASCVD). We aimed to assess the utility of Lp(a) as an ASCVD risk-enhancing factor, as recommended by the 2019 ACC/AHA guidelines on ASCVD primary prevention, and to determine whether C-reactive protein (CRP) modifies the association of elevated Lp(a) with ASCVD risk. Methods: Lp(a), high sensitivity CRP (hs-CRP), and other ASCVD risk factors, including blood lipids, blood pressure, diabetes status, body weight and height, and smoking, were measured in 15,933 participants (median age 61.7 years with 25th-75th percentiles 57-68 years, 56.7% female, 19.7% Black, free of ASCVD at baseline) in the Atherosclerosis Risk in Communities Study, Framingham Offspring Study, and Multi-Ethnic Study of Atherosclerosis. Participants were followed for 10 years for incident ASCVD (coronary heart disease (CHD) or stroke) and CHD (including angioplasty and/or coronary artery bypass but minus stroke). These endpoints occurred in 9.7% and 7.4% of subjects, respectively. Results: Compared with the lowest Lp(a) category (<10 mg/dL), the highest Lp(a) category (≥50 mg/dL) carried a significantly increased incidence of ASCVD (hazard ratio [HR] = 1.31; 95% confidence interval [CI] 1.15-1.50; p < 0.001) and CHD (HR = 1.49; 95%CI 1.27-1.75; p < 0.001). The association of elevated Lp(a) with incident ASCVD was stronger in males and non-Black individuals and was independent of diabetes status. Lp(a) levels ≥ 50 mg/dL predicted the 10-year ASCVD risk for those at intermediate risk (≥7.5%, HR = 1.32; 95%CI 1.15-1.52; p < 0.001). There was a significant interaction between Lp(a) and hs-CRP; individuals with concomitant elevated levels of Lp(a) and hs-CRP had the highest ASCVD risk. Conclusions: Elevated Lp(a) levels were associated with increased ASCVD risk, particularly in individuals with concomitantly elevated hs-CRP levels and those at intermediate 10-year ASCVD risk.
Collapse
Grants
- Contract HHSN268201700001I, HHSN268201700002I, HHSN268201700003I, HHSN268201700005I, and HHSN268201700004I from the National Institutes of Health. Contracts 75N92020D00001, HHSN268201500003I, N01-HC-95159, 75N92020D00005, N01-HC-95160, 75N92020D00002, N01 National Institutes of Health; National Center for Advancing Translation Sciences
Collapse
Affiliation(s)
- Ron C. Hoogeveen
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Margaret R. Diffenderfer
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA;
- Boston Heart Diagnostics, Framingham, MA 01702, USA
| | - Elise Lim
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA; (E.L.); (C.-T.L.)
- The Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA 01702, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA; (E.L.); (C.-T.L.)
- The Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA 01702, USA
| | - Hiroaki Ikezaki
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA;
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Weihua Guan
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN 55414, USA;
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Christie M. Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cardiovascular Disease Prevention, Debakey Heart and Vascular Disease Center, Houston, TX 77030, USA
| |
Collapse
|
11
|
Lopes Almeida Gomes L, Forman Faden D, Xie L, Chambers S, Stone C, Werth VP, Williams KJ. Modern therapy of patients with lupus erythematosus must include appropriate management of their heightened rates of atherosclerotic cardiovascular events: a literature update. Lupus Sci Med 2025; 12:e001160. [PMID: 40204295 PMCID: PMC11979607 DOI: 10.1136/lupus-2024-001160] [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: 12/31/2024] [Accepted: 03/13/2025] [Indexed: 04/11/2025]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the biggest killer of patients with lupus erythematosus (LE) and the general non-autoimmune population. In this literature update on LE and ASCVD, we focused on published work since our earlier review article, meaning from 2021 to the present, with an emphasis on cutaneous LE. Several themes emerged. First, new work shows that patients with lupus still exhibit a high burden of conventional risk factors for ASCVD events. Second, recent studies continue to implicate possible effects of lupus disease activity to worsen rates of ASCVD events beyond predictions from conventional risk factors. Third, new work on estimating the risk of future ASCVD events in patients with lupus supports arterial-wall imaging, inclusion of lupus-specific factors, estimators of ASCVD event risk that take lupus status into account and considering lupus as a diabetes equivalent or even as a diabetes-plus-smoking equivalent in this context. Technologies for arterial-wall imaging continue to improve and will likely play an increasing role in ASCVD assessment and management. Fourth, purported cardiovascular benefits from certain disease-modifying antirheumatic drugs such as antimalarials have become less clear. Fifth, earlier treatment of atherosclerosis, which is a lifelong disease, can be accomplished with diet, exercise, smoking cessation and new classes of safe and effective medications for lipid-lowering and blood pressure control. Benefits on subclinical arterial disease by imaging and on ASCVD events have been reported, supporting the concept that ASCVD is eminently manageable in this autoimmune condition. Sixth, despite the heightened risk for ASCVD events in patients with lupus, available therapeutic approaches remain unused or underused and, accordingly, event rates remain high.Raising awareness among patients and healthcare providers about ASCVD assessment and management in patients with LE is essential. Greater vigilance is needed to prevent ASCVD events in patients with lupus by addressing dyslipidaemias, hypertension, smoking, obesity and physical inactivity.
Collapse
Affiliation(s)
- Lais Lopes Almeida Gomes
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Dermatology, Corporal Michael J Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Daniella Forman Faden
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Dermatology, Corporal Michael J Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Lillian Xie
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Dermatology, Corporal Michael J Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Shae Chambers
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Dermatology, Corporal Michael J Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Caroline Stone
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Dermatology, Corporal Michael J Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Victoria P Werth
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Dermatology, Corporal Michael J Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Kevin Jon Williams
- Departments of Cardiovascular Sciences and Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Revankar S, Shakra N, DiMaio JM, Agarwala A. Key Concepts in Cardiovascular Secondary Prevention: A Case-Based Review. Am J Cardiol 2025; 248:32-40. [PMID: 40188902 DOI: 10.1016/j.amjcard.2025.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/12/2025] [Accepted: 03/30/2025] [Indexed: 04/26/2025]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) continues to be a growing global health concern with ischemic heart disease and stroke as leading causes of years of life lost. While aging is a major ASCVD risk factor, recent trends show a concerning rise in its incidence among younger adults driven, in part, by increased rates of risk factors such as hypertension and diabetes. These individuals with ASCVD are at elevated risk of recurrence years following their initial event, further underscoring the need for aggressive implementation of secondary prevention strategies to reduce morbidity and mortality. This case-based review discusses evidence-based pharmacological approaches to ASCVD secondary prevention-focusing on the roles of antiplatelets, lipid lowering therapies, antihypertensive medications, and glucose lowering treatments, in practical clinical settings.
Collapse
Affiliation(s)
- Shruti Revankar
- Department of Internal Medicine, Baylor College of Medicine, Houston, Texas
| | - Nezar Shakra
- Center for Cardiovascular Disease Prevention, Baylor Scott and White Health The Heart Hospital, Plano, Texas
| | - John Michael DiMaio
- Cardiovascular Department, Baylor Scott and White Health The Heart Hospital, Plano, Texas; Texas A & M Department of Biomedical Engineering, College Station, Texas; Cardiovascular Department, Baylor Scott and White Research Institute, Dallas, Texas
| | - Anandita Agarwala
- Center for Cardiovascular Disease Prevention, Baylor Scott and White Health The Heart Hospital, Plano, Texas.
| |
Collapse
|
13
|
Yadalam AK, Gangavelli A, Razavi AC, Ko YA, Alkhoder A, Haroun N, Lodhi R, Eldaidamouni A, Kasem MA, Quyyumi AA. Lipoprotein(a) Levels and Adverse Outcomes in Heart Failure. J Card Fail 2025:S1071-9164(25)00160-5. [PMID: 40189094 DOI: 10.1016/j.cardfail.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Although lipoprotein(a) [Lp(a)] level elevation is associated with new-onset heart failure (HF), it is unclear if elevated Lp(a) levels predict cardiovascular events in patients with chronic HF. Thus, we examined the association between Lp(a) levels and adverse cardiovascular outcomes in patients with HF. METHODS AND RESULTS A total of 1088 patients with HF undergoing cardiac catheterization at Emory-affiliated hospitals from 2004 to 2022 were divided into low (<30 mg/dL), intermediate (30-49 mg/dL), and high (≥50 mg/dL) Lp(a) groups. The primary outcome was the composite of cardiovascular death and HF hospitalization. Outcomes were assessed by Lp(a) group with competing risk modeling accounting for noncardiovascular death after adjustment for demographics, traditional cardiovascular risk factors, ejection fraction, ischemic HF etiology, and N-terminal prohormone of brain natriuretic peptide. Sensitivity analyses were performed to explore for heterogeneity of effect. The median age was 67 years, 34% were women, 18% were Black, 74% had ischemic HF, and 60% had an ejection fraction of ≤40%. During a median follow-up time of 4.3 years, 474 composite events (44%) occurred. When compared with participants with Lp(a) <30 mg/dL after multivariable adjustment, those with Lp(a) 30-49 mg/dL (subdistribution hazard ratio [sHR] 1.35, 95% confidence interval 1.04-1.76, P = .025) and Lp(a) ≥50 mg/dL (sHR 1.38, 95% confidence interval 1.11-1.72, P = .004) had a significantly higher risk of cardiovascular death or HF hospitalization. This relationship seemed to diminish over time and was nominally stronger in those with ischemic versus nonischemic HF (Pinteraction = .06), but did not meet significance after adjustment for multiple hypothesis testing. CONCLUSIONS In patients with HF, Lp(a) ≥30 mg/dL independently predicts the risk of cardiovascular death or HF hospitalization.
Collapse
Affiliation(s)
- Adithya K Yadalam
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Apoorva Gangavelli
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Alexander C Razavi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Yi-An Ko
- Rollins School of Public Health, Department of Biostatistics and Bioinformatics Emory University, Atlanta, Georgia
| | - Ayman Alkhoder
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Nisreen Haroun
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Rafia Lodhi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Ahmed Eldaidamouni
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Mahmoud Al Kasem
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A Quyyumi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
14
|
Zhang P, Wang W, Xu Q, Cui J, Zhu M, Li Y, Liu Y, Liu Y. Genetic association of circulating lipids and lipid-lowering drug targets with vascular calcification. Atherosclerosis 2025; 403:119136. [PMID: 39985880 DOI: 10.1016/j.atherosclerosis.2025.119136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/27/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND AND AIMS Vascular calcification (VC) significantly increases the incidence and mortality of many diseases. The causal relationships of dyslipidaemia and lipid-lowering drug use with VC severity remain unclear. This study explores the genetic causal associations of different circulating lipids and lipid-lowering drug targets with coronary artery calcification (CAC) and abdominal aortic artery calcification (AAC). METHODS We obtained single-nucleotide polymorphisms (SNPs) and expression quantitative trait loci (eQTLs) associated with seven circulating lipids and 13 lipid-lowering drug targets from publicly available genome-wide association studies and eQTL databases. Causal associations were investigated by univariable, multivariable, drug-target, and summary data-based Mendelian randomization (MR) analyses. Potential mediation effects of metabolic risk factors were evaluated. RESULTS MR analysis revealed that genetic proxies for low-density lipoprotein cholesterol (LDL-C), triglycerides (TC) and Lipoprotein (a) (Lp(a)) were causally associated with CAC severity, and apolipoprotein B (apoB) level was causally associated with AAC severity. A significant association was detected between hepatic Lipoprotein(A) (LPA) gene expression and CAC severity. Colocalisation analysis supported the hypothesis that the association between LPA expression and CAC quantity is driven by different causal variant sites within the ±1 Mb flanking region of LPA. Serum calcium and phosphorus had causal associations with CAC severity. CONCLUSIONS Inhibitors targeting LPA might represent CAC drug candidates. Moreover, T2DM, hypercalcemia, and hyperphosphatemia are positively causally associated with CAC severity, while chronic kidney disease and estimated glomerular filtration rate are not.
Collapse
Affiliation(s)
- Pengfei Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Wenting Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Qian Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Jing Cui
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Mengmeng Zhu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yiwen Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanfei Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China; The Second Department of Gerontology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yue Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
15
|
Fujii E, Ako J, Takahashi Y, Toda M, Iekushi K, Yamashita S. Serum Lipoprotein(a) Levels and Their Association with Atherosclerotic Cardiovascular Disease in Japan. J Atheroscler Thromb 2025; 32:421-438. [PMID: 39370270 PMCID: PMC11973527 DOI: 10.5551/jat.64953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/19/2024] [Indexed: 10/08/2024] Open
Abstract
AIMS To investigate the distribution of lipoprotein(a) (Lp(a)) and its association with atherosclerotic cardiovascular disease (ASCVD) in Japanese patients at high risk for ASCVD using a health insurance database. METHODS Between July 2013 and June 2021, patients eligible for ASCVD prevention according to the 2017 Japan Atherosclerosis Society (JAS) guidelines with documented Lp(a) test results were extracted from the Medical Data Vision claims database and divided into three groups: primary prevention high-risk (Group I), secondary prevention (Group II) and secondary prevention high-risk (Group III). Data on lipid levels, cardiovascular morbidity risk factors and lipid-lowering treatments were extracted. RESULTS Of 700,580 patients with documented low-density lipoprotein cholesterol (LDL-C), 2,967 (0.42%) were tested for Lp(a). In 2,170 eligible patients, the median [interquartile range] serum concentration of Lp(a) was 13.9 [7.5-24.6] mg/dL, with 151 patients (7.0%) above the recommended risk threshold of ≥ 50 mg/dL. Lp(a) levels increased with risk across all prevention groups. Being in the highest Lp(a) quintile (Q5) was associated with an increased frequency of ASCVD (28.9% versus 18.9% in the lowest quintile (Q1) for unstable angina; 18.7% versus 10.1% for myocardial infarction; 27.9% versus 17.0% for ischemic stroke). In the secondary prevention groups, the proportion of patients meeting an LDL-C target of <70 mg/dL decreased from 30.2% in Q1 to 19.0% in Q5 for Group II and from 32.9% to 16.3% for Group III. CONCLUSIONS Despite a high prevalence of Lp(a) ≥ 50mg/dL in Japanese patients at high risk for ASCVD, it found that the Lp(a) testing rate was very low.
Collapse
Affiliation(s)
- Emi Fujii
- Medical Affairs Division, Novartis Pharma K.K
| | - Junya Ako
- Department of Cardiovascular Medicine, Kitasato University School of Medicine
| | | | | | | | | |
Collapse
|
16
|
Girard AS, Paulin A, Manikpurage HD, Lajeunesse E, Clavel MA, Pibarot P, Krege JH, Mathieu P, Thériault S, Arsenault BJ. Impact of Lipoprotein(a) on Valvular and Cardiovascular Outcomes in Patients With Calcific Aortic Valve Stenosis. J Am Heart Assoc 2025; 14:e038955. [PMID: 40079323 DOI: 10.1161/jaha.124.038955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/16/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Lp(a) (lipoprotein(a)) is an independent risk factor for calcific aortic valve stenosis (CAVS). Whether patients with CAVS and high Lp(a) levels are at higher risk of valvular or cardiovascular events is unknown. The aim of this study is to determine whether higher Lp(a) levels are associated with valvular and cardiovascular outcomes in patients with CAVS. METHODS AND RESULTS We identified 1962 patients from the UK Biobank with an electronic health record or self-reported CAVS diagnosis but who did not previously undergo aortic valve replacement (AVR) and had a minimal follow-up time of 2.5 years. Cox proportional hazard regression was used to evaluate the effect of Lp(a) on AVR, AVR or cardiac death, and valvular or cardiovascular events (AVR, cardiac death, myocardial infarction, stroke, heart failure, or coronary artery bypass grafting). The maximal follow-up time was set to 5 years. During the follow-up, 198 patients underwent AVR, 260 had AVR or cardiac death, and 435 had at least 1 valvular or cardiovascular event. Patients with Lp(a) levels ≥125 versus <125 nmol/L were at higher risk of AVR (hazard ratio [HR], 1.58 [95% CI, 1.17-2.12]), AVR or cardiac death (HR, 1.43 [95% CI, 1.10-1.86]), and cardiovascular or valvular events (HR, 1.36 [95% CI, 1.11-1.68]). Point estimates were comparable in men versus women, younger versus older patients, and in patients with higher versus lower plasma C-reactive protein levels. CONCLUSIONS In patients with CAVS, Lp(a) levels predicted a higher risk of valvular and cardiovascular outcomes. The impact of Lp(a)-lowering therapies on valvular and cardiovascular health should be assessed in a long-term randomized clinical trial.
Collapse
Affiliation(s)
- Arnaud S Girard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Audrey Paulin
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Hasanga D Manikpurage
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Emma Lajeunesse
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Marie-Annick Clavel
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Medicine, Faculty of Medicine Université Laval Québec QC Canada
| | - Philippe Pibarot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Medicine, Faculty of Medicine Université Laval Québec QC Canada
| | | | - Patrick Mathieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Surgery, Faculty of Medicine Université Laval Québec QC Canada
| | - Sébastien Thériault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine Université Laval Québec QC Canada
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Medicine, Faculty of Medicine Université Laval Québec QC Canada
| |
Collapse
|
17
|
Sherrill C, Ehrhardt-Humbert L, Salem D. The Role of Lipoprotein (a) in the Progression of Aortic Stenosis. Am J Med 2025:S0002-9343(25)00138-X. [PMID: 40049573 DOI: 10.1016/j.amjmed.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025]
Abstract
Calcific aortic stenosis is a complex process that involves several factors, including genetics, lipid infiltration, inflammation, and hemodynamic stress. This process begins at an early age and progresses throughout one's lifetime. Lipoprotein (a), or Lp(a), has been shown to drive progression of aortic stenosis in several studies. Additionally, analyses have demonstrated that a higher serum level of Lp(a) is associated with earlier onset of aortic stenosis and faster progression of mean pressure across the aortic valve. The current cornerstone medications to lower low-density lipoprotein do not have a meaningful impact on Lp(a) levels. In this article, we will review current therapeutics that are in development to lower serum Lp(a), including several that involve ribonucleic acid modification. Although these novel therapeutics are promising, it is to be determined whether lowering serum Lp(a) correlates to reduced calcific aortic stenosis and improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Clay Sherrill
- Department of Medicine, Tufts Medical Center, Boston, Mass
| | | | - Deeb Salem
- Center for Cardiovascular Disease, Department of Medicine, Tufts Medical Center, Boston, Mass
| |
Collapse
|
18
|
Ramírez-Cortés F, Ménová P. Hepatocyte targeting via the asialoglycoprotein receptor. RSC Med Chem 2025; 16:525-544. [PMID: 39628900 PMCID: PMC11609720 DOI: 10.1039/d4md00652f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
This review highlights the potential of asialoglycoprotein receptor (ASGPR)-mediated targeting in advancing liver-specific treatments and underscores the ongoing progress in the field. First, we provide a comprehensive examination of the nature of ASGPR ligands, both natural and synthetic. Next, we explore various drug delivery strategies leveraging ASGPR, with a particular emphasis on the delivery of therapeutic nucleic acids such as small interfering RNAs (siRNAs) and antisense oligonucleotides (ASOs). An in-depth analysis of the current status of RNA interference (RNAi) and ASO-based therapeutics is included, detailing approved therapies and those in various stages of clinical development (phases 1 to 3). Afterwards, we give an overview of other ASGPR-targeted conjugates, such as those with peptide nucleic acids or aptamers. Finally, targeted protein degradation of extracellular proteins through ASGPR is briefly discussed.
Collapse
Affiliation(s)
| | - Petra Ménová
- University of Chemistry and Technology, Prague Technická 5 16628 Prague 6 Czech Republic
| |
Collapse
|
19
|
Greco A, Finocchiaro S, Spagnolo M, Faro DC, Mauro MS, Raffo C, Sangiorgio G, Imbesi A, Laudani C, Mazzone PM, Ammirabile N, Giacoppo D, Landolina D, Capodanno D. Lipoprotein(a) as a Pharmacological Target: Premises, Promises, and Prospects. Circulation 2025; 151:400-415. [PMID: 39928714 DOI: 10.1161/circulationaha.124.069210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 10/29/2024] [Indexed: 02/12/2025]
Abstract
Atherosclerotic cardiovascular disease is a major health concern worldwide and requires effective preventive measures. Lp(a) (lipoprotein [a]) has recently garnered attention as an independent risk factor for astherosclerotic cardiovascular disease, with proinflammatory and prothrombotic mechanisms contributing to its atherogenicity. On an equimolar basis, Lp(a) is ~5 to 6 times more atherogenic than particles that have been widely associated with adverse cardiovascular outcomes, such as LDL (low-density lipoprotein). Lp(a) can enter the vessel wall, leading to the accumulation of oxidized phospholipids in the arterial intima, which are crucial for initiating plaque inflammation and triggering vascular disease progression. In addition, Lp(a) may cause atherothrombosis through interactions between apoA (apolipoprotein A) and the platelet PAR-1 (protease-activated receptor 1) receptor, as well as competitive inhibition of plasminogen. Because Lp(a) is mostly determined on genetic bases, a 1-time assessment in a lifetime can suffice to identify patients with elevated levels. Mendelian randomization studies and post hoc analyses of randomized trials of LDL cholesterol-lowering drugs showed a causal link between Lp(a) concentrations and cardiovascular outcomes, with therapeutic reduction of Lp(a) expected to contribute to estimated cardiovascular risk mitigation. Many Lp(a)-lowering drugs, including monoclonal antibodies, small interfering ribonucleic acids, antisense oligonucleotides, small molecules, and gene editing compounds, are at different stages of clinical investigation and show promise for clinical use. In particular, increased Lp(a) testing and treatment are expected to have a substantial impact at the population level, enabling the identification of high-risk individuals and the subsequent prevention of a large number of cardiovascular events. Ongoing phase 3 trials will further elucidate the cardiovascular benefits of Lp(a) reduction over the long term, offering potential avenues for targeted interventions and improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Antonio Greco
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Simone Finocchiaro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Marco Spagnolo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Denise Cristiana Faro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Maria Sara Mauro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Carmelo Raffo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Giuseppe Sangiorgio
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Antonino Imbesi
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Claudio Laudani
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Placido Maria Mazzone
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Nicola Ammirabile
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Daniele Giacoppo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Davide Landolina
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| |
Collapse
|
20
|
Lemešić DL, Šimičević L, Ganoci L, Gelemanović A, Šućur N, Pećin I. Association of rs3798220 Polymorphism with Cardiovascular Incidents in Individuals with Elevated Lp(a). Diagnostics (Basel) 2025; 15:404. [PMID: 40002555 PMCID: PMC11854902 DOI: 10.3390/diagnostics15040404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Lipoprotein (a) [Lp(a)] plays a significant role in atherosclerosis and cardiovascular disease (CVD). Genetic regulation of Lp(a) involves variations in the apo(a) LPA gene, as specific polymorphisms like rs10455872 and rs3798220, both linked to higher Lp(a) levels and CVD. CVD remains the leading global cause of death, with high Lp(a) levels increasingly recognized as a significant factor in younger patients with no other CVD risk factors. We aimed to evaluate the association of LPA genetic variations with Lp(a) levels and its effect on cardiovascular risk as there are existing inconsistent findings. Methods: This case-control study included 251 subjects with a median age of 52 years (interquartile range, IQR = 17) and elevated Lp(a) levels. Cases were subjects who experienced early cardiovascular incidents (women < 65, men < 55 years old), and the control group included subjects without such history. Genotyping of LPA gene polymorphisms (rs10455872 and rs3798220) was performed, and demographic data with Lp(a) levels were collected. To evaluate the association between the LPA genotypes and the risk of cardiovascular incidents (CVI), several logistic regression models were performed. The cut-off points for Lp(a) levels were determined using diagnostic test accuracy measures. Results: The rs3798220-C allele was associated with higher Lp(a) levels (288 ± 166 nmol/L in cases vs. 189 ± 102 nmol/L in controls, p < 0.001) and myocardial infarction (53% in cases vs. 36% in controls, p = 0.036). Among cases, 28.9% carried the rs3798220-C allele, compared to 18.7% in controls. The rs10455872-G allele was slightly more prevalent in controls (34.15% vs. 29.69%) but without further significant associations. In this study, the cut-off Lp(a) value of 151 nmol/L, for patients with a positive family history of early CVD, is associated with a higher chance of developing CVI. Conclusions: This study demonstrates an association between the LPA rs3798220-C allele and higher Lp(a) levels, as well as an increased risk of early onset myocardial infarction. However, the obtained association should further be evaluated at a much larger scale.
Collapse
Affiliation(s)
- Dunja Leskovar Lemešić
- Division for Metabolic Diseases, Department of Internal Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia; (N.Š.); (I.P.)
| | - Livija Šimičević
- Division for Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, 10000 Zagreb, Croatia;
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Lana Ganoci
- Division for Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, 10000 Zagreb, Croatia;
- Department of Basic and Clinical Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Andrea Gelemanović
- Mediterranean Institute for Life Sciences (MedILS), University of Split, 21000 Split, Croatia;
| | - Nediljko Šućur
- Division for Metabolic Diseases, Department of Internal Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia; (N.Š.); (I.P.)
| | - Ivan Pećin
- Division for Metabolic Diseases, Department of Internal Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia; (N.Š.); (I.P.)
- Department of Internal Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
21
|
Wissel S, Scharnagl H, Kleber ME, Delgado G, Moissl A, Krämer B, März W. Inflammation, Lp(a) and cardiovascular mortality: results from the LURIC study. Clin Res Cardiol 2025:10.1007/s00392-025-02609-4. [PMID: 39912915 DOI: 10.1007/s00392-025-02609-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
OBJECTIVE Lipoprotein(a) [Lp(a)] concentrations have been associated with cardiovascular risk. Recent studies suggested an interaction between systemic inflammation assessed via high-sensitivity C-reactive protein (hsCRP) and Lp(a). This study aimed to evaluate whether Lp(a), hsCRP, and interleukin-6 (IL-6) levels are associated with cardiovascular mortality in a German hospital-based cohort. METHODS Data were drawn from the Ludwigshafen Risk and Cardiovascular Health (LURIC) study, including 3,316 patients undergoing coronary angiography. Lp(a) was measured by immunoturbidimetry and categorized into three strata (< 50 mg/dL, n = 2668; 50-100 mg/dL, n = 482; > 100 mg/dL, n = 163). HsCRP was measured by immunonephelometry and categorized by intervals (1st < 1 mg/L, 2nd 1-2 mg/L and 3rd interval > 2 mg/L). IL-6 was measured by ELISA and categorized into two groups (1st < 3.2 ng/L, 2nd ≥ 3.2 ng/L). The primary outcome was cardiovascular disease (CVD) mortality, analyzed using Cox proportional hazards models and logistic regression. RESULTS Participants were predominantly male, with a mean age of 62.6 years. Extremely high Lp(a) (> 100 mg/dL) was associated with increased cardiovascular mortality (HR 1.5, 95% CI 1.06-2.12) compared to Lp(a) < 50 mg/dl. Both hsCRP (> 2 mg/L, HR 1.39, 95% CI 1.08-1.79 third vs. first interval) and more so IL-6 (HR 1.92, 95% CI 1.64-2.23, upper vs. lower half), were independently associated with higher CVD mortality. While hsCRP did not increase the Lp(a)-CVD mortality in stratified analysis, high IL-6 conferred an increased risk at Lp(a) levels > 100 mg/dL (HR 1.25, 95% CI 1.09-1.44). CONCLUSION HsCRP and IL-6 are associated with cardiovascular mortality. Markedly elevated Lp(a) is associated with an increased risk of cardiovascular mortality in the context of high systemic inflammation. Anti-inflammatory treatments may provide additional therapeutic benefits for individuals with high Lp(a).
Collapse
Affiliation(s)
- Stephanie Wissel
- Department of Medicine I, University of Würzburg, Würzburg, Germany.
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Marcus E Kleber
- Department of Medicine I, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Synlab Center of Human Genetics, SYNLAB Holding Deutschland GmbH, Mannheim, Germany
| | - Graciela Delgado
- Department of Medicine I, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Angela Moissl
- Department of Medicine I, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Bernhard Krämer
- Department of Medicine I, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Department of Medicine I, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany
| |
Collapse
|
22
|
Ntaios G, Dalakoti M. Treat the patient, not the disease: The embolic stroke of undetermined source as an opportunity to optimize cardiovascular prevention in a holistic approach. Eur J Intern Med 2025; 132:9-17. [PMID: 39443247 DOI: 10.1016/j.ejim.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
For any physician treating a patient with a medical condition of unclear etiology, the differential diagnosis aims to identify the actual most probable cause among various potential etiologies, in order to tailor treatment options. In patients with embolic stroke of undetermined source (ESUS), this can be challenging due to the frequent presence of multiple potential embolic sources, raising difficulties to identify the most likely cause. Additionally, despite targeted preventive measures for the presumed embolic source, patients may remain at risk for stroke and cardiovascular events due to other unrecognized or underestimated pathologies. The multi-level complexity and multimorbidity typically associated with ESUS, represents a challenge that requires broad knowledge of the cardiovascular pathophysiology, deep expertise of the available diagnostic and therapeutic options, and interdisciplinary approach. At the same time, it is an ideal opportunity to assess thoroughly the overall cardiovascular status of the patient, which in turn can allow us to optimize therapeutic and preventive strategies in a holistic approach, and prevent future strokes, cardiovascular events and disability through different parallel pathways. In this context, rather than narrowing our perspective on identifying the specific embolic source presumed to be the most likely cause of ESUS, it is crucial to shift our focus from the disease to the patient, and evaluate the overall cardiovascular profile by assessing the risk of all cardiovascular comorbidities present, no matter if causally associated with ESUS or not. In order to bring across these points and more, this article is centred around a clinical case that serves as a starting point to illustrate the holistic approach to the management of patients with ESUS. After all, this is the beauty, the magic and the art of Internal Medicine: to treat the patient, not the disease, the system or the organ.
Collapse
Affiliation(s)
- George Ntaios
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa 41110, Greece.
| | - Mayank Dalakoti
- Cardiovascular Metabolic Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Cardiology, National University Heart Centre, National University Health System, Singapore
| |
Collapse
|
23
|
Kulus M, Farzaneh M, Sheykhi-Sabzehpoush M, Ghaedrahmati F, Mehravar F, Józkowiak M, Piotrowska-Kempisty H, Bukowska D, Antosik P, Podhorska-Okołów M, Zabel M, Mozdziak P, Dzięgiel P, Kempisty B. Exosomes and non-coding RNAs: Exploring their roles in human myocardial dysfunction. Biomed Pharmacother 2025; 183:117853. [PMID: 39827809 DOI: 10.1016/j.biopha.2025.117853] [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: 09/07/2024] [Revised: 12/29/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Myocardial dysfunction, characterized by impaired cardiac muscle function, arises from diverse etiologies, including coronary artery disease, myocardial infarction, cardiomyopathies, hypertension, and valvular heart disease. Recent advancements have highlighted the roles of exosomes and non-coding RNAs in the pathophysiology of myocardial dysfunction. Exosomes are small extracellular vesicles released by cardiac and other cells that facilitate intercellular communication through their molecular cargo, including ncRNAs. ncRNAs are known to play critical roles in gene regulation through diverse mechanisms, impacting oxidative stress, fibrosis, and other factors associated with myocardial dysfunction. Dysregulation of these molecules correlates with disease progression, presenting opportunities for therapeutic interventions. This review explores the mechanistic interplay between exosomes and ncRNAs, underscoring their potential as biomarkers and therapeutic agents in myocardial dysfunction. Emerging evidence supports the use of engineered exosomes and modified ncRNAs to enhance cardiac repair by targeting signaling pathways associated with fibrosis, apoptosis, and angiogenesis. Despite promising preclinical results, delivery, stability, and immunogenicity challenges remain. Further research is needed to optimize clinical translation. Understanding these intricate mechanisms may drive the development of innovative strategies for diagnosing and treating myocardial dysfunction, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun 87-100, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Mehravar
- Department of Biostatistics and Epidemiology, School of Health, Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Małgorzata Józkowiak
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland; Department of Toxicology, Poznan University of Medical Sciences, Poznan, Poland
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, Poznan, Poland; Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun 87-100, Poland
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun 87-100, Poland
| | - Marzenna Podhorska-Okołów
- Department of Human Morphology and Embryology, Division of Ultrastructure Research, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Maciej Zabel
- Division of Anatomy and Histology, University of Zielona Góra, Zielona Góra 65-046, Poland; Department of Human Morphology and Embryology, Division of Histology and Embryology, Wroclaw Medical University, Wroclaw 50-368, Poland
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA; Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA
| | - Piotr Dzięgiel
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wroclaw Medical University, Wroclaw 50-368, Poland; Department of Physiotherapy, Wroclaw University School of Physical Education, Wroclaw, Poland
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun 87-100, Poland; Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland; Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA; Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czech Republic.
| |
Collapse
|
24
|
Hafiane A, Pisaturo A, Favari E, Bortnick AE. Atherosclerosis, calcific aortic valve disease and mitral annular calcification: same or different? Int J Cardiol 2025; 420:132741. [PMID: 39557087 DOI: 10.1016/j.ijcard.2024.132741] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
There are similarities in the pathophysiologic mechanisms of atherosclerosis, calcific aortic valve disease (CAVD) and mitral annular calcification (MAC), however, medical treatment to slow or stop the progression of CAVD or MAC has been more elusive as compared to atherosclerosis. Atherosclerosis and CAVD share common demographic, clinical, protein, and genetic factors even more so than with MAC, which supports the possibility of shared medical therapies, though abrogating calcific extracellular vesicle shedding could be a common target for all three conditions. Herein, we summarize the overlapping and distinct pathways for further investigation, as well as key areas where additional research is needed.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Institute of the McGill University Health Centre, McGill University, Montreal, Canada.
| | | | - Elda Favari
- Department of Food and Drug, University of Parma, Parma, Italy.
| | - Anna E Bortnick
- Department of Medicine, Divisions of Cardiology and Geriatrics, and Department of Obstetrics & Gynecology and Women's Health, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, United States of America.
| |
Collapse
|
25
|
Bhatia HS, Wandel S, Willeit P, Lesogor A, Bailey K, Ridker PM, Nestel P, Simes J, Tonkin A, Schwartz GG, Colhoun H, Wanner C, Tsimikas S. Independence of Lipoprotein(a) and Low-Density Lipoprotein Cholesterol-Mediated Cardiovascular Risk: A Participant-Level Meta-Analysis. Circulation 2025; 151:312-321. [PMID: 39492722 PMCID: PMC11771346 DOI: 10.1161/circulationaha.124.069556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDL-C) and lipoprotein(a) (Lp[a]) levels are independently associated with atherosclerotic cardiovascular disease (ASCVD). However, the relationship between Lp(a) level, LDL-C level, and ASCVD risk at different thresholds is not well defined. METHODS A participant-level meta-analysis of 27 658 participants enrolled in 6 placebo-controlled statin trials was performed to assess the association of LDL-C and Lp(a) levels with risk of fatal or nonfatal coronary heart disease events, stroke, or any coronary or carotid revascularization (ASCVD). The multivariable-adjusted association between baseline Lp(a) level and ASCVD risk was modeled continuously using generalized additive models, and the association between baseline LDL-C level and ASCVD risk by baseline Lp(a) level by Cox proportional hazards models with random effects. The joint association between Lp(a) level and statin-achieved LDL-C level with ASCVD risk was evaluated using Cox proportional hazards models. RESULTS Compared with an Lp(a) level of 5 mg/dL, increasing levels of Lp(a) were log-linearly associated with ASCVD risk in statin- and placebo-treated patients. Among statin-treated individuals, those with Lp(a) level >50 mg/dL (≈125 nmol/L) had increased risk across all quartiles of achieved LDL-C level and absolute change in LDL-C level. Even among those with the lowest quartile of achieved LDL-C level (3.1-77.0 mg/dL), those with Lp(a) level >50 mg/dL had greater ASCVD risk (hazard ratio, 1.38 [95% CI, 1.06-1.79]) than those with Lp(a) level ≤50 mg/dL. The greatest risk was observed with both Lp(a) level >50 mg/dL and LDL-C level in the fourth quartile (hazard ratio, 1.90 [95% CI, 1.46-2.48]). CONCLUSIONS These findings demonstrate the independent and additive nature of Lp(a) and LDL-C levels for ASCVD risk, and that LDL-C lowering does not fully offset Lp(a)-mediated risk.
Collapse
Affiliation(s)
- Harpreet S. Bhatia
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (H.S.B., S.T.)
| | - Simon Wandel
- Novartis Pharma AG, Basel, Switzerland (S.W., A.L., K.B.)
| | - Peter Willeit
- Department of Medical Statistics, Informatics and Health Economics at the Medical University of Innsbruck, Austria (P.W.)
| | | | - Keith Bailey
- Novartis Pharma AG, Basel, Switzerland (S.W., A.L., K.B.)
| | - Paul M. Ridker
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (P.M.R.)
| | - Paul Nestel
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (P.N.)
| | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, NSW, Australia (J.S.)
| | - Andrew Tonkin
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia (A.T.)
| | - Gregory G. Schwartz
- Division of Cardiology, Rocky Mountain Regional VA Medical Center and University of Colorado School of Medicine, Aurora (G.G.S.)
| | - Helen Colhoun
- MRC Human Genetics Unit, Centre for Genomic and Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, Edinburgh, UK (H.C.)
| | - Christoph Wanner
- Division of Nephrology, Department of Internal Medicine and Comprehensive Heart Failure Centre, University Hospital of Würzburg, Germany (C.W.)
| | - Sotirios Tsimikas
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (H.S.B., S.T.)
| |
Collapse
|
26
|
Mousavi I, Suffredini J, Virani SS, Ballantyne CM, Michos ED, Misra A, Saeed A, Jia X. Early-onset atherosclerotic cardiovascular disease. Eur J Prev Cardiol 2025; 32:100-112. [PMID: 39041374 DOI: 10.1093/eurjpc/zwae240] [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: 05/09/2024] [Revised: 06/24/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Recent trends indicate a concerning increase in early-onset atherosclerotic cardiovascular disease (ASCVD) among younger individuals (men aged <55 years women aged <65 years). These findings highlight the pathobiology of ASCVD as a disease process that begins early in life and underscores the need for more tailored screening methods and preventive strategies. Increasing attention has been placed on the growing burden of traditional cardiometabolic risk factors in young individuals while also recognizing unique factors that mediate risk of pre-mature atherosclerosis in this demographic such as substance use, socioeconomic disparities, adverse pregnancy outcomes, and chronic inflammatory states that contribute to the increasing incidence of early ASCVD. Additionally, mounting evidence has pointed out significant disparities in the diagnosis and management of early ASCVD and cardiovascular outcomes based on sex and race. Moving towards a more personalized approach, emerging data and technological developments using diverse tools such as polygenic risk scores and coronary artery calcium scans have shown potential in earlier detection of ASCVD risk. Thus, we review current evidence on causal risk factors that drive the increase in early ASCVD and highlight emerging tools to improve ASCVD risk assessment in young individuals.
Collapse
Affiliation(s)
- Idine Mousavi
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - John Suffredini
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Salim S Virani
- Office of the Vice Provost, Research, The Aga Khan University, Karachi, Pakistan
- Section of Cardiology, Department of Medicine, Baylor College of Medicine and Texas Heart Institute, Houston, TX, USA
| | - Christie M Ballantyne
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Erin D Michos
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Arunima Misra
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Anum Saeed
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Xiaoming Jia
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
27
|
Ogieuhi IJ, Callender K, Odukudu GDO, Obi ES, Muzofa K, Babalola AE, Ugiomoh OMA, Umenzeakor KH, Akingbola A, Ayoson CO, Agbo EU, Odoeke MC. Antisense Oligonucleotides in Dyslipidemia Management: A Review of Clinical Trials. High Blood Press Cardiovasc Prev 2025; 32:33-47. [PMID: 39476283 DOI: 10.1007/s40292-024-00682-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/06/2024] [Indexed: 01/31/2025] Open
Abstract
INTRODUCTION Elevated serum total cholesterol levels, very low-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides, or a decreased serum high-density lipoprotein cholesterol concentration characterize dyslipidemia. Antisense Oligonucleotide therapy in dyslipidemia targets apolipoprotein B (ApoB), an essential component of low-density lipoprotein (LDL) associated with atherosclerosis development. AIM This review aims to critically evaluate the efficacy and safety of this group of medications in mitigating dyslipidemia in at-risk individuals and its potential role in advancing personalized medicine in the management of dyslipidemias. METHODS A detailed search was conducted from multiple databases adhering to the PRISMA guidelines. Clinical trials and randomized controlled trials on antisense oligonucleotides for management of dyslipidemias were included, excluding non-English studies, case reports and all forms of reviews. Data was screened, with duplicates removed, and key findings were synthesized using a narrative approach. RESULTS AND CONCLUSION The potential of antisense oligonucleotides (ASOs) to treat dyslipidemia and other disorders has attracted much interest. Several studies and clinical trials have been conducted on the safety and tolerability of ASOs for dyslipidemia. Although statins are the mainstay management of hypercholesterolemia, there is evidence from clinical trials that ASOs can even be more effective with little to no side effects. Novel therapeutic approaches such as antisense oligonucleotides (ASOs) offer tailored therapeutic alternatives. ASOs such as Mipomersen and Volanesorsen provide additional treatment options for patients with inherited lipid abnormalities by lowering certain atherogenic lipoproteins such as apo B and ApoC-III, respectively.
Collapse
Affiliation(s)
| | - Kristen Callender
- Queen Elizabeth Hospital, Martindales Road, Bridgetown, St. Michael, Barbados
| | | | | | | | | | | | | | - Adewunmi Akingbola
- Department of Public Health, University of Cambridge, Cambridge, England, UK
| | | | | | | |
Collapse
|
28
|
Daghlas I, Gill D. Leveraging Mendelian randomization to inform drug discovery and development for ischemic stroke. J Cereb Blood Flow Metab 2024:271678X241305916. [PMID: 39628323 PMCID: PMC11615907 DOI: 10.1177/0271678x241305916] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/24/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024]
Abstract
Discovery and development of efficacious and safe pharmacological therapies is fraught with challenges. As proteins constitute the majority of drug targets and are encoded by genes, naturally occurring genetic variation within populations can provide valuable insights to inform drug discovery and development efforts. The drug target Mendelian randomization (MR) paradigm leverages these principles to investigate the causal effects of drug targets in humans. This review examines the application of drug target MR in informing the efficacy and development of therapeutics for ischemic stroke prevention and treatment. We consider applications of MR for existing and novel treatment strategies, including targeting blood pressure, lipid metabolism, coagulation, inflammation and glycemic control. Several of these genetically supported targets are under evaluation in late-stage clinical trials. Methodological limitations of drug target MR are addressed, followed by an outline of future research directions. We anticipate that careful application of drug target MR will enhance the efficiency of drug development for ischemic stroke, consequently accelerating the delivery of effective medications to patients.
Collapse
Affiliation(s)
- Iyas Daghlas
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
29
|
Masana L, Ibarretxe D. New drugs for treating dyslipidemias. From small molecules to small interfering RNAs. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36 Suppl 1:S15-S23. [PMID: 39645293 DOI: 10.1016/j.arteri.2024.07.004] [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: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 12/09/2024]
Abstract
Despite the various therapeutic tools available, many patients do not achieve therapeutic goals, and cardiovascular diseases remain a significant cause of death in our setting. Furthermore, even in patients who manage to reduce their LDL-C levels to the recommended targets, cardiovascular events continue to occur. The therapeutic challenge and the persistent risk have led to active research into new drugs targeting novel therapeutic pathways in the field of lipoprotein metabolism disorders. The therapeutic approach involves new pharmacological mechanisms, ranging from small molecules and monoclonal antibodies to RNA interference, with inclisiran being the first drug approved for clinical use in the cardiovascular domain. In this review, we aim to provide a comprehensive overview of the new therapeutic targets and pharmacological mechanisms under development, as well as their potential clinical impact.
Collapse
Affiliation(s)
- Lluís Masana
- Unitat de Recerca en Lípids i Arteriosclerosi, Unitat de Medicina Vascular i Metabolisme, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Tarragona, España.
| | - Daiana Ibarretxe
- Unitat de Recerca en Lípids i Arteriosclerosi, Unitat de Medicina Vascular i Metabolisme, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Tarragona, España
| |
Collapse
|
30
|
Khan MI, Zahir RS, Dominguez AC, Romeo FJ. Role of Lipoprotein (A) in aortic valve stenosis: Novel disease mechanisms and emerging pharmacotherapeutic approaches. IJC HEART & VASCULATURE 2024; 55:101543. [PMID: 39555492 PMCID: PMC11564994 DOI: 10.1016/j.ijcha.2024.101543] [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: 03/24/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024]
Abstract
Lipoprotein(a) (Lp(a)) has garnered increasing attention as a significant contributor to the pathogenesis of aortic stenosis (AS), prompting a focused investigation into innovative pharmacological strategies to target this lipoprotein and its associated risks. Despite its recognized role in AS progression, Lp(a) often remains overlooked in clinical assessments, mirroring the broader challenges observed in holistic disease management. This review delves into the mechanistic intricacies of Lp(a) involvement in AS pathophysiology and its potential as a therapeutic target. Drawing parallels with the imperative for healthcare providers to proactively engage with patients regarding treatment regimens, this review underscores the essential role of cardiologists and physicians in recognizing and addressing Lp(a) as a modifiable risk factor in AS management. Furthermore, it explores promising avenues of novel drug approaches, including emerging pharmacotherapies and targeted interventions, aimed at modulating Lp(a) levels and attenuating AS progression. By navigating the complexities of Lp(a) modulation and its implications for AS management, this review aims to bridge critical gaps in understanding and clinical practice, ultimately optimizing treatment strategies and improving patient outcomes in the realm of AS therapeutics.
Collapse
Affiliation(s)
- Mohammad Ishrak Khan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raisa Subaita Zahir
- College of Allopathic Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Abel Casso Dominguez
- Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francisco José Romeo
- Department of Cardiology, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, FL, USA
| |
Collapse
|
31
|
Hanna DB, Rodriguez CJ. Lipoprotein(a) and Cardiovascular Disease in People Living With HIV: The Next Horizon. J Am Heart Assoc 2024:e039074. [PMID: 39609261 DOI: 10.1161/jaha.124.039074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Affiliation(s)
- David B Hanna
- Division of Epidemiology, Departments of Epidemiology and Population Health Albert Einstein College of Medicine and Montefiore Medical Center Bronx NY
| | - Carlos J Rodriguez
- Division of Epidemiology, Departments of Epidemiology and Population Health Albert Einstein College of Medicine and Montefiore Medical Center Bronx NY
- Division of Cardiology, Department of Medicine Albert Einstein College of Medicine and Montefiore Medical Center Bronx NY
| |
Collapse
|
32
|
Shah NP, Mulder H, Lydon E, Chiswell K, Hu X, Lampron Z, Cohen L, Patel MR, Taubes S, Song W, Mulukutla SR, Saeed A, Morin DP, Bradley SM, Hernandez AF, Pagidipati NJ. Lipoprotein (a) Testing in Patients With Atherosclerotic Cardiovascular Disease in 5 Large US Health Systems. J Am Heart Assoc 2024; 13:e035610. [PMID: 39494552 PMCID: PMC11935684 DOI: 10.1161/jaha.124.035610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/12/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Lipoprotein (a) is an independent risk factor for atherosclerotic cardiovascular disease. However, lipoprotein (a) testing remains variable and it is unclear what factors influence testing and if testing changes clinical management. METHODS AND RESULTS A retrospective study using electronic medical record data from 5 health systems identified an atherosclerotic cardiovascular disease cohort divided into those with and without a lipoprotein (a) test between 2019 and 2021. Baseline characteristics and lipid-lowering therapy patterns were assessed. Multivariable regression modeling was used to determine factors associated with lipoprotein (a) testing. Among 595 684 patients with atherosclerotic cardiovascular disease, only 2587 (0.4%) were tested for lipoprotein (a). Those who were older or Black individuals were less likely to have lipoprotein (a) testing, while those with familial hypercholesterolemia, ischemic stroke/transient ischemic attack, peripheral artery disease, prior lipid-lowering therapy, or low-density lipoprotein cholesterol ≥130 mg/dL were more likely to be tested. Those with a lipoprotein (a) test, regardless of the lipoprotein (a) value, were more frequently initiated on any statin therapy (30.3% versus 10.6%, P < 0.001), ezetimibe (7.65% versus 0.8%, P < 0.001), or proprotein convertase substilisin/kexin type 9 inhibitor (6.7% versus 0.3%, P < 0.001) compared with those without a test. Those with an elevated lipoprotein (a) level more frequently initiated ezetimibe (11.5% versus 5.9%, P < 0.001) or proprotein convertase substilisin/kexin type 9 inhibitor (10.9% versus 4.8%, P < 0.001). CONCLUSIONS Lipoprotein (a) testing in patients with atherosclerotic cardiovascular disease is infrequent, with evidence of disparities among older or Black individuals. Testing for lipoprotein (a), regardless of level, is associated with greater initiation of any lipid-lowering therapy, while elevated lipoprotein (a) is associated with greater initiation of nonstatin lipid-lowering therapy. There is a critical need for multidisciplinary and inclusive approaches to raise awareness for lipoprotein (a) testing, and its implications on management.
Collapse
Affiliation(s)
- Nishant P. Shah
- Division of CardiologyDuke University HospitalDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| | | | | | | | - Xingdi Hu
- Novartis Pharmaceuticals CorporationEast HanoverNJUSA
| | | | | | - Manesh R. Patel
- Division of CardiologyDuke University HospitalDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| | - Susan Taubes
- Novartis Pharmaceuticals CorporationEast HanoverNJUSA
| | - Wenliang Song
- Division of CardiologyVanderbilt UniversityNashvilleTNUSA
| | | | - Anum Saeed
- Division of CardiologyUniversity of PittsburghPittsburghPAUSA
| | - Daniel P. Morin
- Division of CardiologyOchsner Medical CenterNew OrleansLAUSA
| | | | - Adrian F. Hernandez
- Division of CardiologyDuke University HospitalDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| | - Neha J. Pagidipati
- Division of CardiologyDuke University HospitalDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| |
Collapse
|
33
|
Ma Z, Zhong J, Tu W, Li S, Chen J. The functions of apolipoproteins and lipoproteins in health and disease. MOLECULAR BIOMEDICINE 2024; 5:53. [PMID: 39465476 PMCID: PMC11513782 DOI: 10.1186/s43556-024-00218-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
Lipoproteins and apolipoproteins are crucial in lipid metabolism, functioning as essential mediators in the transport of cholesterol and triglycerides and being closely related to the pathogenesis of multiple systems, including cardiovascular. Lipoproteins a (Lp(a)), as a unique subclass of lipoproteins, is a low-density lipoprotein(LDL)-like particle with pro-atherosclerotic and pro-inflammatory properties, displaying high heritability. More and more strong evidence points to a possible link between high amounts of Lp(a) and cardiac conditions like atherosclerotic cardiovascular disease (ASCVD) and aortic stenosis (AS), making it a risk factor for heart diseases. In recent years, Lp(a)'s role in other diseases, including neurological disorders and cancer, has been increasingly recognized. Although therapies aimed at low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) have achieved significant success, elevated Lp(a) levels remain a significant clinical management problem. Despite the limited efficacy of current lipid-lowering therapies, major clinical advances in new Lp(a)-lowering therapies have significantly advanced the field. This review, grounded in the pathophysiology of lipoproteins, seeks to summarize the wide-ranging connections between lipoproteins (such as LDL-C and HDL-C) and various diseases, alongside the latest clinical developments, special emphasis is placed on the pivotal role of Lp(a) in cardiovascular disease, while also examining its future potential and mechanisms in other conditions. Furthermore, this review discusses Lp(a)-lowering therapies and highlights significant recent advances in emerging treatments, advocates for further exploration into Lp(a)'s pathogenic mechanisms and its potential as a therapeutic target, proposing new secondary prevention strategies for high-risk individuals.
Collapse
Affiliation(s)
- Zijun Ma
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, 430030, Hubei, China
| | - Wei Tu
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Shiliang Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun Chen
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China.
| |
Collapse
|
34
|
Sharma P, Judy R, Yuan S, Gellatly C, Saxby KL, Bown MJ, Levin MG, Damrauer SM. Lipoprotein (a) is associated with increased risk of Abdominal Aortic Aneurysm. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.13.24313646. [PMID: 39314961 PMCID: PMC11419246 DOI: 10.1101/2024.09.13.24313646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Introduction Lipoprotein(a) (Lp(a)) is a circulating apolipoprotein B (ApoB) containing particle that has been observationally linked to atherosclerotic cardiovascular disease and is the target of emerging therapeutics. Recent work has highlighted the role of circulating lipoproteins in abdominal aortic aneurysm (AAA). We sought to triangulate human observational and genetic evidence to evaluate the role of Lp(a) in AAA. Methods We tested the association between circulating levels of Lp(a) and clinically diagnosed abdominal aortic aneurysms while controlling for traditional AAA risk factors and levels of ApoB using logistic regression among 795 individuals with and 374,772 individuals without AAA in the UK Biobank (UKB). Multivariable Mendelian randomization (MVMR) was used to test for putatively causal associations between Lp(a) and AAA controlling for ApoB. Genetic instruments for Lp(a) and ApoB were created from genome-wide association studies (GWAS) of Lp(a) and ApoB comprising 335,796 and 418,505 UKB participants, respectively. The instruments were tested for association with AAA using data from a GWAS of 39,221 individuals with and 1,086,107 without AAA. Results Elevated Lp(a) levels were observationally associated with an increased risk of AAA (OR 1.04 per 10 nmol/L Lp(a); 95%CI 1.02-1.05; P<0.01). Clinically elevated Lp(a) levels (>150nmol/L) were likewise associated with an increased risk of AAA (OR 1.47; 95% CI 1.15-1.88; P < 0.01) when compared to individuals with Lp(a) levels <150nmol/L. MVMR confirmed a significant, ApoB-independent association between increased Lp(a) and increased risk of AAA (OR 1.13 per SD increase in Lp(a); 95%CI 1.02-1.24; P<0.02). Conclusion Both observational and genetic analyses support an association between increased Lp(a) and AAA risk that is independent of ApoB. These findings suggest that Lp(a) may be a therapeutic target for AAA and drive the inclusion of AAA as an outcome in clinical trials of Lp(a) antagonists.
Collapse
|
35
|
Wu T, Hu Y, Tang LV. Gene therapy for polygenic or complex diseases. Biomark Res 2024; 12:99. [PMID: 39232780 PMCID: PMC11375922 DOI: 10.1186/s40364-024-00618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
Gene therapy utilizes nucleic acid drugs to treat diseases, encompassing gene supplementation, gene replacement, gene silencing, and gene editing. It represents a distinct therapeutic approach from traditional medications and introduces novel strategies for genetic disorders. Over the past two decades, significant advancements have been made in the field of gene therapy, leading to the approval of various gene therapy drugs. Gene therapy was initially employed for treating genetic diseases and cancers, particularly monogenic conditions classified as orphan diseases due to their low prevalence rates; however, polygenic or complex diseases exhibit higher incidence rates within populations. Extensive research on the etiology of polygenic diseases has unveiled new therapeutic targets that offer fresh opportunities for their treatment. Building upon the progress achieved in gene therapy for monogenic diseases and cancers, extending its application to polygenic or complex diseases would enable targeting a broader range of patient populations. This review aims to discuss the strategies of gene therapy, methods of gene editing (mainly CRISPR-CAS9), and carriers utilized in gene therapy, and highlight the applications of gene therapy in polygenic or complex diseases focused on applications that have either entered clinical stages or are currently undergoing clinical trials.
Collapse
Affiliation(s)
- Tingting Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| | - Liang V Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| |
Collapse
|
36
|
Makhmudova U, Steinhagen-Thiessen E, Volpe M, Landmesser U. Advances in nucleic acid-targeted therapies for cardiovascular disease prevention. Cardiovasc Res 2024; 120:1107-1125. [PMID: 38970537 DOI: 10.1093/cvr/cvae136] [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: 01/30/2024] [Revised: 04/14/2024] [Accepted: 05/03/2024] [Indexed: 07/08/2024] Open
Abstract
Nucleic acid-based therapies are being rapidly developed for prevention and management of cardiovascular diseases (CVD). Remarkable advancements have been achieved in the delivery, safety, and effectiveness of these therapeutics in the past decade. These therapies can also modulate therapeutic targets that cannot be sufficiently addressed using traditional drugs or antibodies. Among the nucleic acid-targeted therapeutics under development for CVD prevention are RNA-targeted approaches, including antisense oligonucleotides (ASO), small interfering RNAs (siRNA), and novel genome editing techniques. Genetic studies have identified potential therapeutic targets that are suggested to play a causative role in development and progression of CVD. RNA- and DNA-targeted therapeutics can be particularly well delivered to the liver, where atherogenic lipoproteins and angiotensinogen (AGT) are produced. Current targets in lipid metabolism include proprotein convertase subtilisin/kexin type 9 (PCSK9), apolipoprotein A (ApoA), apolipoprotein C3 (ApoC3), angiopoietin-like 3 (ANGPTL3). Several large-scale clinical development programs for nucleic acid-targeted therapies in cardiovascular prevention are under way, which may also be attractive from a therapy adherence point of view, given the long action of these therapeutics. In addition to genome editing, the concept of gene transfer is presently under assessment in preclinical and clinical investigations as a potential approach for addressing low-density lipoprotein receptor deficiency. Furthermore, ongoing research is exploring the use of RNA-targeted therapies to treat arterial hypertension by reducing hepatic angiotensinogen (AGT) production. This review summarizes the rapid translation of siRNA and ASO therapeutics as well as gene editing into clinical studies to treat dyslipidemia and arterial hypertension for CVD prevention. It also outlines potential innovative therapeutic options that are likely relevant to the future of cardiovascular medicine.
Collapse
Affiliation(s)
- Umidakhon Makhmudova
- Department of Cardiology, Angiology, and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik/Centrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Elisabeth Steinhagen-Thiessen
- Friede Springer Cardiovascular Prevention Center at Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik/Centrum, Charitéplatz 1, 10117 Berlin, Germany
- Department of Endocrinology and Metabolic Diseases, Charite Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035-1039, Rome 00189, Italy
- Cardiology Department, IRCCS San Raffaele Roma, Via di Valcannuta 250, Rome 00166, Italy
| | - Ulf Landmesser
- Department of Cardiology, Angiology, and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik/Centrum, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Centre for Cardiovascular Research, DZHK, Partner Site Berlin, Germany
| |
Collapse
|
37
|
Mach F, Visseren FLJ, Cater NB, Salhi N, Soronen J, Ray KK, Delgado V, Jukema JW, Laufs U, Zamorano JL, Ros E, Plat J, Gesztes AG, Tokgozoglu L, Packard C, Libby P. Addressing residual risk beyond statin therapy: New targets in the management of dyslipidaemias-A report from the European Society of Cardiology Cardiovascular Round Table. J Clin Lipidol 2024; 18:e685-e700. [PMID: 39289123 DOI: 10.1016/j.jacl.2024.07.001] [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: 09/12/2023] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 09/19/2024]
Abstract
Cardiovascular (CV) disease is the most common cause of death in Europe. Despite proven benefits, use of lipid-lowering therapy remains suboptimal. Treatment goals are often not achieved, even in patients at high risk with atherosclerotic CV disease (ASCVD). The occurrence of CV events in patients on lipid-lowering drugs is defined as "residual risk", and can result from inadequate control of plasma lipids or blood pressure, inflammation, diabetes, and environmental hazards. Assessment of CV risk factors and vascular imaging can aid in the evaluation and management decisions for individual patients. Lifestyle measures remain the primary intervention for lowering CV risk. Where drug therapies are required to reach lipid treatment targets, their effectiveness increases when they are combined with lifestyle measures delivered through formal programs. However, lipid drug dosage and poor adherence to treatment remain major obstacles to event-free survival. This article discusses guideline-supported treatment algorithms beyond statin therapy that can help reduce residual risk in specific patient profiles while also likely resulting in substantial healthcare savings through better patient management and treatment adherence.
Collapse
Affiliation(s)
- François Mach
- Department of Cardiology, Geneva University Hospitals, Geneva, Switzerland (Dr Mach).
| | - Frank L J Visseren
- Department of Internal Medicine, University Medical Center, Utrecht, the Netherlands (Dr Visseren)
| | - Nilo B Cater
- Global Medical Affairs, Pfizer, New York, NY, USA (Dr Cater)
| | - Nejoua Salhi
- Global Medical Affairs, AMGEN Europe GmbH, Rotkreuz, Switzerland (Dr Salhi)
| | - Jarkko Soronen
- Scientific Communication and Strategy, Daiichi-Sankyo Europe GmbH, Munich, Germany (Dr Soronen)
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK (Dr Ray)
| | - Victoria Delgado
- University Hospital Germans Trias i Pujol, Badalona, Spain (Dr Delgado)
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands (Dr Jukema); Netherlands Heart Institute, Utrecht, the Netherlands (Dr Jukema)
| | - Ulrich Laufs
- Department of Cardiology, Leipzig University Hospital, Leipzig, Germany (Dr Laufs)
| | - Jose-Luis Zamorano
- Department of Cardiology, University Hospital Ramón y Cajal, Madrid, Spain (Dr Zamorano)
| | - Emilio Ros
- Lipid Clinic, Department of Endocrinology and Nutrition, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain (Dr Ros)
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, the Netherlands (Dr Plat)
| | - Akos Gabor Gesztes
- SZÍVSN - Heartily (We help you) National Patient Association, Budapest, Hungary (Dr Gesztes)
| | - Lale Tokgozoglu
- Department of Cardiology, Hacettepe University, Ankara, Turkey (Dr Tokgozoglu)
| | - Chris Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK (Dr Packard)
| | - Peter Libby
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Boston, MA, USA (Dr Libby)
| |
Collapse
|
38
|
Saad ALGhasab N, Fogacci F, Avagimyan A, Cicero AFG. Expanding therapeutic options: overview of novel pharmacotherapies for dyslipidemia. Expert Opin Pharmacother 2024; 25:1795-1805. [PMID: 39286934 DOI: 10.1080/14656566.2024.2406270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 08/18/2024] [Accepted: 09/16/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION Dyslipidemia plays a crucial role in the development of atherosclerotic cardiovascular diseases. AREAS COVERED This article explores the emerging therapeutic targets for the treatment of dyslipidemia and provides novel insights into this field. Thus, it aims to contribute to the understanding and advancement of therapeutic options for managing dyslipidemia. EXPERT OPINION Optimizing the use of available first- and second-line lipid-lowering drugs allows us to adequately control low-density lipoprotein cholesterol (LDL-C) levels, even in statin-intolerant individuals and in patients at high and very high risk of developing cardiovascular diseases who must reach more aggressive LDL-C targets. The drugs under development will further improve our ability to manage the overall lipid-related cardiovascular disease risk and target other dyslipidemia biomarkers.
Collapse
Affiliation(s)
- Naif Saad ALGhasab
- Department of Internal Medicine, Medical College, Ha'il University, Ha'il, Saudi Arabia
- Department of Cardiology, Libin cardiovascular institute, Calgary University, Calgary, Canada
| | - Federica Fogacci
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Ashot Avagimyan
- Anatomical Pathology and Clinical Morphology Department, Yerevan State Medical University, Yerevan, Armenia
| | - Arrigo F G Cicero
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Cardiovascular Medicine Unit, IRCCS AOUBO, Bologna, Italy
| |
Collapse
|
39
|
O'Donoghue ML, Rosenson RS, López JAG, Lepor NE, Baum SJ, Stout E, Gaudet D, Knusel B, Kuder JF, Murphy SA, Wang H, Wu Y, Shah T, Wang J, Wilmanski T, Sohn W, Kassahun H, Sabatine MS. The Off-Treatment Effects of Olpasiran on Lipoprotein(a) Lowering: OCEAN(a)-DOSE Extension Period Results. J Am Coll Cardiol 2024; 84:790-797. [PMID: 39168564 DOI: 10.1016/j.jacc.2024.05.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Olpasiran, a small interfering RNA (siRNA), blocks lipoprotein(a) (Lp(a)) production by preventing translation of apolipoprotein(a) mRNA. In phase 2, higher doses of olpasiran every 12 weeks (Q12W) reduced circulating Lp(a) by >95%. OBJECTIVES This study sought to assess the timing of return of Lp(a) to baseline after discontinuation of olpasiran, as well as longer-term safety. METHODS OCEAN(a)-DOSE (Olpasiran Trials of Cardiovascular Events And LipoproteiN[a] Reduction-DOSE Finding Study) was a phase 2, dose-finding trial that enrolled 281 participants with atherosclerotic cardiovascular disease and Lp(a) >150 nmol/L to 1 of 4 active doses of olpasiran vs placebo (10 mg, 75 mg, 225 mg Q12W, or an exploratory dose of 225 mg Q24W given subcutaneously). The last dose of olpasiran was administered at week 36; after week 48, there was an extended off-treatment follow-up period for a minimum of 24 weeks. RESULTS A total of 276 (98.2%) participants entered the off-treatment follow-up period. The median study exposure (treatment combined with off-treatment phases) was 86 weeks (Q1-Q3: 79-99 weeks). For the 75 mg Q12W dose, the off-treatment placebo-adjusted mean percent change from baseline in Lp(a) was -76.2%, -53.0%, -44.0%, and -27.9% at 60, 72, 84, and 96 weeks, respectively (all P < 0.001). The respective off-treatment changes in Lp(a) for the 225 mg Q12W dose were -84.4%, -61.6%, -52.2%, and -36.4% (all P < 0.001). During the extension follow-up phase, no new safety concerns were identified. CONCLUSIONS Olpasiran is a potent siRNA with prolonged effects on Lp(a) lowering. Participants receiving doses ≥75 mg Q12W sustained a ∼40% to 50% reduction in Lp(a) levels close to 1 year after the last dose. (Olpasiran Trials of Cardiovascular Events And LipoproteiN[a] Reduction-DOSE Finding Study [OCEAN(a)-DOSE]; NCT04270760).
Collapse
Affiliation(s)
- Michelle L O'Donoghue
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| | - Robert S Rosenson
- Icahn School of Medicine, Mount Sinai Hospital, New York, New York, USA
| | | | - Norman E Lepor
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Seth J Baum
- Flourish Research, Boca Raton, Florida, USA; Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Elmer Stout
- Crossroads Clinical Research Inc, Mooresville, North Carolina, USA
| | - Daniel Gaudet
- Department of Medicine, University of Montréal, Chicoutimi, Quebec, Canada
| | - Beat Knusel
- Global Development, Amgen, Thousand Oaks, California, USA
| | - Julia F Kuder
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sabina A Murphy
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Huei Wang
- Global Development, Amgen, Thousand Oaks, California, USA
| | - You Wu
- Global Development, Amgen, Thousand Oaks, California, USA
| | - Trupti Shah
- Global Development, Amgen, Thousand Oaks, California, USA
| | - Jingying Wang
- Global Development, Amgen, Thousand Oaks, California, USA
| | | | - Winnie Sohn
- Global Development, Amgen, Thousand Oaks, California, USA
| | | | - Marc S Sabatine
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Moore MK, Jones GT, McCormick S, Williams MJA, Coffey S. Association between lipoprotein(a), LPA genetic risk score, aortic valve disease, and subsequent major adverse cardiovascular events. Eur J Prev Cardiol 2024; 31:1303-1311. [PMID: 38593219 DOI: 10.1093/eurjpc/zwae100] [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: 11/19/2023] [Revised: 02/18/2024] [Accepted: 03/01/2024] [Indexed: 04/11/2024]
Abstract
AIMS Cohort studies have demonstrated associations between calcific aortic valve disease (CAVD) and Lp(a). As Lp(a) is almost entirely genetically determined, in this study, we aim to determine whether Lp(a), when predicted from genetic data, is associated with CAVD and major adverse cardiovascular events (MACEs). METHODS AND RESULTS Patients undergoing coronary angiography between January 2012 and May 2013 were invited to participate in the study. Of 752 analysable participants, 446 had their Lp(a) measured and 703 had a calculable LPA genetic risk score (GRS). The primary outcomes were the presence of CAVD at baseline and MACE over a 7-year follow-up. The GRS explained 45% of variation in Lp(a). After adjustment for cardiac risk factors and coronary artery disease (CAD), the odds of CAVD increased with increasing Lp(a) [odds ratio (OR) 1.039 per 10-unit increase, 95% confidence interval (CI) 1.022-1.057, P < 0.001] and GRS (OR 1.054 per 10-unit increase, 95% CI 1.024-1.086; P < 0.001). Lipoprotein(a) and the GRS as continuous variables were not associated with subsequent MACEs. A dichotomized GRS (>54) was associated with MACE, but this relationship became non-significant when CAD classification was added into the model (OR 1.333, 95% CI 0.927-1.912; P = 0.12). CONCLUSION An LPA GRS can explain 45% of variation in Lp(a) levels, and both Lp(a) and the GRS are associated with CAVD. An elevated GRS is associated with future cardiac events in a secondary risk setting, but, if the CAD status is known, it does not provide additional prognostic information.
Collapse
Affiliation(s)
- Matthew K Moore
- Department of Medicine, HeartOtago, Dunedin School of Medicine, University of Otago, 201 Great King Street, Dunedin 9016, New Zealand
| | - Gregory T Jones
- Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, 201 Great King Street, Dunedin 9016, New Zealand
| | - Sally McCormick
- Department of Biochemistry, University of Otago, 710 Cumberland Street, Dunedin 9016, New Zealand
| | - Michael J A Williams
- Department of Medicine, HeartOtago, Dunedin School of Medicine, University of Otago, 201 Great King Street, Dunedin 9016, New Zealand
- Department of Cardiology, Dunedin Hospital, Te Whatu Ora/Health New Zealand, 201 Great King Street, Dunedin 9016, New Zealand
| | - Sean Coffey
- Department of Medicine, HeartOtago, Dunedin School of Medicine, University of Otago, 201 Great King Street, Dunedin 9016, New Zealand
- Department of Cardiology, Dunedin Hospital, Te Whatu Ora/Health New Zealand, 201 Great King Street, Dunedin 9016, New Zealand
| |
Collapse
|
41
|
Hou G, Alissa M, Alsuwat MA, Ali Alarjany HM, Alzahrani KJ, Althobaiti FM, Mujalli HM, Alotaiby MM, Al-Doaiss AA, Anthony S. The art of healing hearts: Mastering advanced RNA therapeutic techniques to shape the evolution of cardiovascular medicine in biomedical science. Curr Probl Cardiol 2024; 49:102627. [PMID: 38723793 DOI: 10.1016/j.cpcardiol.2024.102627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide and are associated with increasing financial health burden that requires research into novel therapeutic approaches. Since the early 2000s, the availability of next-generation sequencing techniques such as microRNAs, circular RNAs, and long non-coding RNAs have been proven as potential therapeutic targets for treating various CVDs. Therapeutics based on RNAs have become a viable option for addressing the intricate molecular pathways that underlie the pathophysiology of CVDs. We provide an in-depth analysis of the state of RNA therapies in the context of CVDs, emphasizing various approaches that target the various stages of the basic dogma of molecular biology to effect temporary or long-term changes. In this review, we summarize recent methodologies used to screen for novel coding and non-coding RNA candidates with diagnostic and treatment possibilities in cardiovascular diseases. These methods include single-cell sequencing techniques, functional RNA screening, and next-generation sequencing.Lastly, we highlighted the potential of using oligonucleotide-based chemical products such as modified RNA and RNA mimics/inhibitors for the treatment of CVDs. Moreover, there will be an increasing number of potential RNA diagnostic and therapeutic for CVDs that will progress to expand for years to come.
Collapse
Affiliation(s)
- Guoliang Hou
- Department of Cardiology, Tengzhou Central People's Hospital, Shandong 277599, China
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Meshari A Alsuwat
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21974, Saudi Arabia
| | | | - Khalid J Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21974, Saudi Arabia
| | - Fahad M Althobaiti
- Department of Nursing Leadership and Education, Nursing College, Taif University, Taif 21974, Saudi Arabia
| | | | - Monearah M Alotaiby
- Department of Laboratory, King Faisal Medical Complex, Ministry of Health, Taif 26514, Saudi Arabia
| | - Amin A Al-Doaiss
- Biology Department, College of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Stefan Anthony
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University Liaoning Provence China, China.
| |
Collapse
|
42
|
Hang S, Hegele RA. Elderly patients with very high plasma lipoprotein(a) concentrations and few cardiovascular consequences: a case series. J Int Med Res 2024; 52:3000605241271876. [PMID: 39197866 PMCID: PMC11375631 DOI: 10.1177/03000605241271876] [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] [Indexed: 09/01/2024] Open
Abstract
Lipoprotein(a) (Lp(a)) is an atherogenic low-density lipoprotein (LDL)-like particle that is currently regarded as a non-modifiable risk factor for atherosclerotic cardiovascular disease. The number of patients detected with elevated Lp(a) concentrations has been increasing in recent years, although the implication of this finding is unclear for patients and physicians. We screened our lipid clinic database for patients aged >65 years with very high Lp(a) concentrations, which were defined as >230 nmol/L, and cardiovascular outcomes were assessed. The patients' (n = 16) mean (±standard deviation) age was 72.2 ± 7.1 years and the mean Lp(a) concentration was 313 ± 68 nmol/L. After a cumulative 129.0 patient-year follow-up (mean: 8.1 ± 4.2 years), the mean age was 80.3 ± 7.0 years. We observed a low baseline prevalence of cardiovascular events, with only two patients having a history of cardiovascular events. Furthermore, zero incident adverse cardiovascular events were recorded over the follow-up. Therefore, very high Lp(a) concentrations and disease-free old age are not mutually exclusive. Our aggregated clinical experience is that there is only a modest association between elevated Lp(a) concentrations and adverse outcomes. Nonetheless, we still advise treating modifiable risk factors in these patients.
Collapse
Affiliation(s)
| | - Robert A Hegele
- Departments of Medicine and
- Biochemistry, and
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
43
|
Tsimikas S, Yeang C, Kronenberg F. In Search of an Accurate Measurement of LDL-C: Correction for Lp(a)-Cholesterol to Predict Clinical Outcomes. J Am Coll Cardiol 2024; 84:178-181. [PMID: 38960511 DOI: 10.1016/j.jacc.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 07/05/2024]
Affiliation(s)
| | - Calvin Yeang
- University of California-San Diego, La Jolla, California, USA. https://twitter.com/CalvinYeang
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria. https://twitter.com/KronenbergLab
| |
Collapse
|
44
|
Wang P, Yuan D, Zhao X, Zhu P, Guo X, Jiang L, Xu N, Wang Z, Liu R, Wang Q, Chen Y, Zhang Y, Xu J, Liu Z, Song Y, Zhang Z, Yao Y, Feng Y, Tang X, Wang X, Gao R, Han Y, Yuan J. Inverse Association of Lipoprotein(a) on Long-Term Bleeding Risk in Patients with Coronary Heart Disease: Insight from a Multicenter Cohort in Asia. Thromb Haemost 2024; 124:684-694. [PMID: 37487540 PMCID: PMC11199048 DOI: 10.1055/s-0043-1771188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/08/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Lipoprotein(a), or Lp(a), has been recognized as a strong risk factor for atherosclerotic cardiovascular disease. However, the relationship between Lp(a) and bleeding remains indistinct, especially in the secondary prevention population of coronary artery disease (CAD). This investigation aimed to evaluate the association of Lp(a) with long-term bleeding among patients with CAD. METHODS Based on a prospective multicenter cohort of patients with CAD consecutively enrolled from January 2015 to May 2019 in China, the current analysis included 16,150 participants. Thus, according to Lp(a) quintiles, all subjects were divided into five groups. The primary endpoint was bleeding at 2-year follow-up, and the secondary endpoint was major bleeding at 2-year follow-up. RESULTS A total of 2,747 (17.0%) bleeding and 525 (3.3%) major bleeding were recorded during a median follow-up of 2.0 years. Kaplan-Meier survival analysis showed the highest bleeding incidence in Lp(a) quintile 1, compared with patients in Lp(a) quintiles 2 to 5 (p < 0.001), while the incidence of major bleeding seemed similar between the two groups. Moreover, restricted cubic spline analysis suggested that there was an L-shaped association between Lp(a) and 2-year bleeding after adjustment for potential confounding factors, whereas there was no significant association between Lp(a) and 2-year major bleeding. CONCLUSION There was an inverse and L-shaped association of Lp(a) with bleeding at 2-year follow-up in patients with CAD. More attention and effort should be made to increase the clinician awareness of Lp(a)'s role, as a novel marker for bleeding risk to better guide shared-decision making in clinical practice.
Collapse
Affiliation(s)
- Peizhi Wang
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Deshan Yuan
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xueyan Zhao
- Special Demand Medical Care Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Zhu
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Lin Jiang
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Xu
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhifang Wang
- Department of Cardiology, Xinxiang Central Hospital, Xinxiang, Henan Province, China
| | - Ru Liu
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qingsheng Wang
- Department of Cardiology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei Province, China
| | - Yan Chen
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongzhen Zhang
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Jingjing Xu
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenyu Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Song
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Zhang
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yi Yao
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingqing Feng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou, Guangdong Province, China
| | - Xiaofang Tang
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaozeng Wang
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning Province, China
| | - Runlin Gao
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaling Han
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning Province, China
| | - Jinqing Yuan
- Department of Cardiology, Center for Coronary Heart Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
45
|
Bellomo TR, Liu Y, Gilliland TC, Miksenas H, Haidermota S, Wong M, Hu X, Cristino JR, Browne A, Plutzky J, Tsimikas S, Januzzi JL, Natarajan P. Associations between lipoprotein(a), oxidized phospholipids, and extracoronary vascular disease. J Lipid Res 2024; 65:100585. [PMID: 38942114 PMCID: PMC11298641 DOI: 10.1016/j.jlr.2024.100585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024] Open
Abstract
The roles of lipoprotein(a) [Lp(a)] and related oxidized phospholipids (OxPLs) in the development and progression of coronary disease is known, but their influence on extracoronary vascular disease is not well-established. We sought to evaluate associations between Lp(a), OxPL apolipoprotein B (OxPL-apoB), and apolipoprotein(a) (OxPL-apo(a)) with angiographic extracoronary vascular disease and incident major adverse limb events (MALEs). Four hundred forty-six participants who underwent coronary and/or peripheral angiography were followed up for a median of 3.7 years. Lp(a) and OxPLs were measured before angiography. Elevated Lp(a) was defined as ≥150 nmol/L. Elevated OxPL-apoB and OxPL-apo(a) were defined as greater than or equal to the 75th percentile (OxPL-apoB ≥8.2 nmol/L and OxPL-apo(a) ≥35.8 nmol/L, respectively). Elevated Lp(a) had a stronger association with the presence of extracoronary vascular disease compared to OxPLs and was minimally improved with the addition of OxPLs in multivariable models. Compared to participants with normal Lp(a) and OxPL concentrations, participants with elevated Lp(a) levels were twice as likely to experience a MALE (odds ratio: 2.14, 95% confidence interval: 1.03, 4.44), and the strength of the association as well as the C statistic of 0.82 was largely unchanged with the addition of OxPL-apoB and OxPL-apo(a). Elevated Lp(a) and OxPLs are risk factors for progression and complications of extracoronary vascular disease. However, the addition of OxPLs to Lp(a) does not provide additional information about risk of extracoronary vascular disease. Therefore, Lp(a) alone captures the risk profile of Lp(a), OxPL-apoB, and OxPL-apo(a) in the development and progression of atherosclerotic plaque in peripheral arteries.
Collapse
Affiliation(s)
- Tiffany R Bellomo
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Yuxi Liu
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas C Gilliland
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Hannah Miksenas
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara Haidermota
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Megan Wong
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xingdi Hu
- Novartis Pharmaceuticals Corporation, Novartis, East Hanover, NJ, USA
| | | | - Auris Browne
- Novartis Pharmaceuticals Corporation, Novartis, East Hanover, NJ, USA
| | - Jorge Plutzky
- Division of Cardiology, Brigham and Women's Hospital, Boston, Harvard Medical School, Boston, MA, USA
| | - Sotirios Tsimikas
- Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA
| | - James L Januzzi
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cardiology Division, Baim Institute for Clinical Research, Boston, MA, USA
| | - Pradeep Natarajan
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
46
|
Januzzi JL, van Kimmenade RRJ, Liu Y, Hu X, Browne A, Plutzky J, Tsimikas S, Blankstein R, Natarajan P. Lipoprotein(a), Oxidized Phospholipids, and Progression to Symptomatic Heart Failure: The CASABLANCA Study. J Am Heart Assoc 2024; 13:e034774. [PMID: 38860394 PMCID: PMC11255745 DOI: 10.1161/jaha.124.034774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/09/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Higher lipoprotein(a) and oxidized phospholipid concentrations are associated with increased risk for coronary artery disease and valvular heart disease. The role of lipoprotein(a) or oxidized phospholipid as a risk factor for incident heart failure (HF) or its complications remains uncertain. METHODS AND RESULTS A total of 1251 individuals referred for coronary angiography in the Catheter Sampled Blood Archive in Cardiovascular Diseases (CASABLANCA) study were stratified on the basis of universal definition of HF stage; those in stage A/B (N=714) were followed up for an average 3.7 years for incident stage C/D HF or the composite of HF/cardiovascular death. During follow-up, 105 (14.7%) study participants in stage A/B progressed to symptomatic HF and 57 (8.0%) had cardiovascular death. In models adjusted for multiple HF risk factors, including severe coronary artery disease and aortic stenosis, individuals with lipoprotein(a) ≥150 nmol/L were at higher risk for progression to symptomatic HF (hazard ratio [HR], 1.90 [95% CI, 1.15-3.13]; P=0.01) or the composite of HF/cardiovascular death (HR, 1.71 [95% CI, 1.10-2.67]; P=0.02). These results remained significant after further adjustment of the model to include prior myocardial infarction (HF: HR, 1.89, P=0.01; HF/cardiovascular death: HR, 1.68, P=0.02). Elevated oxidized phospholipid concentrations were similarly associated with risk, particularly when added to higher lipoprotein(a). In Kaplan-Meier analyses, individuals with stage A/B HF and elevated lipoprotein(a) had shorter time to progression to stage C/D HF or HF/cardiovascular death (both log-rank P<0.001). CONCLUSIONS Among individuals with stage A or B HF, higher lipoprotein(a) and oxidized phospholipid concentrations are independent risk factors for progression to symptomatic HF or cardiovascular death. REGISTRATION URL: https://wwwclinicaltrials.gov; Unique identifier: NCT00842868.
Collapse
Affiliation(s)
- James L. Januzzi
- Division of CardiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMA
- Baim Institute for Clinical ResearchBostonMA
| | | | - Yuxi Liu
- Division of CardiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Xingdi Hu
- Novartis Pharmaceuticals CorporationEast HanoverNJ
| | - Auris Browne
- Novartis Pharmaceuticals CorporationEast HanoverNJ
| | - Jorge Plutzky
- Cardiovascular DivisionBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Sotirios Tsimikas
- Sulpizio Cardiovascular CenterUniversity of California San DiegoLa JollaCA
| | - Ron Blankstein
- Cardiovascular DivisionBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Pradeep Natarajan
- Division of CardiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMA
- Program in Medical and Population Genetics and the Cardiovascular Disease InitiativeBroad Institute of Harvard and MITCambridgeMA
| |
Collapse
|
47
|
Yao S, Kasargod A, Chiu R, Torgerson TR, Kupiec-Weglinski JW, Dery KJ. The Coming Age of Antisense Oligos for the Treatment of Hepatic Ischemia/Reperfusion (IRI) and Other Liver Disorders: Role of Oxidative Stress and Potential Antioxidant Effect. Antioxidants (Basel) 2024; 13:678. [PMID: 38929116 PMCID: PMC11200799 DOI: 10.3390/antiox13060678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Imbalances in the redox state of the liver arise during metabolic processes, inflammatory injuries, and proliferative liver disorders. Acute exposure to intracellular reactive oxygen species (ROS) results from high levels of oxidative stress (OxS) that occur in response to hepatic ischemia/reperfusion injury (IRI) and metabolic diseases of the liver. Antisense oligonucleotides (ASOs) are an emerging class of gene expression modulators that target RNA molecules by Watson-Crick binding specificity, leading to RNA degradation, splicing modulation, and/or translation interference. Here, we review ASO inhibitor/activator strategies to modulate transcription and translation that control the expression of enzymes, transcription factors, and intracellular sensors of DNA damage. Several small-interfering RNA (siRNA) drugs with N-acetyl galactosamine moieties for the liver have recently been approved. Preclinical studies using short-activating RNAs (saRNAs), phosphorodiamidate morpholino oligomers (PMOs), and locked nucleic acids (LNAs) are at the forefront of proof-in-concept therapeutics. Future research targeting intracellular OxS-related pathways in the liver may help realize the promise of precision medicine, revolutionizing the customary approach to caring for and treating individuals afflicted with liver-specific conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Kenneth J. Dery
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
48
|
Leistner DM, Laguna-Fernandez A, Haghikia A, Abdelwahed YS, Schatz AS, Erbay A, Roehle R, Fonseca AF, Ferber P, Landmesser U. Impact of elevated lipoprotein(a) on coronary artery disease phenotype and severity. Eur J Prev Cardiol 2024; 31:856-865. [PMID: 38348689 DOI: 10.1093/eurjpc/zwae007] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 03/19/2024]
Abstract
AIMS A thorough characterization of the relationship between elevated lipoprotein(a) [Lp(a)] and coronary artery disease (CAD) is lacking. This study aimed to quantitatively assess the association of increasing Lp(a) levels and CAD severity in a real-world population. METHODS AND RESULTS This non-interventional, cross-sectional, LipidCardio study included patients aged ≥21 years undergoing angiography (October 2016-March 2018) at a tertiary cardiology centre, who have at least one Lp(a) measurement. The association between Lp(a) and CAD severity was determined by synergy between PCI with taxus and cardiac surgery (SYNTAX)-I and Gensini scores and angiographic characteristics. Overall, 975 patients (mean age: 69.5 years) were included; 70.1% were male, 97.5% had Caucasian ancestry, and 33.2% had a family history of premature atherosclerotic cardiovascular disease. Median baseline Lp(a) level was 19.3 nmol/L. Patients were stratified by baseline Lp(a): 72.9% had < 65 nmol/L, 21.0% had ≥100 nmol/L, 17.2% had ≥125 nmol/L, and 12.9% had ≥150 nmol/L. Compared with the normal (Lp(a) < 65 nmol/L) group, elevated Lp(a) groups (e.g. ≥ 150 nmol/L) had a higher proportion of patients with prior CAD (48.4% vs. 62.7%; P < 0.01), prior coronary revascularization (39.1% vs. 51.6%; P = 0.01), prior coronary artery bypass graft (6.0% vs. 15.1%; P < 0.01), vessel(s) with lesions (68.5% vs. 81.3%; P = 0.03), diffusely narrowed vessels (10.9% vs. 16.5%; P = 0.01) or chronic total occlusion lesions (14.3% vs. 25.2%; P < 0.01), and higher median SYNTAX-I (3.0 vs. 5.5; P = 0.01) and Gensini (10.0 vs. 16.0; P < 0.01) scores. CONCLUSION Elevated Lp(a) was associated with a more severe presentation of CAD. Awareness of Lp(a) levels in patients with CAD may have implications in their clinical management.
Collapse
Affiliation(s)
- David M Leistner
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
- University Hospital Frankfurt and Wolfgang Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | | | - Arash Haghikia
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
| | - Youssef S Abdelwahed
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Anne-Sophie Schatz
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Aslihan Erbay
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- University Hospital Frankfurt and Wolfgang Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Robert Roehle
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ana F Fonseca
- Novartis Pharma AG, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Philippe Ferber
- Novartis Pharma AG, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Ulf Landmesser
- Department of Cardiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Herzkreislaufforschung (DZHK), partner site Berlin, DZHK-Geschäftsstelle, Potsdamer Str. 58, 10785 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center, Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
| |
Collapse
|
49
|
Sharma P, Klarin D, Voight BF, Tsao PS, Levin MG, Damrauer SM. Evaluation of Plasma Biomarkers for Causal Association With Peripheral Artery Disease. Arterioscler Thromb Vasc Biol 2024; 44:1114-1123. [PMID: 38545784 PMCID: PMC11043009 DOI: 10.1161/atvbaha.124.320674] [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: 01/04/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Hundreds of biomarkers for peripheral artery disease (PAD) have been reported in the literature; however, the observational nature of these studies limits causal inference due to the potential of reverse causality and residual confounding. We sought to evaluate the potential causal impact of putative PAD biomarkers identified in human observational studies through genetic causal inference methods. METHODS Putative circulating PAD biomarkers were identified from human observational studies through a comprehensive literature search based on terms related to PAD using PubMed, Cochrane, and Embase. Genetic instruments were generated from publicly available genome-wide association studies of circulating biomarkers. Two-sample Mendelian randomization was used to test the association of genetically determined biomarker levels with PAD using summary statistics from a genome-wide association study of 31 307 individuals with and 211 753 individuals without PAD in the Veterans Affairs Million Veteran Program and replicated in data from FinnGen comprised of 11 924 individuals with and 288 638 individuals without PAD. RESULTS We identified 204 unique circulating biomarkers for PAD from the observational literature, of which 173 were genetically instrumented using genome-wide association study results. After accounting for multiple testing (false discovery rate, <0.05), 10 of 173 (5.8%) biomarkers had significant associations with PAD. These 10 biomarkers represented categories including plasma lipoprotein regulation, lipid homeostasis, and protein-lipid complex remodeling. Observational literature highlighted different pathways including inflammatory response, negative regulation of multicellular organismal processes, and regulation of response to external stimuli. CONCLUSIONS Integrating human observational studies and genetic causal inference highlights several key pathways in PAD pathophysiology. This work demonstrates that a substantial portion of biomarkers identified in observational studies are not well supported by human genetic evidence and emphasizes the importance of triangulating evidence to understand PAD pathophysiology. Although the identified biomarkers offer insights into atherosclerotic development in the lower limb, their specificity to PAD compared with more widespread atherosclerosis requires further study.
Collapse
Affiliation(s)
- Pranav Sharma
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Derek Klarin
- Veterans Affairs Palo Alto Healthcare System, CA
- Division of Vascular Surgery, Stanford University School of Medicine, CA
| | - Benjamin F. Voight
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United State
| | - Philip S. Tsao
- Veterans Affairs Palo Alto Healthcare System, CA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, CA
| | - Michael G. Levin
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Scott M. Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
50
|
Abou-Karam R, Cheng F, Gady S, Fahed AC. The Role of Genetics in Advancing Cardiometabolic Drug Development. Curr Atheroscler Rep 2024; 26:153-162. [PMID: 38451435 DOI: 10.1007/s11883-024-01195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE OF REVIEW The objective of this review is to explore the role of genetics in cardiometabolic drug development. The declining costs of sequencing and the availability of large-scale genomic data have deepened our understanding of cardiometabolic diseases, revolutionizing drug discovery and development methodologies. We highlight four key areas in which genetics is empowering drug development for cardiometabolic disease: (1) identifying drug candidates, (2) anticipating drug target failures, (3) silencing and editing genes, and (4) enriching clinical trials. RECENT FINDINGS Identifying novel drug targets through genetic discovery studies and the use of genetic variants as indicators of potential drug efficacy and safety have become critical components of cardiometabolic drug discovery. We highlight the successes of genetically-informed therapeutic strategies, such as PCSK9 and ANGPTL3 inhibitors in lipid lowering and the emerging role of polygenic risk scores in improving the efficiency of clinical trials. Additionally, we explore the potential of gene silencing and editing technologies, such as antisense oligonucleotides and small interfering RNA, showcasing their promise in addressing diseases refractory to conventional treatments. In this review, we highlight four use cases that demonstrate the vital role of genetics in cardiometabolic drug development: (1) identifying drug candidates, (2) anticipating drug target failures, (3) silencing and editing genes, and (4) enriching clinical trials. Through these advances, genetics has paved the way to increased efficiency of drug development as well as the discovery of more personalized and effective treatments for cardiometabolic disease.
Collapse
Affiliation(s)
- Roukoz Abou-Karam
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street|CPZN 3.128, Boston, MA, 02114, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Fangzhou Cheng
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street|CPZN 3.128, Boston, MA, 02114, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shoshana Gady
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street|CPZN 3.128, Boston, MA, 02114, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Akl C Fahed
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street|CPZN 3.128, Boston, MA, 02114, USA.
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|