Published online Aug 26, 2025. doi: 10.4330/wjc.v17.i8.109657
Revised: June 14, 2025
Accepted: August 4, 2025
Published online: August 26, 2025
Processing time: 96 Days and 7.9 Hours
Elevated lipoprotein(a) [Lp(a)] is a major independent risk factor for atheroscle
Core Tip: Lepodisiran is a novel N-acetylgalactosamine-conjugated small interfering RNA that durably reduces lipoprotein(a) [Lp(a)] levels by silencing hepatic LPA message RNA expression. It offers up to 94% sustained Lp(a) reduction with minimal side effects and extended dosing intervals (6-12 months). This therapeutic breakthrough addresses a key residual cardiovascular risk factor previously untreatable with conventional lipid-lowering agents.
- Citation: Faisal A, Basit A, Iftikhar A, Saifullah M, Rehmaan MKU, Basil AM. Lepodisiran: From genetic targeting to cardiovascular promise: A detailed narrative review of the literature. World J Cardiol 2025; 17(8): 109657
- URL: https://www.wjgnet.com/1949-8462/full/v17/i8/109657.htm
- DOI: https://dx.doi.org/10.4330/wjc.v17.i8.109657
Cardiovascular disease (CVD) remains the foremost global health burden, accounting for an estimated 17.9 million deaths each year, representing approximately 32% of all global deaths, according to the World Health Organization[1]. In recent decades, research has shifted toward understanding residual cardiovascular risk beyond traditional lipid parameters. One such factor gaining considerable attention is lipoprotein(a) [Lp(a)], a genetically determined lipoprotein that confers both atherogenic and thrombogenic risk. Elevated Lp(a) has been strongly linked with premature atherosclerotic CVD (ASCVD), including coronary artery disease, ischemic stroke, and aortic valve stenosis[2,3].
Despite its clinical relevance, Lp(a) has long posed a therapeutic challenge. Traditional lipid-lowering therapies such as statins exert minimal impact, and in some cases paradoxically elevate Lp(a) levels[4]. Lepodisiran, a novel small inter
Lp(a) consists of a low-density lipoprotein (LDL)-like particle containing apolipoprotein B-100 attached via a disulfide bond to apolipoprotein(a) [apo(a)][5]. The apo(a) segment shares extensive homology with plasminogen, particularly in its kringle IV and V domains, contributing to its antifibrinolytic and pro-inflammatory properties[6]. The kringle IV type 2 (KIV-2) domain. A repeat region in apo(a) gene which exists in variable tandem repeats; plays a central role in deter
The LPA gene, located on chromosome 6q26-27, encodes for apo(a) and is the principal determinant of Lp(a) plasma levels[7,8]. Variations in KIV-2 repeat number explain up to 90% of inter-individual differences in Lp(a). Genome-wide association studies have validated LPA as a key locus influencing CVD risk[8,9].
Multiple large-scale epidemiological studies have confirmed a causal relationship between elevated Lp(a) levels and ASCVD. In a pivotal meta-analysis by Lu et al[2], individuals in the top third of Lp(a) distribution had a 2.5-fold higher risk of coronary heart disease compared to those in the lowest third[2]. Further, elevated Lp(a) levels have been inde
While statins remain foundational in lipid management, their effect on Lp(a) is negligible or even adverse. Several studies have shown that statin therapy can increase Lp(a) by up to 20%-25%[4]. PCSK9 inhibitors, such as evolocumab and alirocumab, have shown modest Lp(a) reductions of around 20%-30%, although their primary mechanism targets LDL cholesterol[12]. Niacin, once considered a potential therapy for Lp(a), showed reductions of 20%-30% but failed to demonstrate meaningful cardiovascular benefits in clinical outcomes and has since fallen out of favor due to poor tole
RNA interference offers a precision-based method to silence genes post-transcriptionally. siRNAs are short, double-stranded RNA molecules that, once inside hepatocytes, are incorporated into the RNA-induced silencing complex (RISC)[15]. The guide strand of the siRNA directs RISC to complementary sequences of message RNA (mRNA), leading to its cleavage and degradation. N-acetylgalactosamine (GalNAc)-conjugated siRNAs specifically bind to the asialoglycoprotein receptor, enabling targeted delivery to the liver where Lp(a) is synthesized[16].
Lepodisiran is a GalNAc-conjugated siRNA that targets LPA mRNA in hepatocytes. Upon subcutaneous injection, the GalNAc moiety facilitates receptor-mediated endocytosis via the asialoglycoprotein receptor[16]. Inside the cell, lepo
A key pharmacokinetic advantage of lepodisiran is its prolonged duration of action, attributed to the chemical stabilization of the siRNA strands and intracellular RISC residence. This allows for extended dosing intervals-potentially as infrequent as every 6 to 12 months-thus improving adherence[19]. Importantly, lepodisiran does not interfere with LDL receptor pathways, preserving compatibility with other lipid-lowering agents like statins and PCSK9 inhibitors[20].
Preclinical studies in rodents and non-human primates demonstrated robust reductions in Lp(a) concentrations up to 90% with sustained effects for several months[18]. No significant hepatotoxicity, nephrotoxicity, or immune responses were noted. These findings set the stage for first-in-human clinical evaluation.
In the phase 1 trial led by Sahebkar et al[11], healthy volunteers with elevated Lp(a) received a single subcutaneous dose ranging from 100 mg to 400 mg. Lepodisiran produced dose-dependent reductions in plasma Lp(a), with the highest dose achieving a 94% decrease that persisted for over 180 days. Importantly, no serious adverse events or laboratory abnormalities were reported.
The ALPACA trial [apo(a) Lp(a) siRNA in patients with CVD] was a multicenter, randomized, double-blind, placebo-controlled phase 2 study enrolling 320 patients with established ASCVD and Lp(a) levels above 125 nmol/L[21]. Some of the key findings are given below: (1) A single 400 mg dose of lepodisiran reduced Lp(a) by 93.9% from day 60 through 180; (2) Two doses, administered six months apart, maintained Lp(a) reduction of 94.8% up to one year; (3) Lp(a) levels remained 74.2% below baseline even 360 days after the second dose; and (4) Adverse events were comparable between treatment and placebo arms, with no dose-limiting toxicities or discontinuations due to adverse effects[21]. These results underscore lepodisiran’s potential as a durable, safe, and effective agent for Lp(a) lowering.
Olpasiran, another GalNAc-siRNA developed by Amgen, targets the same LPA mRNA. The OCEAN(a)-DOSE trial demonstrated > 95% reductions in Lp(a), but its shorter duration necessitates more frequent dosing (monthly to quar
Pelacarsen, an antisense oligonucleotide developed by Novartis/Ionis, achieves up to 80% Lp(a) reductions but requires monthly injections and has a higher incidence of injection site reactions[13]. It is currently being evaluated in the Lp(a) HORIZON outcome trial.
Lepodisiran advantages: (1) Longest duration of effect (6-12 months dosing); (2) Comparable or superior efficacy; and (3) High tolerability and minimal systemic side effects[17,21]. The comparison is given in Table 1.
Feature | Lepodisiran | Olpasiran | Pelacarsen |
Type | GalNAc-conjugated small interfering RNA (siRNA) | GalNAc-conjugated siRNA | Antisense oligonucleotide |
Target | LPA mRNA (hepatocytes) | LPA mRNA (hepatocytes) | LPA mRNA (hepatocytes) |
Mechanism | RNA interference degrades LPA mRNA | RNA interference inhibits apo(a) synthesis | RNase H-mediated degradation of LPA mRNA |
Dosing frequency | Twice yearly (biannual) | Every 12-24 weeks depending on dose | Monthly subcutaneous injection |
Route | Subcutaneous | Subcutaneous | Subcutaneous |
Lp(a) reduction | > 90% reduction sustained for up to 48 weeks | Approximately 90%-95% reduction in phase 2 data | Approximately 80% reduction (dose-dependent) |
Duration of action | Long (effects up to 1 year with single dose) | Moderate to long (dose-dependent) | Shorter; Requires regular monthly dosing |
Phase of trials | Phase 2 completed; Phase 3 ongoing (ALPINE program) | Phase 2 completed; Phase 3 ongoing [OCEAN(a)-outcomes] | Phase 3 ongoing [Lp(a) HORIZON trial] |
Safety profile | Well tolerated; Mild injection-site reactions | Well tolerated; Mild side effects noted | Generally safe; Injection-site reactions, minor flu-like symptoms |
Notable advantage | Longest dosing interval | Rapid onset, high potency | Extensive phase 3 data under development |
A pivotal phase 3 cardiovascular outcome trial is underway to determine whether Lp(a) lowering with lepodisiran reduces MACE in high-risk populations[21]. Subgroup analyses are expected to clarify: (1) Genetic variability in Lp(a) response[8]; (2) Efficacy in diverse ethnic populations[20]; (3) Effects on aortic stenosis progression[10]; (4) Long-term hepatic and renal safety[19]; and (5) Combination strategies with statins or PCSK9 inhibitors[12,20].
Currently, routine Lp(a) screening is recommended for individuals with premature ASCVD or a family history of early cardiovascular events[11]. However, no specific pharmacologic treatment targets Lp(a) in current guidelines due to a lack of conclusive outcome data. Lepodisiran’s durability, high efficacy [> 90% Lp(a) reduction], and favorable safety profile may eventually shift this paradigm. With accumulating evidence, especially if ongoing phase 3 trials confirm a reduction in MACE, guideline bodies may incorporate Lp(a) reduction as a formal therapeutic objective for high-risk populations[11,20]. If MACE benefit is validated, clinical guidelines could expand to recommend Lp(a) screening more broadly i.e., potentially for all patients with established ASCVD or those with Lp(a) ≥ 50 mg/dL, even in the absence of other lipid abnormalities. Moreover, treatment thresholds may be redefined, and Lp(a)-targeted therapies like lepodisiran may become part of standard lipid-lowering protocols in selected populations[20].
Despite promising early-phase data, several key questions remain regarding the clinical use of lepodisiran for Lp(a)-mediated ASCVD. Foremost among these is whether the substantial reduction in Lp(a) achieved with lepodisiran exceeding 90% in some trials will translate into meaningful reductions in cardiovascular events such as myocardial infarction, stroke, or cardiovascular death. While elevated Lp(a) is a well-established independent risk factor, definitive evidence of outcome benefit is awaited from ongoing phase 3 trials. Another unresolved issue concerns patient selection. It remains unclear whether lepodisiran should be used broadly in individuals with elevated Lp(a) for primary prevention or restricted to those with established ASCVD and markedly elevated levels. Additionally, long-term safety must be monitored closely, as the physiological roles of Lp(a), including wound healing and host defense, may be impacted by chronic suppression of apo(a) synthesis[15].
The optimal dosing strategy also warrants further evaluation. Although current trials support twice-yearly administration, it is uncertain whether dose adjustments based on body weight or Lp(a) levels will be required in clinical practice. Comparative effectiveness is another area of interest; lepodisiran must be evaluated against other emerging Lp(a)-lowering agents such as pelacarsen and olpasiran to determine superiority or potential combination benefits. Moreover, economic considerations will play a significant role in determining accessibility, as the cost-effectiveness of lepodisiran remains to be established, particularly for use in populations without established CVD. Lastly, there is an ongoing debate regarding whether Lp(a) should be routinely measured in all adults as part of cardiovascular risk stratification. If lepodisiran is approved, it may prompt guideline updates advocating universal or risk-based Lp(a) screening[16] as summarized in Table 2.
Section | Key Information |
Global CVD burden | Approximately 17.9 million deaths annually; 32% of global mortality[1] |
Lp(a): Role and structure | LDL-like particle with apoB-100 + apo(a); Apo(a) homologous to plasminogen; Pro-atherogenic and pro-thrombotic[5,6] |
Genetic basis | LPA gene on chromosome 6q26-27; KIV-2 copy number explains up to 90% of Lp(a) variability[7,8,9] |
Clinical significance | Elevated Lp(a) linked with CHD, stroke, aortic stenosis; 2.5 × higher CHD risk in top Lp(a) tertile[2,3,10] |
Guidelines | ACC/AHA recognizes Lp(a) as a risk-enhancing factor[11] |
Limitations of existing therapies | Statins (increase) Lp(a) 20%-25%[4]; PCSK9 inhibitors (decrease); Lp(a) approximately 20%-30%[12]; Niacin poorly tolerated[13]; Apheresis effective but invasive and costly[14] |
siRNA approach | GalNAc-siRNA targets liver via ASGPR; siRNA-RISC complex degrades LPA mRNA[15,16] |
Lepodisiran: Mechanism | Subcutaneous GalNAc-siRNA that inhibits apo(a) synthesis; Long half-life allows 6-12 months dosing[16,17,19] |
Preclinical data | Up to 90% Lp(a) reduction in animal models; No toxicity observed[18] |
Phase 1 data | Single dose (100-400 mg), up to 94% Lp(a) (decrease) lasting > 180 days; No serious adverse events[17] |
Phase 2 (ALPACA) | Single 400 mg dose: 93.9% (decrease) (day 60-180); Two doses: 94.8% (decrease) up to 1 year; 74.2% (decrease) even at day 360; Excellent safety profile[21] |
Comparison with other therapies | Olpasiran: > 95% (decrease); Monthly dosing[22]; Pelacarsen: 80% (decrease); Monthly; More injection site reactions[13]; Lepodisiran: Comparable efficacy; Longest duration; Highly tolerable[17,21] |
Ongoing research | Phase 3 MACE outcomes trial evaluating genetic/ethnic variability, safety, and combination therapies[8,10,19,20] |
Clinical implications | Lp(a) testing in premature ASCVD or family history; Potential future inclusion of Lp(a)-lowering in guidelines[11,20] |
If lepodisiran is proven effective in reducing MACE, its biannual dosing and high adherence potential could lead to substantial long-term economic benefits. These may include reduced medication costs, fewer healthcare visits, and lower rates of hospitalizations and interventions, ultimately making it a cost-effective strategy for ASCVD prevention in patients with elevated Lp(a)[17]. While lepodisiran shows promising efficacy, long-term safety data remain limited, warranting cautious optimism. Additionally, potential racial and ethnic disparities in Lp(a) levels and treatment response require further investigation.
Lepodisiran represents a milestone in cardiovascular therapeutics, uniquely addressing the unmet need of Lp(a) lowering. Its RNA-based mechanism offers durable suppression of apo(a) synthesis, translating to sustained reductions in plasma Lp(a). Clinical data to date affirm its safety and tolerability, and ongoing trials may cement its role in reducing cardiovascular events. However, key concerns remain: The phase 3 trials must validate its long-term impact on MACE, and the current evidence base is largely derived from Caucasian populations, highlighting the need for broader racial and ethnic representation. If these challenges are addressed, lepodisiran could reshape the landscape of personalized cardio
1. | Mendis S, Graham I, Narula J. Addressing the Global Burden of Cardiovascular Diseases; Need for Scalable and Sustainable Frameworks. Glob Heart. 2022;17:48. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 47] [Reference Citation Analysis (0)] |
2. | Lu W, Cheng YC, Chen K, Wang H, Gerhard GS, Still CD, Chu X, Yang R, Parihar A, O'Connell JR, Pollin TI, Angles-Cano E, Quon MJ, Mitchell BD, Shuldiner AR, Fu M. Evidence for several independent genetic variants affecting lipoprotein (a) cholesterol levels. Hum Mol Genet. 2015;24:2390-2400. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 46] [Cited by in RCA: 40] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
3. | Nordestgaard BG, Langsted A. Lipoprotein (a) as a cause of cardiovascular disease: insights from epidemiology, genetics, and biology. J Lipid Res. 2016;57:1953-1975. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 270] [Cited by in RCA: 395] [Article Influence: 43.9] [Reference Citation Analysis (0)] |
4. | Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW, Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith SC Jr, Sperling L, Virani SS, Yeboah J. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2019;73:e285-e350. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1551] [Cited by in RCA: 1650] [Article Influence: 275.0] [Reference Citation Analysis (0)] |
5. | Tsimikas S. A Test in Context: Lipoprotein(a): Diagnosis, Prognosis, Controversies, and Emerging Therapies. J Am Coll Cardiol. 2017;69:692-711. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 460] [Cited by in RCA: 717] [Article Influence: 89.6] [Reference Citation Analysis (0)] |
6. | Kamstrup PR, Tybjaerg-Hansen A, Steffensen R, Nordestgaard BG. Genetically elevated lipoprotein(a) and increased risk of myocardial infarction. JAMA. 2009;301:2331-2339. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 846] [Cited by in RCA: 981] [Article Influence: 61.3] [Reference Citation Analysis (0)] |
7. | Thanassoulis G, Campbell CY, Owens DS, Smith JG, Smith AV, Peloso GM, Kerr KF, Pechlivanis S, Budoff MJ, Harris TB, Malhotra R, O'Brien KD, Kamstrup PR, Nordestgaard BG, Tybjaerg-Hansen A, Allison MA, Aspelund T, Criqui MH, Heckbert SR, Hwang SJ, Liu Y, Sjogren M, van der Pals J, Kälsch H, Mühleisen TW, Nöthen MM, Cupples LA, Caslake M, Di Angelantonio E, Danesh J, Rotter JI, Sigurdsson S, Wong Q, Erbel R, Kathiresan S, Melander O, Gudnason V, O'Donnell CJ, Post WS; CHARGE Extracoronary Calcium Working Group. Genetic associations with valvular calcification and aortic stenosis. N Engl J Med. 2013;368:503-512. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 785] [Cited by in RCA: 737] [Article Influence: 61.4] [Reference Citation Analysis (0)] |
8. | Leren TP, Bogsrud MP. Cascade screening for familial hypercholesterolemia should be organized at a national level. Curr Opin Lipidol. 2022;33:231-236. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 7] [Article Influence: 2.3] [Reference Citation Analysis (0)] |
9. | Camm AJ, Amarenco P, Haas S, Hess S, Kirchhof P, Kuhls S, van Eickels M, Turpie AG; XANTUS Investigators. XANTUS: a real-world, prospective, observational study of patients treated with rivaroxaban for stroke prevention in atrial fibrillation. Eur Heart J. 2016;37:1145-1153. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 341] [Cited by in RCA: 340] [Article Influence: 37.8] [Reference Citation Analysis (0)] |
10. | Tavori H, Giunzioni I, Fazio S. PCSK9 inhibition to reduce cardiovascular disease risk: recent findings from the biology of PCSK9. Curr Opin Endocrinol Diabetes Obes. 2015;22:126-132. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 23] [Cited by in RCA: 26] [Article Influence: 2.6] [Reference Citation Analysis (0)] |
11. | Sahebkar A, Reiner Ž, Simental-Mendía LE, Ferretti G, Cicero AF. Effect of extended-release niacin on plasma lipoprotein(a) levels: A systematic review and meta-analysis of randomized placebo-controlled trials. Metabolism. 2016;65:1664-1678. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 105] [Cited by in RCA: 94] [Article Influence: 10.4] [Reference Citation Analysis (0)] |
12. | Williams L, Wilson DP. Editorial commentary: Dietary management of familial chylomicronemia syndrome. J Clin Lipidol. 2016;10:462-465. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 18] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
13. | Moriarty PM, Thompson PD, Cannon CP, Guyton JR, Bergeron J, Zieve FJ, Bruckert E, Jacobson TA, Baccara-Dinet MT, Zhao J, Donahue S, Ali S, Manvelian G, Pordy R. Efficacy and safety of alirocumab in statin-intolerant patients over 3 years: open-label treatment period of the ODYSSEY ALTERNATIVE trial. J Clin Lipidol. 2020;14:88-97.e2. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 6] [Cited by in RCA: 9] [Article Influence: 1.8] [Reference Citation Analysis (0)] |
14. | Zimmermann TS, Karsten V, Chan A, Chiesa J, Boyce M, Bettencourt BR, Hutabarat R, Nochur S, Vaishnaw A, Gollob J. Clinical Proof of Concept for a Novel Hepatocyte-Targeting GalNAc-siRNA Conjugate. Mol Ther. 2017;25:71-78. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 108] [Cited by in RCA: 153] [Article Influence: 19.1] [Reference Citation Analysis (0)] |
15. | Swerdlow DI, Rider DA, Yavari A, Wikström Lindholm M, Campion GV, Nissen SE. Treatment and prevention of lipoprotein(a)-mediated cardiovascular disease: the emerging potential of RNA interference therapeutics. Cardiovasc Res. 2022;118:1218-1231. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 11] [Cited by in RCA: 36] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
16. | Tsimikas S, Fazio S, Ferdinand KC, Ginsberg HN, Koschinsky ML, Marcovina SM, Moriarty PM, Rader DJ, Remaley AT, Reyes-Soffer G, Santos RD, Thanassoulis G, Witztum JL, Danthi S, Olive M, Liu L. NHLBI Working Group Recommendations to Reduce Lipoprotein(a)-Mediated Risk of Cardiovascular Disease and Aortic Stenosis. J Am Coll Cardiol. 2018;71:177-192. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 230] [Cited by in RCA: 364] [Article Influence: 60.7] [Reference Citation Analysis (0)] |
17. | Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, Tardif JC, Baum SJ, Steinhagen-Thiessen E, Shapiro MD, Stroes ES, Moriarty PM, Nordestgaard BG, Xia S, Guerriero J, Viney NJ, O'Dea L, Witztum JL; AKCEA-APO(a)-LRx Study Investigators. Lipoprotein(a) Reduction in Persons with Cardiovascular Disease. N Engl J Med. 2020;382:244-255. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 406] [Cited by in RCA: 650] [Article Influence: 130.0] [Reference Citation Analysis (0)] |
18. | Viney NJ, van Capelleveen JC, Geary RS, Xia S, Tami JA, Yu RZ, Marcovina SM, Hughes SG, Graham MJ, Crooke RM, Crooke ST, Witztum JL, Stroes ES, Tsimikas S. Antisense oligonucleotides targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind, placebo-controlled, dose-ranging trials. Lancet. 2016;388:2239-2253. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 500] [Cited by in RCA: 583] [Article Influence: 64.8] [Reference Citation Analysis (0)] |
19. | Corrigendum to: HFp2EF: heart failure with pulmonary dysfunction and preserved ejection fraction? Eur Heart J. 2023;44:380. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
20. | Deedwania P, Acharya T. Anticoagulation in Atrial Fibrillation: Is the Paradigm Really Shifting? J Am Coll Cardiol. 2017;69:786-788. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 4] [Article Influence: 0.7] [Reference Citation Analysis (0)] |
21. | Bhatnagar D, Bhatnagar P. Nutrition and its impact on cardiovascular disease. Curr Opin Lipidol. 2022;33:211-212. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
22. | Ho JE, Liu C, Lyass A, Courchesne P, Pencina MJ, Vasan RS, Larson MG, Levy D. Galectin-3, a marker of cardiac fibrosis, predicts incident heart failure in the community. J Am Coll Cardiol. 2012;60:1249-1256. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 460] [Cited by in RCA: 455] [Article Influence: 35.0] [Reference Citation Analysis (0)] |