Abstract

Calcific aortic valve disease (CAVD) resulting in aortic stenosis (AS) is the most common form of valvular heart disease, affecting 2% of those over age 65. Those who develop symptomatic severe AS have an average further lifespan of <2 years without valve replacement, and three-quarters of these patients will develop heart failure, undergo valve replacement, or die within 5 years. There are no approved pharmaceutical therapies for AS, due primarily to a limited understanding of the molecular mechanisms that direct CAVD progression in the complex haemodynamic environment. Here, advances in efforts to understand the pathogenesis of CAVD and to identify putative drug targets derived from recent multi-omics studies [including (epi)genomics, transcriptomics, proteomics, and metabolomics] of blood and valvular tissues are reviewed. The recent explosion of single-cell omics-based studies in CAVD and the pathobiological and potential drug discovery insights gained from the application of omics to this disease area are a primary focus. Lastly, the translation of knowledge gained in valvular pathobiology into clinical therapies is addressed, with a particular emphasis on treatment regimens that consider sex-specific, renal, and lipid-mediated contributors to CAVD, and ongoing Phase I/II/III trials aimed at the prevention/treatment of AS are described.

Calcific aortic valve disease (CAVD) results from a complex physiological microenvironment and multifaceted interplay amongst a number of putative initiators and drivers of disease. Multi-omics enables a holistic assessment of molecular drivers of CAVD via quantitation of the (epi)genome, transcriptome, proteome, and metabolome at both a bulk and single-cell level. Integration of multiple omics modalities facilitates the identification of novel, subtle, or cross-layer molecules and interactions. Target prioritization via systems biology precedes drug development and eventual pharmaceutical alternatives to surgical aortic valve replacement or transcatheter aortic valve replacement.
Graphical abstract

Calcific aortic valve disease (CAVD) results from a complex physiological microenvironment and multifaceted interplay amongst a number of putative initiators and drivers of disease. Multi-omics enables a holistic assessment of molecular drivers of CAVD via quantitation of the (epi)genome, transcriptome, proteome, and metabolome at both a bulk and single-cell level. Integration of multiple omics modalities facilitates the identification of novel, subtle, or cross-layer molecules and interactions. Target prioritization via systems biology precedes drug development and eventual pharmaceutical alternatives to surgical aortic valve replacement or transcatheter aortic valve replacement.

Introduction

Calcific aortic valve disease (CAVD) resulting in aortic stenosis (AS) is a significant driver of cardiovascular morbidity and mortality and encompasses a spectrum of early leaflet fibrosis and thickening [aortic valve (AV) sclerosis, present in ∼25% of seniors] to highly fibrocalcific and symptomatic AS affecting nearly 3% of those over age 75.1 Beyond surgical aortic valve replacement (AVR, SAVR) or transcatheter AVR (TAVR), there are no approved pharmaceutical therapies for AS. As a result, some ∼400 000 AVR procedures are performed annually worldwide.2 These interventions are specialized, resource-intensive (>$10 billion/year in the USA alone3), invasive, and not without risk.4 In addition, structural valve degeneration of implanted bioprosthetic valves limits their lifespan to roughly a decade after implantation.5 As AVRs expand into younger patient cohorts who are projected to require multiple valve re-interventions and replacements in their lives due to repeated bioprosthetic valve failure, these technological limitations have sparked discussion and implementation of lifetime management strategies to the point where preemptive aortic root enlargement, early pulmonary autograft replacements, and valve-in-valve-in-valve procedures are contemplated and enacted.6,7 If left untreated after the onset of symptoms, most patients with severe AS die within 2 years,8 and the vast majority will require invasive AVR or suffer from heart failure or death within half a decade.9 This grim prognosis—worse than that of many cancers—only serves to place in stark relief the limited therapeutic options available for patients suffering from AS. There are also important implications for global health equity: concurrent epidemics of aging, obesity, diabetes, and chronic kidney disease are predicted to lead to a rapid 2.5-fold increase in global AS incidence in the next 15 years which will be particularly concentrated in low- and middle-income countries, developing countries and the global south.10–12 In turn, these regions have a critical lack of capacity for cardiac surgery or interventional cardiology,13–15 and will be unable to leverage the otherwise-revolutionary benefits to access and equity of care provided by TAVR. Even within high-income and developed countries, massive racial, ethnic, and socioeconomic disparities exist in risk and/or diagnosis of AS,16,17 access to TAVR,18 and outcomes after AVR.19,20 In the face of these many challenges, there is a clear and urgent unmet need to unravel CAVD mechanisms, discover molecular targets, and drive the development of novel pharmacotherapies.21

The anatomy of the AV is intricate, with a tri-layered architecture ∼1 mm thick:22 the collagen-rich fibrosa faces the ascending aorta, the elastin-rich ventricularis is positioned next to the left ventricle, and the proteoglycan-rich spongiosa rests in between.23 In normal tissues, two main mechanosensitive cell types dominate the valve: quiescent, fibroblastic valvular interstitial cells (VICs) are found throughout the three leaflet layers,24 while a monolayer of valvular endothelial cells (VECs) lines the blood-contacting surfaces of the leaflet.25 Disease occurs preferentially in the fibrosa,26 where tissue and blood biomechanics, lipid-mediated inflammation, extracellular vesicles (EVs), platelet activation, myofibroblastic/osteoblastic/chondrogenic differentiation, immune cell infiltration, endothelial-to-mesenchymal transition (EndMT), aberrant extracellular matrix (ECM) turnover, fibrosis, impaired calcium and phosphate metabolism, cellular senescence, and other as-yet undiscovered mechanisms all conspire, to one degree or another, to actively drive tissue fibrosis, leaflet thickening, and mineralization.24,27–36 This leads inexorably towards impaired leaflet opening, left ventricular remodelling, heart failure, and eventual death (Figure 1).

Natural progression of calcific aortic valve disease. Prior to the onset of symptoms or impairment of cardiac function, initial activation of resident valvular interstitial cells and valvular endothelial cells towards a myofibrogenic lineage by a combination of cardiometabolic, genetic, renal, and congenital risk factors leads to an intermediate disease stage marked by sclerotic valve leaflet thickening. Subsequently, in a small proportion of patients with valvular sclerosis (∼2% yearly), still largely unknown mechanisms drive valve cell osteogenesis and result in leaflet mineralization which reduces valve opening, increases left ventricular afterload, and induces cardiac hypertrophy. Left untreated, these patients typically progress rapidly to heart failure: within 5 years of developing symptomatic aortic stenosis, ∼75% will require aortic valve replacement or die
Figure 1

Natural progression of calcific aortic valve disease. Prior to the onset of symptoms or impairment of cardiac function, initial activation of resident valvular interstitial cells and valvular endothelial cells towards a myofibrogenic lineage by a combination of cardiometabolic, genetic, renal, and congenital risk factors leads to an intermediate disease stage marked by sclerotic valve leaflet thickening. Subsequently, in a small proportion of patients with valvular sclerosis (∼2% yearly), still largely unknown mechanisms drive valve cell osteogenesis and result in leaflet mineralization which reduces valve opening, increases left ventricular afterload, and induces cardiac hypertrophy. Left untreated, these patients typically progress rapidly to heart failure: within 5 years of developing symptomatic aortic stenosis, ∼75% will require aortic valve replacement or die

Much of the failure to bring effective CAVD treatments to bear is the result of still-limited understanding of the molecular mechanisms that govern pathogenesis in the AV. Initiation and progression of CAVD is highly complex and multi-factorial, existing in vitro and in vivo models of disease remain imperfect,37,38 and the standard of care-driven absence of excised human tissue samples from normal, sclerotic, or mild/moderate AS has potentiated disease stage-specific segmentation of end-stage specimens.30,39–41 When paired with cutting-edge systems biology approaches, multi-omics of the valvular (epi)genome, transcriptome, proteome, and metabolome hold significant promise to enable greater holistic comprehension of the molecular fingerprint of CAVD via improved identification of risk genes, circulating biomarkers, or disease-driving proteins, lipids, and metabolites.42,43 These strategies may enable the rigorous definition of healthy baselines, building new prediction models of disease progression, the benchmarking of in vitro, in vivo, or in silico disease model performance against CAVD in humans, or improved characterization of valve cell heterogeneity to allow cell- or tissue-specific drug targeting. Most importantly, these technologies permit the bench-to-bedside prioritization of novel signalling pathways and individual molecules as promising targets befitting of investment in validation, drug development, and clinical trials (Figure 2). While pharmacotherapy development is challenged by the multifaceted nature of AS initiation and progression, multi-omics, systems biology, and network medicine approaches may allow the hierarchical characterization of disease to enable the targeting of mechanisms that are highly upstream or downstream of calcification, and the quantitative prioritization of molecular pathways with an outsized contribution to disease progression. Due to the rising popularity and near-ubiquity of this approach, contemporary (multi-)omics studies are an important and useful lens through which to observe the progress of basic and translational science. Prior multi-omics-driven advances in CAVD have been reviewed elsewhere,42,43 and we will focus the bulk of this review on recent progress in the study of pathogenic mechanisms of CAVD, and position integrative physiology as one means by which new technologies may deliver pharmacotherapeutic relief of this disease via identification of novel drug targets and treatments.44

Multi-omics drug discovery pipeline in calcific aortic valve disease. Depending on study goals (e.g. mechanisms of pathogenesis, cell type-specific targets, biomarker discovery), multi-omic screening studies can be performed on valvular cells, tissues, plasma/serum, blood-/tissue-derived extracellular vesicles, and other relevant sample types. Quantification of differential molecular abundances between experimental groups of interest, integration of insights from multiple omics modalities, and unbiased target prioritization via network analysis derives target lists for follow-up translational studies. Validation of top targets can occur via well-established in silico, in vitro, or in vivo disease models that recapitulate specific aspects of calcific aortic valve disease pathogenesis relevant to the target’s putative mechanism of action. High-throughput screening of pre-existing small molecule libraries, engineering of targeted biologics, and other drug development approaches lead to pre-clinical and clinical trials to assess safety and efficacy. Iteration and optimization using samples derived from pre-clinical models and Phase I/II/III studies in humans may add further insight into (non-)responding sub-groups, off-target effects, mechanisms of action, and label extension/expansion.
Figure 2

Multi-omics drug discovery pipeline in calcific aortic valve disease. Depending on study goals (e.g. mechanisms of pathogenesis, cell type-specific targets, biomarker discovery), multi-omic screening studies can be performed on valvular cells, tissues, plasma/serum, blood-/tissue-derived extracellular vesicles, and other relevant sample types. Quantification of differential molecular abundances between experimental groups of interest, integration of insights from multiple omics modalities, and unbiased target prioritization via network analysis derives target lists for follow-up translational studies. Validation of top targets can occur via well-established in silico, in vitro, or in vivo disease models that recapitulate specific aspects of calcific aortic valve disease pathogenesis relevant to the target’s putative mechanism of action. High-throughput screening of pre-existing small molecule libraries, engineering of targeted biologics, and other drug development approaches lead to pre-clinical and clinical trials to assess safety and efficacy. Iteration and optimization using samples derived from pre-clinical models and Phase I/II/III studies in humans may add further insight into (non-)responding sub-groups, off-target effects, mechanisms of action, and label extension/expansion.

Assessing (epi-)genetic regulation and risk in aortic stenosis

After early landmark studies revealed LPA as the first genetic driver of AS risk,45 follow-on work in larger cohorts continued to confirm this association and identified additional risk alleles in PALMD, TEX41, IL6, and ALPL.46–48 More recently, an explosion of high-quality, large-scale GWAS (genome-wide association studies) has revolutionized our understanding of the depth and breadth of genetic risk in AS. These meta-analysis studies leverage large cohorts (nearly 15 000 cases and 1 000 000 controls) to both replicate prior findings and add tens of novel risk genes to the pool of AS contributors, including the replicated loci CEP85L, FTO, SLMAP, CELSR2-SORT1, MECOM, CDAN1, NLRP6, and SMC2.49–51 Small et al.49 performed a multi-ancestry GWAS analysis that identified ancestry-specific SNPs and identified shared and differential genomic drivers of AS and atherosclerosis, Larsson et al.52 found via Mendelian randomization (MR) that genetically predicted body mass index and adiposity were most strongly associated with AS vs. other cardiovascular diseases, Chen et al.50 generated cross-ancestry analyses, genetic risk scores, and linked causal roles for dyslipidemia and adiposity to AS, while Thériault et al.51 performed RNA-sequencing in a cohort of 500 human aortic valves for confirmatory genome-wide eQTL (expression quantitative trait loci) analyses and TWAS (transcriptome-wide association studies). In all cases, leveraging of sophisticated phenome-wide association studies, MR, colocalization analyses with tissue expression data, TWAS, etc. have become powerful and standard tools for interpretation and translation in genomic studies of AS. One notable challenge resulting from this dramatic expansion of cohort size, study power, and genome-wide significant risk alleles is the need to accurately annotate/map tens or hundreds of significant SNPs to onto protein-coding genes; this is particularly non-trivial as the complexity of genomic regulation continues to be unravelled. As additional risk alleles are identified, polygenic risk scores (PRSs) in AS have now been generated that reveal a weak correlation between PRS of AS vs. coronary artery disease (CAD), consistent with reports of unique whole blood transcriptional signatures between these conditions.53,54 While the AS PRS performs comparably to the vast majority of clinical risk factors, they do not yet exceed these traditional clinical approaches and further development is required.53

Bicuspid aortic valve (BAV) is the most common congenital heart defect and drives the early and accelerated onset of AS through mechanisms which remain largely unelucidated.55 One hypothetical aetiology arises from the colocalization of BAV calcification with well-described abnormal haemodynamic forces and altered intra-leaflet mechanical stress concentrations found in BAVs,56,57 though a causal role has yet to be established and separation of mechanical and genetic drivers in vivo is challenging. Genotype–phenotype linkages in BAV are complicated, the heritability of BAV is highly polygenic, and BAVs are found in only ∼2% of humans.58 This context complicates the identification of genetic causes of BAV and BAV-associated AS. Initial GWAS in 466 cases and 4660 controls found a non-coding loss-of-function variant of the cardiac-specific transcription factor GATA4,59 while recent studies in larger cohorts detected BAV risk loci in MUC4 (deleterious), PALMD (loss-of-function), TEX41, and MIB1 (loss-of-function).58,60 Confirmatory experiments revealed that disruption of Muc4, a gene involved in epithelial-to-mesenchymal transformation, impaired AV development in zebrafish and linked genomic risk drivers to aberrant endothelial transcriptomic profiles. This finding, along with evidence from CRISPR-iPSC lines showing GATA4 reductions inhibited EndMT, raise the importance of this pathway in AV development and disease.58,61 In turn, the E3-ubiquitin ligase MIB1 was shown to be essential for the activation of NOTCH signalling during AV development and implicated in ∼2% of non-syndromic cases of BAV.60 Other studies have leveraged targeted DNA sequencing of HOXA1 and ADAMTS family genes, phenotype screening in zebrafish, and RNA-sequencing in knockout mice to implicate loss-of-function of these well-described regulators of cardiovascular development in human BAV incidence.62,63 Beyond early transcriptomics work,64–66 there is a notable absence of multi-omics studies aimed at differentiating mechanisms of BAV-driven AS from those of prototypical tricuspid aortic valve (TAV)-associated AS. In practice, it is challenging to conduct GWAS studies of AS exclusively on individuals with TAV and most GWAS studies of AS make no distinction between valvular morphotypes.

Germline point mutations are not the only DNA-associated drivers of CAVD, as evidenced by prior seminal works in AS epigenetics that found (i) hypomethylation drove expression of the lncRNA H19, impaired NOTCH signalling, and VIC calcification67 and (ii) methylation of the intronic enhancer of PLPP3 (phospholipid phosphatase-3) attenuated the degradation of lipoprotein(a) [Lp(a)] metabolites and drove pro-osteogenic VIC responses.68 In a particularly elegant study, Han and colleagues69 revealed a putative pathogenic mechanism for the GWAS-identified CAVD risk gene and m6A methyltransferase FTO.49–51 By sequencing non-coding PIWI-interacting RNAs (piRNA) in calcified human AVs, they identified elevated hsa-piR-25624, or AVCAPIR (AV calcification-associated piRNA), which induced VIC calcification in vitro. Using mass spectrometric proteomics, they identified FTO as an AVCAPIR binding partner, then employed methylated RNA IP-seq (m6A-seq) and RNA-seq to show this binding blocks FTO-mediated demethylation of CD36 mRNA. Highly methylated CD36 mRNA is recognized by the m6a reader IGF2BP1, which enhances the stability and abundance of CD36 mRNA and, thus, protein. Elevated CD36 protein levels directly bind to, stabilize, and promote the abundance of pro-calcific PCSK9 protein, leading to progressive CAVD through pleiotropic (non-lipid) downstream mechanisms.69 Clonal haematopoiesis of indeterminate potential (CHIP)-driving mutations have recently emerged as a key and novel cause of atherosclerosis70 and targeted amplicon sequencing has revealed that approximately a third of patients undergoing AVR carry these somatic mutations and suffer from elevated mortality after TAVR.71 Mechanistic insight into this phenomenon came from bulk transcriptomics (RNA-seq) of human valve tissues from SAVR recipients who were CHIP- and non-CHIP-mutation carriers; these experiments revealed broad inflammatory cell infiltration and inflammatory signalling in CHIP-carrier AVs with a particular exacerbation of activated B cells.72 Somatic alterations may also contribute to well-described sex-based differences in CAVD pathogenesis, whereby in both TAVs and BAVs, female AS patients have lower calcification levels but elevated leaflet fibrosis while incurring similar haemodynamic severity of disease.73–75 Mosaic loss of chromosome Y (LOY) in circulating blood cells is the most common acquired mutation and is strongly associated with major adverse cardiovascular events (MACE) and heart failure in men.76,77 After identifying exacerbated LOY and correlations with increased mortality in male TAVR recipients, Mas-Peiro et al.78 performed single-cell RNA-seq (scRNA-seq) on monocytes with LOY to identify a TGFβ-mediated pro-fibrotic transcriptional programme in these patients, though the relative contributions of LOY to cardiac vs. valvular fibrosis remains to be elucidated. On the other hand, genes that escape X-chromosome inactivation also appear to regulate myofibrogenic VIC differentiation and resultant CAVD: transcriptomics of male- and female-derived porcine VICs in hydrogel culture systems found that elevated Rho-associated protein kinase (ROCK) signalling enabled the genes BMX and STS to avoid inactivation and elevate female VIC myofibrogenesis.79 As a whole, these findings are consistent with several prior transcriptomics studies which have demonstrated elevated pro-fibrotic pathways in female AS leaflet tissues.80,81

Multi-omics and integrative approaches to pathobiological insight

Mechanisms of genetic risk

Beyond the genome, efforts to capture global, omics-based insights into valvular phenotypes have been successfully applied in a widespread variety of contexts, and fall under two primary avenues: first, the identification of new mechanisms of homeostasis or pathogenesis through non-biased screening. Second, to confirm clinical findings or validate targets generated by hypothesis-driven research. In some cases, omics approaches can assist in understanding AV-targeted drug side effects: transcriptome arrays have revealed that downregulation of discoidin domain receptor 2 (DDR2), the target of the chronic myeloid leukaemia treatment Nilotinib, is present in calcified human AVs. In vitro experiments found that VICs treated with Nilotinib underwent robust osteogenesis and mineralization, suggesting a potential contribution to CAVD-associated cardiotoxicity.82 In other ways, multi-omics enables the construction of mechanistic linkages between genotypic drivers of CAVD and disease phenotypes. The paralemmin protein Palmdelphin (PALMD) has also long been connected by genomic evidence to AS onset in humans, though only recently have the downstream mechanisms been clarified. By mining murine scRNA-seq datasets, Sáinz-Jaspeado et al.41 found Palmd was highly expressed in the endothelium and controlled endothelial actin cap formation and nuclear alignment in response to fluid flow, then went on to show how PALMD exerted RAN GTPase-dependent control of nucleocytoplasmic shuttling via XPO1. Human CAVD tissue transcriptomics found disease stage-specific reductions in PALMD and suggested that nuclear fragility caused by loss of PALMD-dependent nucleocytoplasmic shuttling underpins genetic risk associated with SNPs in this gene. Importantly, the association of PALMD with bicuspid AS47,58 could be explained by this shear-sensitive mechanosensitive nuclear resilience mechanism, as well as potential control over EndMT.83 Palmdelphin appears to exert pleiotropic effects on cellular actin stress fibre dynamics: Chignon et al.84 employed 3D genetic mapping, CRISPR activation screens, and DNA-binding assays to demonstrate that an AS risk variant at 1p21.2 controlled PALMD expression, and that the risk variant altered DNA shape in such a way as to lower binding of the transcription factor NFATC2 and resultant PALMD expression. The authors went on to examine the PALMD protein–protein interactome by mass spectrometry and identified a role for PALMD in regulating actin polymerization and exerted control of VIC fibrogenesis via myocardin-related transcription factor. Contemporary AS GWAS studies have also identified the CELSR2–SORT1 risk locus.49–51 The SORT1 protein Sortilin has been extensively studied in the context of EV-mediated vascular calcification,85 though its role in CAVD was unclear until Iqbal and colleagues showed Sortilin-deficient mice with wire-injured AVs were protected against aortic valve fibrocalcification. Proteomics and scRNA-seq of cultured human VICs revealed that Sortilin loss-of-function prevented calcification, and that Sortilin signalled via p38 MAPK to drive expansion of a complex inflammatory-myofibroblastic-osteogenic VIC subtype in vitro.86

Inflammatory drivers of calcific aortic valve disease

Some of the earliest indications of a potential role for inflammatory drivers in AS stem from those associated with infection. Indeed, the incidence of infective endocarditis in patients with AS is a function of AS severity,87 multiple bacterial species are present in the majority of stenotic aortic valves that undergo replacement,88 inoculation of the rabbit aortic valve with Corynebacterium matruchotti and/or Streptococcus sanguis II is sufficient to induce large macrocalcifications,89 and proteomics has recently implicated autoimmunity-associated inflammatory responses in the progression of rheumatic aortic valve disease.90 Among the genetically identified targets (Table 1), the pivotal incrimination of the IL6 gene (encoding the pro-inflammatory cytokine interleukin 6) as a driver of genetic AS risk48 emphasized the role of inflammatory processes in CAVD. Toll-like receptors (TLRs), an evolutionarily conserved family of pattern recognition receptors, are typically activated by pathogen detection and endogenous alarm signals from injured cells. Activation of TLR2 and TLR4 on VICs is directly linked to procalcification pathways.94 In addition, recent evidence identified TLR3 activation in aortic valves leading to osteogenic VIC differentiation through type 1 interferon (IFN) signalling.95 Systematic screening identified the proteoglycan Biglycan (BGN) as a novel TLR3 ligand, requiring a xylosyltransferase-mediated maturation of BGN’s glycosaminoglycan side chains. The functional impact of TLR3 signalling in AS has received support from the protective effects against CAVD of in vivo models with a genetically targeted BGN–TLR3–IFN pathway.95,96 In contrast to TLR2, 3, and 4, the TLR family member TLR7 co-localized with anti-inflammatory M2 macrophages in stenotic human valves, and TLR7 ligand administration drove the secretion of anti-inflammatory cytokines.97 Taken together, aortic valve TLRs may represent a starting point of aortic valve inflammation triggered by cellular damage and ECM components, and are coupled to immunomodulatory signalling.

Table 1

Genetic associations with calcific aortic valve stenosis

ChromosomeSNPLociNameFunction related to CAVD
6q25.3-q26rs1045587245,47–51,91
rs14183470951
rs14057088650,51
rs11803927851
LPALipoprotein(a)Carrier of oxidized phospholipids and the lysophospholipase autotaxin32
11q12–11q13.1rs17454791
rs17453349,50
rs17455151
FADS1/2
MYRF/FADS
Fatty acid desaturaseEnzyme metabolizing fatty acids92
1p13.3rs152238749
rs1274037450
CELSR2-SORT1 gene clusterSortilin 1Type I membrane glycoprotein with protein-sorting functions.86
1p21.2rs754313047,49
rs754303947
rs670261948,50,51
PALMDPalmdelphinParalemmin protein regulating endothelial actin cap formation and nuclear alignment.41
7p15.3rs206983248
rs180079548
rs147434749
rs180079750,51
rs209767851
IL6Interleukin 6Pro-inflammatory cytokine
1p36.12rs1214156948
rs754712848
rs375378249
rs669606650
ALPLAlkaline phosphataseHydrolysis of organic phosphate and pyrophosphate93
2q22.3rs183032147,48
rs224636349
rs759333647,50
rs7285446251
TEX41Testis expressed 41Long non-protein-coding RNA (lncRNA)
1q32.1rs753598948
rs66577048
rs66583448
rs63155650
rs68211251
NAV1Neuron Navigator 1Unknown
ChromosomeSNPLociNameFunction related to CAVD
6q25.3-q26rs1045587245,47–51,91
rs14183470951
rs14057088650,51
rs11803927851
LPALipoprotein(a)Carrier of oxidized phospholipids and the lysophospholipase autotaxin32
11q12–11q13.1rs17454791
rs17453349,50
rs17455151
FADS1/2
MYRF/FADS
Fatty acid desaturaseEnzyme metabolizing fatty acids92
1p13.3rs152238749
rs1274037450
CELSR2-SORT1 gene clusterSortilin 1Type I membrane glycoprotein with protein-sorting functions.86
1p21.2rs754313047,49
rs754303947
rs670261948,50,51
PALMDPalmdelphinParalemmin protein regulating endothelial actin cap formation and nuclear alignment.41
7p15.3rs206983248
rs180079548
rs147434749
rs180079750,51
rs209767851
IL6Interleukin 6Pro-inflammatory cytokine
1p36.12rs1214156948
rs754712848
rs375378249
rs669606650
ALPLAlkaline phosphataseHydrolysis of organic phosphate and pyrophosphate93
2q22.3rs183032147,48
rs224636349
rs759333647,50
rs7285446251
TEX41Testis expressed 41Long non-protein-coding RNA (lncRNA)
1q32.1rs753598948
rs66577048
rs66583448
rs63155650
rs68211251
NAV1Neuron Navigator 1Unknown
Table 1

Genetic associations with calcific aortic valve stenosis

ChromosomeSNPLociNameFunction related to CAVD
6q25.3-q26rs1045587245,47–51,91
rs14183470951
rs14057088650,51
rs11803927851
LPALipoprotein(a)Carrier of oxidized phospholipids and the lysophospholipase autotaxin32
11q12–11q13.1rs17454791
rs17453349,50
rs17455151
FADS1/2
MYRF/FADS
Fatty acid desaturaseEnzyme metabolizing fatty acids92
1p13.3rs152238749
rs1274037450
CELSR2-SORT1 gene clusterSortilin 1Type I membrane glycoprotein with protein-sorting functions.86
1p21.2rs754313047,49
rs754303947
rs670261948,50,51
PALMDPalmdelphinParalemmin protein regulating endothelial actin cap formation and nuclear alignment.41
7p15.3rs206983248
rs180079548
rs147434749
rs180079750,51
rs209767851
IL6Interleukin 6Pro-inflammatory cytokine
1p36.12rs1214156948
rs754712848
rs375378249
rs669606650
ALPLAlkaline phosphataseHydrolysis of organic phosphate and pyrophosphate93
2q22.3rs183032147,48
rs224636349
rs759333647,50
rs7285446251
TEX41Testis expressed 41Long non-protein-coding RNA (lncRNA)
1q32.1rs753598948
rs66577048
rs66583448
rs63155650
rs68211251
NAV1Neuron Navigator 1Unknown
ChromosomeSNPLociNameFunction related to CAVD
6q25.3-q26rs1045587245,47–51,91
rs14183470951
rs14057088650,51
rs11803927851
LPALipoprotein(a)Carrier of oxidized phospholipids and the lysophospholipase autotaxin32
11q12–11q13.1rs17454791
rs17453349,50
rs17455151
FADS1/2
MYRF/FADS
Fatty acid desaturaseEnzyme metabolizing fatty acids92
1p13.3rs152238749
rs1274037450
CELSR2-SORT1 gene clusterSortilin 1Type I membrane glycoprotein with protein-sorting functions.86
1p21.2rs754313047,49
rs754303947
rs670261948,50,51
PALMDPalmdelphinParalemmin protein regulating endothelial actin cap formation and nuclear alignment.41
7p15.3rs206983248
rs180079548
rs147434749
rs180079750,51
rs209767851
IL6Interleukin 6Pro-inflammatory cytokine
1p36.12rs1214156948
rs754712848
rs375378249
rs669606650
ALPLAlkaline phosphataseHydrolysis of organic phosphate and pyrophosphate93
2q22.3rs183032147,48
rs224636349
rs759333647,50
rs7285446251
TEX41Testis expressed 41Long non-protein-coding RNA (lncRNA)
1q32.1rs753598948
rs66577048
rs66583448
rs63155650
rs68211251
NAV1Neuron Navigator 1Unknown

Another genetic indication for the importance of immune activation and suppression in CAVD is represented by SNPs in the fatty acid desaturase FADS1/2 loci which were associated with plasma phospholipid fatty acid levels and, specifically, AS incidence.98 Indeed, transcriptomics and lipidomics of stenotic human valves revealed that the FADS genotype associates with fatty acid desaturase (FADS) mRNA expression, tissue fatty acid content/metabolism, and leaflet calcification.92 Other work has further reinforced genetic associations between AS, FADS1, and pro-inflammatory omega-6 polyunsaturated fatty acids (n-6 PUFAs).91 Lipid-driven regulation of valvular inflammation is also fundamentally important to CAVD initiation and progression. Classical pro-inflammatory mediators of inflammation from the 5-lipoxygenase and cyclooxygenase enzymatic pathways have been associated with AS severity.99 Furthermore, there is lipidomics-based evidence that the inflammation-resolving, omega-3 polyunsaturated fatty acid (n-3 PUFA)-derived lipid mediator resolvin E1 is dysregulated in calcified AV areas.100 Mice with endogenous synthesis of n-3 PUFA presented with reduced calcification, improved valvular haemodynamics, and elevated M2 macrophage burden in a manner dependent on the resolvin E1 receptor ChemR23. These findings suggest that as in the vasculature,101 valvular calcification may result from improper resolution of inflammatory processes.

Bioactive extracellular vesicle cargoes

EVs are nanometer-sized membrane-bound particles actively released by all eukaryotic cell types and loaded with bioactive cargoes including non-coding RNA, proteins, lipids, and metabolites.102 They are well known to play key roles in cell–cell communication,103 and miRNA transcriptomics of circulating EV cargoes has identified that shuttling of the proapoptotic EV-incorporated miR-122-5p into cardiomyocytes is a driver of reduced expression of the antiapoptotic gene BCL2 and responsible for reduced cardiac function after TAVR.104 In cardiovascular tissues, EVs act as the biophysical building blocks of microcalcification,105 and osteogenic culture conditions induce alterations to EV proteomic cargoes, extracellular EV aggregation, and formation of microcalcifications in the valve and vasculature.29,85,106,107 Notably, proteomics of VIC and vascular smooth muscle cell (SMC)-derived EVs from calcifying culture conditions found elevated abundances of the tethering protein ANXA1 (annexin A1).29 ANXA1 was localized to collagen–EV contacts in calcified human cardiovascular tissues, while loss-of-function experiments rescued VIC and SMC calcification in vitro, and abrogated EV-driven microcalcification in 3D collagen hydrogels. Trailblazing efforts to quantitatively and systematically examine the cargoes of EVs entrapped in calcifying vascular and valvular tissue were motivated by the first comparative global proteomic study of disease progression in human atherosclerotic plaques and calcified aortic valves,40 which identified unique protein drivers of CAVD and atherosclerosis and implicated EVs in both conditions. Enrichment of EVs directly from fibrocalcific cardiovascular tissues enabled vesiculomics assessments of tissue-entrapped EV protein and miR cargoes, and unbiased network medicine approaches integrated and prioritized 6 vesicle-borne targets for in vitro validation: FGFR2, PPP2CA, ADAM17, APC, APP, and WNT5A. Strikingly, knockdown of all six targets significantly modulated VIC and SMC calcification in vitro, including cell type-specific effects of FGFR2 (a pro-osteogenic fibroblast growth factor receptor) and WNT5A (control of osteogenesis via noncanonical PKC/JNK signalling and β-catenin antagonization), which mark them as high-priority candidates for future translational efforts aimed at targeted and specific treatment of vascular vs. valvular calcification.40 Vesiculomics techniques have also been applied to tissue-engineered, biomechanically relevant, multi-layered, 3D-bioprinted, VIC-laden hydrogel valve organoids as a means of benchmarking model performance against native human tissues. Notably, network analyses of organoid-derived VIC and secreted EV proteomics determined that organoid culture mimicked 94% of the valve tissue proteome, while 2D monolayer culture only mirrored 70%.108,109

Target discovery at the single-cell level

Early single-cell omics studies of CAVD were focused on transcriptomics atlas development in mouse and human tissues, and identified the presence of substantial cellular heterogeneity in both VIC/VEC and immune/inflammatory cell types.110,111 Further studies have dissected and compared basal differences in cellular landscapes across all four cardiac valves112 When applied to hyperlipidemic mouse models of CAVD, scRNA-seq was suggestive of lipid-driven recruitment of pro-inflammatory macrophages to the valve and protective activation of peroxisome proliferator-activated receptor-gamma (PPARγ) signalling in VECs, and further reinforced the notion that (non-)resident immune cells contribute to CAVD progression.113 Beyond atlas development, scRNA-seq has begun to support CAVD drug target identification: study of cells enzymatically isolated from young non-diseased, aged non-diseased, and CAVD human valves found the proteoglycan Lumican (LUM) marked a pathological pro-osteogenic VIC subpopulation. Multi-omics assessments revealed that LUM drove lactylation of histone H3 and activation of prototypical osteogenic transcriptional programmes.114 Studies of cultured VICs or VECs have been utilized to successfully identify key pathogenic subpopulations: scRNA-seq has identified sortilin-driven differentiation of an inflammatory-myofibroblastic-osteogenic VIC subtype in response to calcifying media conditions,86 and single-cell multi-omics (scRNA-seq and single-cell mass cytometry) of VIC cultures isolated a disease-driving CD44highCD29+CD59+CD73+CD45low population with exacerbated osteogenic potential. Temporal bulk proteomics of this disease-driving VIC subpopulation and loss-of-function experiments identified monoamine oxidase (MAOA) as a potential therapeutic target.24 As we noted above, the AV has a particularly elaborate anatomy, and spatial segregation of disease may hold the key to effective therapeutics. Early efforts at spatial omics in CAVD employed laser-capture microdissection of tissue sections and prototypical downstream bulk transcriptomics to examine layer-specific gene expression in diseased human valves.39 Subsequent studies have leveraged 10× Genomics slide-based molecular profiling techniques to examine embryonic development of the murine AV and associate defective retinol-binding protein 1 (RBP1) signalling with BAV formation, though the application of high-resolution spatial omics approaches to CAVD remain to be performed.115

Clinical translational towards pharmacotherapy for aortic stenosis

Lipoprotein-lowering strategies

AS pharmacotherapeutics represent a global and rapidly growing potential market with the opportunity to cover the gap in care caused by inequalities in access to TAVR/SAVR throughout much of the world. Unfortunately, the cost and challenging nature of cardiovascular outcomes trials in general, and for AS in particular, has limited industry interest in this area to the point where any successful therapy would enjoy a first-to-market advantage. Despite a total lack of success to date (reviewed in Kraler et al.21 and Lindman et al.116), there are a number of ongoing clinical trials aimed at testing potential medical therapies for AS (Table 2). Thus far, these programmes have focused on attempts to repurpose a wide variety of drugs initially approved or developed for other indications; few if any first-in-class drugs have been developed against AS (Figure 3). Despite many shared risk factors and certain gross histological similarities, anti-atherosclerosis drugs (e.g. statins) which successfully reduce pro-inflammatory lipid accumulation have thus far failed to provide any protective effect against CAVD progression. Though low-density lipoprotein-cholesterol (LDL-C) is a strong and well-described clinical risk factor of AS,9 statin-mediated LDL-C lowering has continually failed in a number of prospective trials (e.g. SEAS,117 ASTRONOMER,118 SALTIRE119). While post-hoc analyses have evaluated the influence of baseline AS disease severity and LDL-C levels,120,121 the window in which LDL-C lowering by statins may have therapeutic benefits remains to be established. Another contributor to such therapeutic benefits against AS may related to the degree of LDL-C lowering, which can be obtained to a greater extent by inhibitors of the LDL receptor (LDL-R)-degrading proprotein convertase subtilisin/kexin type 9 (PCSK9). In the FOURIER trial, which was one of the studies showing additional beneficial effects of PCSK9 inhibition in addition to statin therapy in atherosclerotic cardiovascular disease prevention, an exploratory analysis pointed to potential protective effects of the PCSK9 inhibitor Evolocumab on AS measures when compared with placebo beyond 1 year of treatment.122 The EPISODE trial is evaluating three such PCSK9 inhibitors (Evolocumab, Alirocumab, and Tafolecimab) in patients with mild, moderate, or asymptomatic severe AS.123,124 In addition to systemic LDL-C lowering, PCSK9 inhibitors may directly affect local valve inflammation and calcification processes, as demonstrated by the presence of PCSK9 secretion by VICs and in vitro reductions in VIC calcification upon PCSK9 inhibition (Figure 3).125

Disease mechanisms, drug targets, and pharmaceutical therapies for aortic stenosis. A clear causal role for low-density lipoprotein cholesterol in aortic stenosis has driven extensive trials of lipid-lowering therapies including statins, proprotein convertase subtilisin/kexin type 9 inhibitors, and lipoprotein(a) inhibitors. Sodium–glucose cotransporter-2 inhibitors and angiotensin II receptor blockers appear to reduce mortality in aortic stenosis via mitigation of heart failure and normalization of blood pressure, though inhibition of the renin–angiotensin system by angiotensin II receptor blockers or angiotensin-converting enzyme inhibitors may prevent monocyte activation and/or pro-inflammatory macrophage-derived signalling. Other therapeutic efforts have also targeted the inflammatory impact of these cells and signalling pathways on resident valvular interstitial cells and valvular endothelial cells, and include inhibition of dipeptidyl peptidase 4, peroxisome proliferator-activated receptor gamma activation, colchicine, denosumab (receptor activator of nuclear factor kappa beta ligand inhibition), and glucagon-like peptide 1 receptor agonists. Notably, pharmacological modulation dipeptidyl peptidase 4 and glucagon-like peptide 1 may also directly regulate valvular interstitial cell myofibrogenesis and osteogenesis. Drug targets implicated in pathological valvular interstitial cell differentiation also include cadherin 11 (blocked by the monoclonal antibody SYN0012) and soluble guanylate cyclase (activated by Ataciguat). Other therapeutics aim to directly target calcification by impairing hydroxyapatite mineral formation (inositol-based SNF472 and INS-3001) or by stabilizing common constituents of amyloidosis (Tafamidis) that may be contained in extracellular vesicles released from osteogenic valvular interstitial cells. Novel omics-derived pharmaceutical targets that remain to be tested in clinical trials include Sortilin (driver of valvular interstitial cell myofibro/osteogenesis), major facilitator superfamily domain containing 5 [lipoprotein(a) receptor], Apolipoprotein C-III, annexin A1/A5 (extracellular vesicle cargo/tethering), amyloid precursor protein, monoamine oxidase A, and aortic valve calcification-associated PIWI-interacting RNA
Figure 3

Disease mechanisms, drug targets, and pharmaceutical therapies for aortic stenosis. A clear causal role for low-density lipoprotein cholesterol in aortic stenosis has driven extensive trials of lipid-lowering therapies including statins, proprotein convertase subtilisin/kexin type 9 inhibitors, and lipoprotein(a) inhibitors. Sodium–glucose cotransporter-2 inhibitors and angiotensin II receptor blockers appear to reduce mortality in aortic stenosis via mitigation of heart failure and normalization of blood pressure, though inhibition of the renin–angiotensin system by angiotensin II receptor blockers or angiotensin-converting enzyme inhibitors may prevent monocyte activation and/or pro-inflammatory macrophage-derived signalling. Other therapeutic efforts have also targeted the inflammatory impact of these cells and signalling pathways on resident valvular interstitial cells and valvular endothelial cells, and include inhibition of dipeptidyl peptidase 4, peroxisome proliferator-activated receptor gamma activation, colchicine, denosumab (receptor activator of nuclear factor kappa beta ligand inhibition), and glucagon-like peptide 1 receptor agonists. Notably, pharmacological modulation dipeptidyl peptidase 4 and glucagon-like peptide 1 may also directly regulate valvular interstitial cell myofibrogenesis and osteogenesis. Drug targets implicated in pathological valvular interstitial cell differentiation also include cadherin 11 (blocked by the monoclonal antibody SYN0012) and soluble guanylate cyclase (activated by Ataciguat). Other therapeutics aim to directly target calcification by impairing hydroxyapatite mineral formation (inositol-based SNF472 and INS-3001) or by stabilizing common constituents of amyloidosis (Tafamidis) that may be contained in extracellular vesicles released from osteogenic valvular interstitial cells. Novel omics-derived pharmaceutical targets that remain to be tested in clinical trials include Sortilin (driver of valvular interstitial cell myofibro/osteogenesis), major facilitator superfamily domain containing 5 [lipoprotein(a) receptor], Apolipoprotein C-III, annexin A1/A5 (extracellular vesicle cargo/tethering), amyloid precursor protein, monoamine oxidase A, and aortic valve calcification-associated PIWI-interacting RNA

Table 2

Potential medical therapies for calcific aortic valve stenosis

DrugTargetTrial nameTrial IDNotes
Evolocumab, Alirocumab, TafolecimabPCSK9EPISODE, PCSK9-ASNCT04968509, NCT03051360Along with LDL-C lowering, reduces Lp(a) plasma concentrations; inhibits VIC calcification in vitro
PelacarsenLp(a)Lp(a)FRONTIERS CAVSNCT05646381Strong risk gene for CAVS; plasma levels highly genetically determined
NiacinLp(a)EAVaLLNCT02109614Non-specific Lp(a) lowering
ARBs, FimasartanAT1RARBAS, ALFANCT04913870, NCT01589380Beyond blood pressure, AT1R may directly regulate macrophage, cardiomyocyte, and VIC pathogenesis
EvogliptinDPP4EVOID-AS, DIP-CAVDNCT05143177, NCT04055883Modulation of valvular inflammatory signalling, VIC/VEC pathological differentiation in vitro
ColchicineInflammationCHIANTI, COPAS-PilotNCT05162742, NCT05253794Prior studies show significant reductions in cardiovascular events in patients with chronic coronary disease
PioglitazonePPARγWUHFACAVD02NCT05875675Pre-clinical studies show PPARγ agonists reduce valvular inflammation and calcification in mice
Denosumab, AlendronateRANKL, CalcificationSALTIRE IINCT02132026Failure to demonstrate effect on aortic valve calcification activity/burden nor haemodynamics
AtaciguatsGCCAVSNCT02481258Nitric oxide-dependent sGC activator; pre-clinical studies show reduced mouse aortic valve calcification
Vitamin K2MGPBASIK2, AVADEC, DECAV-K2NCT02917525, NCT03243890,NCT03305536Carboxylation and activation of the ECM-embedded calcification inhibitor matrix Gla protein (MGP)
SNF-472HydroxyapatiteCaLIPSONCT02966028Attenuation of coronary artery and valve calcification in end-stage kidney disease
INS-3001CalcificationNL78901.056.21EUCTR2021-001969-20-NLInositol derivative abolishes rodent cardiovascular calcification
GLP1RAsGLP1Liraglutide reduces CAVS in mice; GLP1 prevents VIC calcification in vitro
SGLT2iSGLT2BIO-ASIncreased cardiac SGLT2 abundance in human CAVS; improves heart failure in rat CAVS model
TafamidisTransthyretinReduced mortality in transthyretin amyloid cardiomyopathy; CAVS impact remains unknown
DTPA-NPCalcification/elastinDTPA nanoparticles conjugated to anti-elastin targeting antibodies reversed ex vivo porcine leaflet mineralization
DrugTargetTrial nameTrial IDNotes
Evolocumab, Alirocumab, TafolecimabPCSK9EPISODE, PCSK9-ASNCT04968509, NCT03051360Along with LDL-C lowering, reduces Lp(a) plasma concentrations; inhibits VIC calcification in vitro
PelacarsenLp(a)Lp(a)FRONTIERS CAVSNCT05646381Strong risk gene for CAVS; plasma levels highly genetically determined
NiacinLp(a)EAVaLLNCT02109614Non-specific Lp(a) lowering
ARBs, FimasartanAT1RARBAS, ALFANCT04913870, NCT01589380Beyond blood pressure, AT1R may directly regulate macrophage, cardiomyocyte, and VIC pathogenesis
EvogliptinDPP4EVOID-AS, DIP-CAVDNCT05143177, NCT04055883Modulation of valvular inflammatory signalling, VIC/VEC pathological differentiation in vitro
ColchicineInflammationCHIANTI, COPAS-PilotNCT05162742, NCT05253794Prior studies show significant reductions in cardiovascular events in patients with chronic coronary disease
PioglitazonePPARγWUHFACAVD02NCT05875675Pre-clinical studies show PPARγ agonists reduce valvular inflammation and calcification in mice
Denosumab, AlendronateRANKL, CalcificationSALTIRE IINCT02132026Failure to demonstrate effect on aortic valve calcification activity/burden nor haemodynamics
AtaciguatsGCCAVSNCT02481258Nitric oxide-dependent sGC activator; pre-clinical studies show reduced mouse aortic valve calcification
Vitamin K2MGPBASIK2, AVADEC, DECAV-K2NCT02917525, NCT03243890,NCT03305536Carboxylation and activation of the ECM-embedded calcification inhibitor matrix Gla protein (MGP)
SNF-472HydroxyapatiteCaLIPSONCT02966028Attenuation of coronary artery and valve calcification in end-stage kidney disease
INS-3001CalcificationNL78901.056.21EUCTR2021-001969-20-NLInositol derivative abolishes rodent cardiovascular calcification
GLP1RAsGLP1Liraglutide reduces CAVS in mice; GLP1 prevents VIC calcification in vitro
SGLT2iSGLT2BIO-ASIncreased cardiac SGLT2 abundance in human CAVS; improves heart failure in rat CAVS model
TafamidisTransthyretinReduced mortality in transthyretin amyloid cardiomyopathy; CAVS impact remains unknown
DTPA-NPCalcification/elastinDTPA nanoparticles conjugated to anti-elastin targeting antibodies reversed ex vivo porcine leaflet mineralization
Table 2

Potential medical therapies for calcific aortic valve stenosis

DrugTargetTrial nameTrial IDNotes
Evolocumab, Alirocumab, TafolecimabPCSK9EPISODE, PCSK9-ASNCT04968509, NCT03051360Along with LDL-C lowering, reduces Lp(a) plasma concentrations; inhibits VIC calcification in vitro
PelacarsenLp(a)Lp(a)FRONTIERS CAVSNCT05646381Strong risk gene for CAVS; plasma levels highly genetically determined
NiacinLp(a)EAVaLLNCT02109614Non-specific Lp(a) lowering
ARBs, FimasartanAT1RARBAS, ALFANCT04913870, NCT01589380Beyond blood pressure, AT1R may directly regulate macrophage, cardiomyocyte, and VIC pathogenesis
EvogliptinDPP4EVOID-AS, DIP-CAVDNCT05143177, NCT04055883Modulation of valvular inflammatory signalling, VIC/VEC pathological differentiation in vitro
ColchicineInflammationCHIANTI, COPAS-PilotNCT05162742, NCT05253794Prior studies show significant reductions in cardiovascular events in patients with chronic coronary disease
PioglitazonePPARγWUHFACAVD02NCT05875675Pre-clinical studies show PPARγ agonists reduce valvular inflammation and calcification in mice
Denosumab, AlendronateRANKL, CalcificationSALTIRE IINCT02132026Failure to demonstrate effect on aortic valve calcification activity/burden nor haemodynamics
AtaciguatsGCCAVSNCT02481258Nitric oxide-dependent sGC activator; pre-clinical studies show reduced mouse aortic valve calcification
Vitamin K2MGPBASIK2, AVADEC, DECAV-K2NCT02917525, NCT03243890,NCT03305536Carboxylation and activation of the ECM-embedded calcification inhibitor matrix Gla protein (MGP)
SNF-472HydroxyapatiteCaLIPSONCT02966028Attenuation of coronary artery and valve calcification in end-stage kidney disease
INS-3001CalcificationNL78901.056.21EUCTR2021-001969-20-NLInositol derivative abolishes rodent cardiovascular calcification
GLP1RAsGLP1Liraglutide reduces CAVS in mice; GLP1 prevents VIC calcification in vitro
SGLT2iSGLT2BIO-ASIncreased cardiac SGLT2 abundance in human CAVS; improves heart failure in rat CAVS model
TafamidisTransthyretinReduced mortality in transthyretin amyloid cardiomyopathy; CAVS impact remains unknown
DTPA-NPCalcification/elastinDTPA nanoparticles conjugated to anti-elastin targeting antibodies reversed ex vivo porcine leaflet mineralization
DrugTargetTrial nameTrial IDNotes
Evolocumab, Alirocumab, TafolecimabPCSK9EPISODE, PCSK9-ASNCT04968509, NCT03051360Along with LDL-C lowering, reduces Lp(a) plasma concentrations; inhibits VIC calcification in vitro
PelacarsenLp(a)Lp(a)FRONTIERS CAVSNCT05646381Strong risk gene for CAVS; plasma levels highly genetically determined
NiacinLp(a)EAVaLLNCT02109614Non-specific Lp(a) lowering
ARBs, FimasartanAT1RARBAS, ALFANCT04913870, NCT01589380Beyond blood pressure, AT1R may directly regulate macrophage, cardiomyocyte, and VIC pathogenesis
EvogliptinDPP4EVOID-AS, DIP-CAVDNCT05143177, NCT04055883Modulation of valvular inflammatory signalling, VIC/VEC pathological differentiation in vitro
ColchicineInflammationCHIANTI, COPAS-PilotNCT05162742, NCT05253794Prior studies show significant reductions in cardiovascular events in patients with chronic coronary disease
PioglitazonePPARγWUHFACAVD02NCT05875675Pre-clinical studies show PPARγ agonists reduce valvular inflammation and calcification in mice
Denosumab, AlendronateRANKL, CalcificationSALTIRE IINCT02132026Failure to demonstrate effect on aortic valve calcification activity/burden nor haemodynamics
AtaciguatsGCCAVSNCT02481258Nitric oxide-dependent sGC activator; pre-clinical studies show reduced mouse aortic valve calcification
Vitamin K2MGPBASIK2, AVADEC, DECAV-K2NCT02917525, NCT03243890,NCT03305536Carboxylation and activation of the ECM-embedded calcification inhibitor matrix Gla protein (MGP)
SNF-472HydroxyapatiteCaLIPSONCT02966028Attenuation of coronary artery and valve calcification in end-stage kidney disease
INS-3001CalcificationNL78901.056.21EUCTR2021-001969-20-NLInositol derivative abolishes rodent cardiovascular calcification
GLP1RAsGLP1Liraglutide reduces CAVS in mice; GLP1 prevents VIC calcification in vitro
SGLT2iSGLT2BIO-ASIncreased cardiac SGLT2 abundance in human CAVS; improves heart failure in rat CAVS model
TafamidisTransthyretinReduced mortality in transthyretin amyloid cardiomyopathy; CAVS impact remains unknown
DTPA-NPCalcification/elastinDTPA nanoparticles conjugated to anti-elastin targeting antibodies reversed ex vivo porcine leaflet mineralization

Other lipoproteins, particularly Lp(a), confer significant and substantial AS risk.9 Over a decade ago, a pioneering GWAS and MR study implicated the first genetic drivers of AS and demonstrated that a SNP within the LPA locus drove genetically determined alterations in circulating Lp(a) levels that were significantly and causally associated with AS (Table 1).45 In the ∼7% of AS cases accounted for by Lp(a), these effects appear to be mediated by the oxidized phospholipids and the ectonucleotide pyrophosphatase/phosphodiesterase autotaxin carried by Lp(a), which directly drive VIC osteogenesis.32,33 Until recently, Lp(a) was challenging to target therapeutically—statins do not lower Lp(a) levels, and niacin lowers circulating concentrations by ∼20%–30% alongside an increased incidence of adverse events. As a result, recruitment to the EAVaLL trial was slow and eventually withdrawn.126 Because LDL-R internalizes both LDL and Lp(a), PCSK9 inhibitors reduce circulating levels of both lipoproteins—though MR studies suggest the expected Lp(a)-lowering capability of existing PCSK9 inhibitors (similar to that of niacin) may be insufficient for hope of a therapeutic benefit.127 In a more specific approach, the Lp(a)FRONTIERS CAVS trial is currently recruiting individuals with mild or moderate AS to test the efficacy of Lp(a) lowering using the anti-LPA antisense oligonucleotide Pelacarsen.128 Outside of LDL-R, a specific valvular receptor responsible for internalization of Lp(a) remained undiscovered until Rogers and colleagues employed unbiased ligand–receptor capture mass spectrometry to identify major facilitator superfamily domain containing 5 (MFSD5) as a putative Lp(a) receptor/cofactor.129In vitro, chemical or genetic reduction of MFSD5 expression impaired Lp(a) uptake and suppressed calcification in both VICs and VECs, and targeted assessment of genetic MFSD5 variants was significantly associated with AS. Together, these data suggest MFSD5 as a high-priority drug target for inhibition of Lp(a)-specific internalization. Beyond Lp(a), targeted proteomics of apolipoproteins has unearthed enrichment of Apolipoprotein C-III (apo-CIII) in calcified valve regions, and shown that apo-CIII induced calcification in primary human VIC cultures via a mitochondrial dysfunction/inflammation-mediated pathway.30 These findings raise the intriguing notion of a potentially novel class of repurposed AS treatments through direct therapeutic reduction of apo-CIII (e.g. via antisense approaches such as Olezarsen130), or by targeting apo-CIII-modulated hypertriglyceridemia through fibrates or icosapent ethyl.131,132

Therapeutic disruption of pathogenic valvular cell mechanisms

Indeed, due to their active roles in driving disease most remaining drug development endeavours have focused on directly modulating pathogenic disease drivers in VICs, VECs, macrophages, and other valve-resident cell types. After notable reductions of cardiovascular events in patients with chronic CAD,133 interest grew in repurposing the anti-inflammatory alkaloid colchicine towards other cardiovascular indications. The CHIANTI and COPAS-Pilot trials are testing the impact of low-dose colchicine therapy on mild to moderate AS progression.134,135 In pre-clinical studies, scRNA-seq studies found pro-inflammatory valvular macrophages were increased in hyperlipidemic murine models of AS along with activation of PPARγ signalling in VECs.113 Notably, PPARγ agonists reduced valvular inflammation, monocyte-derived macrophage content, and calcification in mice and prospective trials of pioglitazone in mild-to-moderate AS are underway.136,137 Transcriptomics has also revealed that oxidative stress and resultant endothelial dysfunction appear to drive elevated VEC expression of the exopeptidase dipeptidyl peptidase-4 (DPP4), which contributes to recruitment of inflammatory macrophages and inhibits homeostatic insulin-like growth factor 1 (IGF-1) signalling in VICs, driving osteogenesis and calcification.138 Inhibition of DPP4 by Sitagliptin in a rabbit AS model drove significant valvular calcium and haemodynamic improvements; the EVOID-AS and DIP-CAVD trials are testing the DPP4 inhibitor Evogliptin in AS outcomes.139,140 Though DIP-CAVD recently reported negative outcomes on aortic valve calcification burden, Evogliptin drove significant improvement in some secondary endpoints (early reductions in calcification as measured by more sensitive radiotracer-based imaging and suppression of increases in calcification burden at later timepoints).141 Many DPP4 inhibitors also prevent catabolism of incretins [e.g. glucagon-like peptide 1 (GLP1)]. In obese individuals without diabetes, the incretin-mimic semaglutide improved cardiovascular outcomes in a manner that may be at least partially independent of body weight reductions and suggested direct physiological effects.142 GLP1 levels are reduced in human AS blood and valvular tissue, and antagonized pro-osteogenic signalling pathways and VIC calcification in vitro.143 In hyperlipidemic mice, liraglutide administration attenuated AV calcification, and leaflet transcriptomics implicated reduced VIC myofibro/osteogenic differentiation and lower inflammatory macrophage accumulation in incretin mimetic-treated mice, suggestive of potential therapeutic benefits in human AS.144 Downstream of endothelial dysfunction and nitric oxide145 or natriuretic peptide signalling, valvular cyclic guanosine monophosphate levels contribute to embryonic leaflet development along with adult VIC/VEC mechanosensation and homeostasis.55,146 The AS trial is current testing the nitric oxide-independent soluble guanylate cyclase (sGC) activator Ataciguat in patients with moderate AS; early readouts in mice and humans were suggestive of positive effects on VIC osteogenesis and progression of calcification.147,148

Extracellular targets

Other efforts have aimed at directly targeting extracellular or ECM-associated targets, with a particular focus on mineral metabolism. After some retrospective evidence that extra-skeletal effects of anti-osteoporosis drugs might slow AV calcification progression, the SALTIRE II trial randomized patients with mild or moderate AS to the RANKL inhibitor denosumab or the bisphosphonate alendronate but failed to alter AV calcium burden nor haemodynamic measures.149,150 Other modes appear more promising: SNF-472 is the hexasodium salt of myo-inositol hexaphosphate (phytate) that inhibits ectopic calcification by selectively binding to the surface of hydroxyapatite crystals, thereby blocking ion binding and further crystal growth. Pre-clinical studies found that SNF-472 inhibited VIC calcification in vitro, abrogated calcium accumulation in an ex vivo porcine AV leaflet culture model, and reduced cardiovascular calcification levels in uraemic rodents.151–153 In patients with end-stage renal disease and on haemodialysis, intravenous delivery of SNF-472 in the CaLIPSO trial significantly slowed imaging-measured progression of both coronary artery and AV calcification.154,155 The closely related compound INS-3001 is an inositol phosphate derivatized with ethylene glycol oligomers, which stabilizes calciprotein particle growth and offers improved pharmacokinetic profiles. INS-3001 inhibited calcification of cultured SMCs, explanted human femoral arteries, and AVs, and reduced vascular calcification in vitamin D-induced murine models without changes in trabecular bone area.156 First-in-human studies of INS-3001 in patients with moderate AS are underway.157 Other direct protein modulators of mineralization include the master calcification inhibitor matrix Gla protein (MGP), a vitamin K-dependent calcium-binding protein that negatively regulates cultured VIC calcification,158 while circulating levels are reduced in AS patients.159 In a trio of larger ongoing trials (BASIK2, AVADEC, DECAV-K2), vitamin K2 supplementation aimed at driving carboxylation and activation of MGP is actively being assessed in patients with both prototypical tricuspid and congenital bicuspid AS.160–162 ECM components have also been directly targeted: covalent binding of the chelator diethylenetriaminepentaacetic acid onto albumin-based nanoparticles conjugated to anti-elastin targeting antibodies arrested and reversed ex vivo mineralization of intact porcine AV leaflets.163 In the valve, misfolded extracellular protein aggregates (amyloids) may act as a strong pro-calcifying substrate. Cardiomyopathic transthyretin amyloidosis (ATTR-CM) is an under-recognized form of heart failure that is often comorbid with AS. Multi-omics has shown amyloid proteins are highly and specifically elevated in human CAVD tissues, carried by valvular tissue-entrapped EVs,40,164 and amyloid plaque co-localizes with valvular nodule formation.165 Recent successes in small molecule (e.g. Tafamidis), RNAi, CRISPR–Cas9, and antibody-based therapies for ATTR-CM are promising future areas for therapeutic discovery and drug repurposing.166

Myocardial mediators of heart failure

In recent years, therapeutic inhibition of sodium–glucose cotransporter 2 (SGLT2) has expanded beyond the treatment of Type 2 diabetes after unexpected reductions in heart failure hospitalization rates. Follow-up studies found broad effects on cellular metabolic processes, inflammation, and fibrosis, though the exact mechanisms of these beneficial effects remain unclear. Regardless, cardiac SGLT2 abundances are elevated in low-flow, low-gradient AS, and in preliminary studies, the SGLT2 inhibitor Empagliflozin improved heart failure phenotypes in supra-aortic-banded rats.167 While inhibition of angiotensin-converting enzyme (ACE) has shown variable and conflicting effects on AS outcomes in a number of trials, retrospective assessments of anti-hypertensive blockade of the renin–angiotensin system in AS via angiotensin II receptor blockers (ARBs) suggests this drug class may leverage its cardioprotective benefits against AS class with a modicum of success.168 The ongoing ARBAS and ALFA trials will assess the efficacy of angiotensin II receptor type 1 (AT1R) inhibition on AS progression, with a focus on valvular haemodynamic parameters and cardiac function.169,170 Notably, outside of impacts on the heart, AT1R is expressed in VICs21,171 with elevated levels found in stenotic AVs172 and may also directly regulate pathological valvular macrophage and VIC phenotypes (Figure 3).173,174 Interestingly, such an effect may be sex-specific and in alignment with the female fibrotic AS phenotype:73 a recent retrospective assessment found that ARB intake was only associated with reduced valvular leaflet fibrosis in female patients, not males.175

Conclusions

As we enter into the age of testing the first pharmaceutical products designed against early genomics-derived anti-AS drug targets [e.g. Lp(a)45], so too is the field now positioned to expand this set of risk genes with large-cohort genomics meta-analyses and MR approaches.49–51 The fusion of large-cohort databases with robust clinical and molecular datasets, or clinomics, promises to improve our understanding and treatment of AS. By integrating deep clinical phenotyping (clinical factors, imaging, events, outcomes, etc.) of large longitudinal cohorts with genomics and unbiased profiling of circulating or tissue molecular profiles (often via affinity- or aptamer-based proteomics methods176,177), MR-based inference may enable precise identification of causal targets, subpopulations with greater potential therapeutic efficacy, anticipation of safety signals, and personalized prediction of AS progression as has been done for CAD.52,178,179 Rapid maturation of both the technical capabilities of multi-omics methods and downstream systems analyses has also begun to spur pre-clinical translation of the first non-genomic targets towards drug development.24,69,86,129 Single-cell technologies have recently expanded beyond the transcriptome and into epigenomics, immune repertoire profiling, and cell surface protein quantitation; these modalities promise to facilitate the specific targeting of crucial cellular subpopulations that contribute to CAVD pathogenesis.110,112–114 Single-cell techniques may also enable differentiation of BAV vs. TAV-associated disease-driving mechanisms, while cusp-specific studies could shed light on how shear-stress or flow-related modulate cellular subpopulations in the valve. More broadly, single-cell studies across various stages of disease (e.g. early initiation, mid-stage fibrosis, end-stage calcification) will help to better understand the complex continuum of pathobiology present in CAVD. In turn, spatial transcriptomics may unravel the complex anatomical and layer-specific cellular regulatory programmes that underpin the disease-prone nature of the fibrosa. Lastly, proteomics techniques have now advanced to the point where circulating biomarkers of AS, AS risk, and AS progression can now be reliably and economically assayed in large cohorts.176,177

These important advances are also tinged by a growing clarity and comprehension of the pitfalls that will be faced along the path towards efficacious AS pharmacotherapies. As easily detected targets with the largest effect sizes are discovered, larger cohorts and costlier/deeper sequencing and phenotyping will be needed to identify significant hits and threaten to provide ever-diminishing returns. Multi-omics promises to ease these concerns by leveraging cross-modality interactions to identify more subtle hits in existing sample sets. The cost, complexity, and length of rigorous clinical trials in AS will also remain a major impediment to translation. Cost-efficient trials will require advances in early detection and diagnosis, improved prediction models of sclerosis-to-stenosis progression, careful selection of trial enrolment populations, and non-traditional clinical endpoints (reviewed elsewhere21,116). Development of targeted therapeutic strategies may be assisted by trial stratification and potentiated via a clearer differentiation of sex-based differences in AS pathogenesis,73–75,79–81,180 of disease-initiating and disease-driving factors in CKD- and non-CKD-associated CAVD,181 between pathogenic mechanisms and markers of CAVD vs. CAD,40,53,182 or via insights into disease stages and vulnerable phases of CAVD development potentially associated with intra-leaflet haemorrhage.183–185 Systematic multi-omics assessments of concordant and paradoxical mechanisms between valvular calcification and skeletal bone formation remain unstudied, and could shed new light on AS pathogenesis and aid in drug repurposing strategies.186,187

In one under-appreciated way, the field will soon be a victim of its own success: in the TAVR era, SAVR volumes have dropped precipitously—some estimates place this reduction at almost 45% over the past decade.188 The resultant scarcity of SAVR-derived diseased human tissue samples is becoming an impediment to basic science efforts and has prompted renewed attempts to develop new animal models (reviewed in Sider et al.37 and Zhu et al.189) or engineer organoids and other suitable 3D models of CAVD.108,109,190–192 Appropriate model selection, where mode(s) of model AS induction match putative target-driven disease mechanisms, is a critical aspect of both the accurate study of pathogenic mechanisms and rigorous pre-clinical target validation.38,193 Robust and multi-factorial benchmarking of model recapitulation of native human CAVD pathobiology via multi-omics will be of fundamental importance to these initiatives.109 Should these surgical trends continue, there may be a narrow window of time within which the field must work to mitigate the spectre of bioprosthetic valve degeneration and gather the resources needed to support successful pharmaceutical translation in the future.

Supplementary data

Supplementary data are not available at European Heart Journal online.

Declarations

Disclosure of Interest

M.C.B. is a consultant for BioMarin Pharmaceuticals, Inc. M.B.’s institution has received speaker and consultant fees from Amarin, Amgen, Heel, Novartis, and Fresenius Kabi. T.F.L. has received educational and/or research grants from Abbot, Amgen, AstraZeneca, BAYER Health Care, Boehringer Ingelheim, Daichi-Sankyo, Eli Lilly, Novartis, Novo Nordisk, Sanofi, and Vifor, and honoraria or consulting fees from Milestone Pharmaceuticals and Novo Nordisk. E.A. is a member of the scientific board of Elastrin Therapeutics, Inc.

Data Availability

No data were generated or analysed for this manuscript.

Funding

Swedish Research Council (grant number 2023-02652) to M.B., Swedish Heart and Lung Foundation (grant number 20210560) to M.B.; and National Institutes of Health grant number R01HL141917 to E.A.

References

1

Nkomo
VT
,
Gardin
JM
,
Skelton
TN
,
Gottdiener
JS
,
Scott
CG
,
Enriquez-Sarano
M
.
Burden of valvular heart diseases: a population-based study
.
Lancet
2006
;
368
:
1005
11
.

2

Zilla
P
,
Brink
J
,
Human
P
,
Bezuidenhout
D
.
Prosthetic heart valves: catering for the few
.
Biomaterials
2008
;
29
:
385
406
.

3

Moore
M
,
Chen
J
,
Mallow
PJ
,
Rizzo
JA
.
The direct health-care burden of valvular heart disease: evidence from US national survey data
.
Clinicoecon Outcomes Res
2016
;
8
:
613
27
.

4

Martinsson
A
,
Nielsen
SJ
,
Milojevic
M
,
Redfors
B
,
Omerovic
E
,
Tønnessen
T
, et al.
Life expectancy after surgical aortic valve replacement
.
J Am Coll Cardiol
2021
;
78
:
2147
57
.

5

Côté
N
,
Pibarot
P
,
Clavel
MA
.
Incidence, risk factors, clinical impact, and management of bioprosthesis structural valve degeneration
.
Curr Opin Cardiol
2017
;
32
:
123
9
.

6

Yerasi
C
,
Rogers
T
,
Forrestal
BJ
,
Case
BC
,
Khan
JM
,
Ben-Dor
I
, et al.
Transcatheter versus surgical aortic valve replacement in young, low-risk patients with severe aortic stenosis
.
JACC Cardiovasc Interv
2021
;
14
:
1169
80
.

7

Russo
G
,
Tang
GHL
,
Sangiorgi
G
,
Pedicino
D
,
Enriquez-Sarano
M
,
Maisano
F
, et al.
Lifetime management of aortic stenosis: transcatheter versus surgical treatment for young and low-risk patients
.
Circ Cardiovasc Interv
2022
;
15
:
915
27
.

8

Clark
MA
,
Arnold
SV
,
Duhay
FG
,
Thompson
AK
,
Keyes
MJ
,
Svensson
LG
, et al.
Five-year clinical and economic outcomes among patients with medically managed severe aortic stenosis: results from a Medicare claims analysis
.
Circ Cardiovasc Qual Outcomes
2012
;
5
:
697
704
.

9

Stewart
BF
,
Siscovick
D
,
Lind
BK
,
Gardin
JM
,
Gottdiener
JS
,
Smith
VE
, et al.
Clinical factors associated with calcific aortic valve disease. Cardiovascular Health Study
.
J Am Coll Cardiol
1997
;
29
:
630
4
.

10

Danielsen
R
,
Aspelund
T
,
Harris
TB
,
Gudnason
V
.
The prevalence of aortic stenosis in the elderly in Iceland and predictions for the coming decades: the AGES-Reykjavik study
.
Int J Cardiol
2014
;
176
:
916
22
.

11

Santangelo
G
,
Bursi
F
,
Faggiano
A
,
Moscardelli
S
,
Simeoli
PS
,
Guazzi
M
, et al.
The global burden of valvular heart disease: from clinical epidemiology to management
.
J Clin Med
2023
;
12
:
2178
.

12

Yu
J
,
Wang
Z
,
Bao
Q
,
Lei
S
,
You
Y
,
Yin
Z
, et al.
Global burden of calcific aortic valve disease and attributable risk factors from 1990 to 2019
.
Front Cardiovasc Med
2022
;
9
:
1003233
.

13

Vervoort
D
,
Meuris
B
,
Meyns
B
,
Verbrugghe
P
.
Global cardiac surgery: access to cardiac surgical care around the world
.
J Thorac Cardiovasc Surg
2020
;
159
:
987
96.e6
.

14

Vervoort
D
,
Lee
G
,
Lin
Y
,
Contreras Reyes
JR
,
Kanyepi
K
,
Tapaua
N
.
6 billion people have no access to safe, timely, and affordable cardiac surgical care
.
JACC Adv
2022
;
1
:
100061
.

15

Ozkan
J
.
A snapshot of cardiology in Africa
.
Eur Heart J
2018
;
39
:
2128
9
.

16

Crousillat
DR
,
Amponsah
DK
,
Camacho
A
,
Kandanelly
RR
,
Bapat
D
,
Chen
C
, et al.
Racial and ethnic differences in the clinical diagnosis of aortic stenosis
.
J Am Heart Assoc
2022
;
11
:
e025692
.

17

Andell
P
,
Li
X
,
Martinsson
A
,
Nilsson
PM
,
Zöller
B
,
Smith
JG
, et al.
Neighborhood socioeconomic status and aortic stenosis: a Swedish study based on nationwide registries and an echocardiographic screening cohort
.
Int J Cardiol
2020
;
318
:
153
9
.

18

Nathan
AS
,
Yang
L
,
Yang
N
,
Eberly
LA
,
Khatana
SAM
,
Dayoub
EJ
, et al.
Racial, ethnic, and socioeconomic disparities in access to transcatheter aortic valve replacement within major metropolitan areas
.
JAMA Cardiol
2022
;
7
:
150
7
.

19

Lachonius
M
,
Giang
KW
,
Lindgren
M
,
Skoglund
K
,
Pétursson
P
,
Silverborn
M
, et al.
Socioeconomic factors and long-term mortality risk after surgical aortic valve replacement
.
Int J Cardiol Cardiovasc Risk Prev
2023
;
19
:
200223
.

20

Sleder
A
,
Tackett
S
,
Cerasale
M
,
Mittal
C
,
Isseh
I
,
Radjef
R
, et al.
Socioeconomic and racial disparities: a case-control study of patients receiving transcatheter aortic valve replacement for severe aortic stenosis
.
J Racial Ethn Health Disparities
2017
;
4
:
1189
94
.

21

Kraler
S
,
Blaser
MC
,
Aikawa
E
,
Camici
GG
,
Luscher
TF
.
Calcific aortic valve disease: from molecular and cellular mechanisms to medical therapy
.
Eur Heart J
2022
;
43
:
683
97
.

22

Otto
CM
,
Kuusisto
J
,
Reichenbach
DD
,
Gown
AM
,
O'Brien
KD
.
Characterization of the early lesion of ‘degenerative’ valvular aortic stenosis. Histological and immunohistochemical studies
.
Circulation
1994
;
90
:
844
53
.

23

Thubrikar
M
.
The Aortic Valve
.
Boca Raton
:
CRC Press, Inc
,
1990
.

24

Decano
JL
,
Iwamoto
Y
,
Goto
S
,
Lee
JY
,
Matamalas
JT
,
Halu
A
, et al.
A disease-driver population within interstitial cells of human calcific aortic valves identified via single-cell and proteomic profiling
.
Cell Rep
2022
;
39
:
110685
.

25

Hjortnaes
J
,
Shapero
K
,
Goettsch
C
,
Hutcheson
JD
,
Keegan
J
,
Kluin
J
, et al.
Valvular interstitial cells suppress calcification of valvular endothelial cells
.
Atherosclerosis
2015
;
242
:
251
60
.

26

Yabusaki
K
,
Hutcheson
JD
,
Vyas
P
,
Bertazzo
S
,
Body
SC
,
Aikawa
M
, et al.
Quantification of calcified particles in human valve tissue reveals asymmetry of calcific aortic valve disease development
.
Front Cardiovasc Med
2016
;
3
:
44
.

27

Aikawa
E
,
Nahrendorf
M
,
Sosnovik
D
,
Lok
VM
,
Jaffer
FA
,
Aikawa
M
, et al.
Multimodality molecular imaging identifies proteolytic and osteogenic activities in early aortic valve disease
.
Circulation
2007
;
115
:
377
86
.

28

Oh
KS
,
Febres-Aldana
CA
,
Kuritzky
N
,
Ujueta
F
,
Arenas
IA
,
Sriganeshan
V
, et al.
Cellular senescence evaluated by P16INK4a immunohistochemistry is a prevalent phenomenon in advanced calcific aortic valve disease
.
Cardiovasc Pathol
2021
;
52
:
107318
.

29

Rogers
MA
,
Buffolo
F
,
Schlotter
F
,
Atkins
SK
,
Lee
LH
,
Halu
A
, et al.
Annexin A1-dependent tethering promotes extracellular vesicle aggregation revealed with single-extracellular vesicle analysis
.
Sci Adv
2020
;
6
:
eabb1244
.

30

Schlotter
F
,
de Freitas
RCC
,
Rogers
MA
,
Blaser
MC
,
Wu
PJ
,
Higashi
H
, et al.
ApoC-III is a novel inducer of calcification in human aortic valves
.
J Biol Chem
2021
;
296
:
100193
.

31

Blaser
MC
,
Aikawa
E
.
Roles and regulation of extracellular vesicles in cardiovascular mineral metabolism
.
Front Cardiovasc Med
2018
;
5
:
187
.

32

Bouchareb
R
,
Mahmut
A
,
Nsaibia
MJ
,
Boulanger
M-C
,
Dahou
A
,
Lepine
J-L
, et al.
Autotaxin derived from lipoprotein(a) and valve interstitial cells promotes inflammation and mineralization of the aortic valve
.
Circulation
2015
;
132
:
677
90
.

33

Bouchareb
R
,
Boulanger
M-C
,
Tastet
L
,
Mkannez
G
,
Nsaibia
MJ
,
Hadji
F
, et al.
Activated platelets promote an osteogenic programme and the progression of calcific aortic valve stenosis
.
Eur Heart J
2019
;
40
:
1362
73
.

34

Wirrig
EE
,
Hinton
RB
,
Yutzey
KE
.
Differential expression of cartilage and bone-related proteins in pediatric and adult diseased aortic valves
.
J Mol Cell Cardiol
2011
;
50
:
561
9
.

35

Salim
MT
,
Esmerats
JF
,
Arjunon
S
,
Villa-Roel
N
,
Nerem
RM
,
Jo
H
, et al.
miR-214 is stretch-sensitive in aortic valve and inhibits aortic valve calcification
.
Ann Biomed Eng
2019
;
47
:
1106
15
.

36

Heath
JM
,
Fernandez Esmerats
J
,
Khambouneheuang
L
,
Kumar
S
,
Simmons
R
,
Jo
H
.
Mechanosensitive microRNA-181b regulates aortic valve endothelial matrix degradation by targeting TIMP3
.
Cardiovasc Eng Technol
2018
;
9
:
141
50
.

37

Sider
KL
,
Blaser
MC
,
Simmons
CA
.
Animal models of calcific aortic valve disease
.
Int J Inflam
2011
;
2011
:
364310
.

38

Hutcheson
JD
,
Blaser
MC
,
Aikawa
E
.
Giving calcification its due: recognition of a diverse disease: a first attempt to standardize the field
.
Circ Res
2017
;
120
:
270
3
.

39

Schlotter
F
,
Halu
A
,
Goto
S
,
Blaser
MC
,
Body
SC
,
Lee
LH
, et al.
Spatiotemporal multi-omics mapping generates a molecular atlas of the aortic valve and reveals networks driving disease
.
Circulation
2018
;
138
:
377
93
.

40

Blaser
MC
,
Buffolo
F
,
Halu
A
,
Turner
ME
,
Schlotter
F
,
Higashi
H
, et al.
Multiomics of tissue extracellular vesicles identifies unique modulators of atherosclerosis and calcific aortic valve stenosis
.
Circulation
2023
;
148
:
661
78
.

41

Sáinz-Jaspeado
M
,
Smith
RO
,
Plunde
O
,
Pawelzik
SC
,
Jin
Y
,
Nordling
S
, et al.
Palmdelphin regulates nuclear resilience to mechanical stress in the endothelium
.
Circulation
2021
;
144
:
1629
45
.

42

Blaser
MC
,
Kraler
S
,
Lüscher
TF
,
Aikawa
E
.
Multi-omics approaches to define calcific aortic valve disease pathogenesis
.
Circ Res
2021
;
128
:
1371
97
.

43

Blaser
MC
,
Kraler
S
,
Lüscher
TF
,
Aikawa
E
.
Network-guided multiomic mapping of aortic valve calcification
.
Arterioscler Thromb Vasc Biol
2023
;
43
:
417
26
.

44

Aikawa
E
,
Blaser
MC
,
Singh
SA
,
Levine
RA
,
Yacoub
MH
.
Challenges and opportunities in valvular heart disease: from molecular mechanisms to the community
.
Arterioscler Thromb Vasc Biol
2024
;
44
:
763
7
.

45

Thanassoulis
G
,
Campbell
CY
,
Owens
DS
,
Smith
JG
,
Smith
AV
,
Peloso
GM
, et al.
Genetic associations with valvular calcification and aortic stenosis
.
N Engl J Med
2013
;
368
:
503
12
.

46

Thériault
S
,
Gaudreault
N
,
Lamontagne
M
,
Rosa
M
,
Boulanger
MC
,
Messika-Zeitoun
D
, et al.
A transcriptome-wide association study identifies PALMD as a susceptibility gene for calcific aortic valve stenosis
.
Nat Commun
2018
;
9
:
6
13
.

47

Helgadottir
A
,
Thorleifsson
G
,
Gretarsdottir
S
,
Stefansson
OA
,
Tragante
V
,
Thorolfsdottir
RB
, et al.
Genome-wide analysis yields new loci associating with aortic valve stenosis
.
Nat Commun
2018
;
9
:
987
–.

48

Thériault
S
,
Dina
C
,
Messika-Zeitoun
D
,
Le Scouarnec
S
,
Capoulade
R
,
Gaudreault
N
, et al.
Genetic association analyses highlight IL6, ALPL, and NAV1 as 3 new susceptibility genes underlying calcific aortic valve stenosis
.
Circ Genom Precis Med
2019
;
12
:
e002617
.

49

Small
AM
,
Peloso
G
,
Linefsky
J
,
Aragam
J
,
Galloway
A
,
Tanukonda
V
, et al.
Multiancestry genome-wide association study of aortic stenosis identifies multiple novel loci in the million veteran program
.
Circulation
2023
;
147
:
942
55

50

Chen
HY
,
Dina
C
,
Small
AM
,
Shaffer
CM
,
Levinson
RT
,
Helgadóttir
A
, et al.
Dyslipidemia, inflammation, calcification, and adiposity in aortic stenosis: a genome-wide study
.
Eur Heart J
2023
;
44
:
1927
39
.

51

Thériault
S
,
Li
Z
,
Abner
E
,
Luan
J
,
Manikpurage
HD
,
Houessou
U
, et al.
Integrative genomic analyses identify candidate causal genes for calcific aortic valve stenosis involving tissue-specific regulation
.
Nat Commun
2024
;
15
:
2407
.

52

Larsson
SC
,
Bäck
M
,
Rees
JMB
,
Mason
AM
,
Burgess
S
.
Body mass index and body composition in relation to 14 cardiovascular conditions in UK Biobank: a Mendelian randomization study
.
Eur Heart J
2020
;
41
:
221
6
.

53

Small
AM
,
Melloni
GEM
,
Kamanu
FK
,
Bergmark
BA
,
Bonaca
MP
,
O'Donoghue
ML
, et al.
Novel polygenic risk score and established clinical risk factors for risk estimation of aortic stenosis
.
JAMA Cardiol
2024
;
9
:
357
66
.

54

Piacentini
L
,
Myasoedova
VA
,
Chiesa
M
,
Vavassori
C
,
Moschetta
D
,
Valerio
V
, et al.
Whole-blood transcriptome unveils altered immune response in acute myocardial infarction patients with aortic valve sclerosis
.
Arterioscler Thromb Vasc Biol
2024
;
44
:
452
64
.

55

Blaser
MC
,
Wei
K
,
Adams
RL
,
Zhou
YQ
,
Caruso
LL
,
Mirzaei
Z
, et al.
Deficiency of natriuretic peptide receptor 2 promotes bicuspid aortic valves, aortic valve disease, left ventricular dysfunction, and ascending aortic dilatations in mice
.
Circ Res
2018
;
122
:
405
16
.

56

Sun
L
,
Chandra
S
,
Sucosky
P
.
Ex vivo evidence for the contribution of hemodynamic shear stress abnormalities to the early pathogenesis of calcific bicuspid aortic valve disease
.
PLoS One
2012
;
7
:
e48843
.

57

Sabet
HY
,
Edwards
WD
,
Tazelaar
HD
,
Daly
RC
.
Congenitally bicuspid aortic valves: a surgical pathology study of 542 cases (1991 through 1996) and a literature review of 2,715 additional cases
.
Mayo Clin Proc
1999
;
74
:
14
26
.

58

Gehlen
J
,
Stundl
A
,
Debiec
R
,
Fontana
F
,
Krane
M
,
Sharipova
D
, et al.
Elucidation of the genetic causes of bicuspid aortic valve disease
.
Cardiovasc Res
2023
;
119
:
857
66
.

59

Yang
B
,
Zhou
W
,
Jiao
J
,
Nielsen
JB
,
Mathis
MR
,
Heydarpour
M
, et al.
Protein-altering and regulatory genetic variants near GATA4 implicated in bicuspid aortic valve
.
Nat Commun
2017
;
8:
15481
.

60

Tessler
I
,
Albuisson
J
,
Piñeiro-Sabarís
R
,
Verstraeten
A
,
Kamber Kaya
HE
,
Siguero-Álvarez
M
, et al.
Novel association of the NOTCH pathway regulator MIB1 gene with the development of bicuspid aortic valve
.
JAMA Cardiol
2023
;
8
:
721
31
.

61

Huang
T
,
Cheng
J
,
Feng
H
,
Zhou
W
,
Qiu
P
,
Zhou
D
, et al.
Bicuspid aortic valve-associated regulatory regions reveal GATA4 regulation and function during human-induced pluripotent stem cell-based endothelial-mesenchymal transition-brief report
.
Arterioscler Thromb Vasc Biol
2023
;
43
:
312
22
.

62

Lin
Y
,
Yang
Q
,
Lin
X
,
Liu
X
,
Qian
Y
,
Xu
D
, et al.
Extracellular matrix disorganization caused by ADAMTS16 deficiency leads to bicuspid aortic valve with raphe formation
.
Circulation
2024
;
149
:
605
26
.

63

Odelin
G
,
Faucherre
A
,
Marchese
D
,
Pinard
A
,
Jaouadi
H
,
Le Scouarnec
S
, et al.
Variations in the poly-histidine repeat motif of HOXA1 contribute to bicuspid aortic valve in mouse and zebrafish
.
Nat Commun
2023
;
14
:
1543
.

64

Guauque-Olarte
S
,
Droit
A
,
Tremblay-Marchand
J
,
Gaudreault
N
,
Kalavrouziotis
D
,
Dagenais
F
, et al.
RNA expression profile of calcified bicuspid, tricuspid, and normal human aortic valves by RNA sequencing
.
Physiol Genomics
2016
;
48
:
749
61
.

65

Yanagawa
B
,
Lovren
F
,
Pan
Y
,
Garg
V
,
Quan
A
,
Tang
G
, et al.
miRNA-141 is a novel regulator of BMP-2-mediated calcification in aortic stenosis
.
J Thorac Cardiovasc Surg
2012
;
144
:
256
62
.

66

Padang
R
,
Bagnall
RD
,
Tsoutsman
T
,
Bannon
PG
,
Semsarian
C
.
Comparative transcriptome profiling in human bicuspid aortic valve disease using RNA sequencing
.
Physiol Genomics
2015
;
47
:
75
87
.

67

Hadji
F
,
Boulanger
MC
,
Guay
SP
,
Gaudreault
N
,
Amellah
S
,
Mkannez
G
, et al.
Altered DNA methylation of long noncoding RNA H19 in calcific aortic valve disease promotes mineralization by silencing NOTCH1
.
Circulation
2016
;
134
:
1848
62
.

68

Mkannez
G
,
Gagné-Ouellet
V
,
Jalloul Nsaibia
M
,
Boulanger
M-C
,
Rosa
M
,
Argaud
D
, et al.
DNA methylation of a PLPP3 MIR transposon-based enhancer promotes an osteogenic programme in calcific aortic valve disease
.
Cardiovasc Res
2018
;
114
:
1525
35
.

69

Han
D
,
Zhou
T
,
Li
L
,
Ma
Y
,
Chen
S
,
Yang
C
, et al.
AVCAPIR: a novel procalcific PIWI-interacting RNA in calcific aortic valve disease
.
Circulation
2024
;
149
:
1578
97
.

70

Jaiswal
S
,
Natarajan
P
,
Silver
AJ
,
Gibson
CJ
,
Bick
AG
,
Shvartz
E
, et al.
Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease
.
N Engl J Med
2017
;
377
:
111
21
.

71

Mas-Peiro
S
,
Hoffmann
J
,
Fichtlscherer
S
,
Dorsheimer
L
,
Rieger
MA
,
Dimmeler
S
, et al.
Clonal haematopoiesis in patients with degenerative aortic valve stenosis undergoing transcatheter aortic valve implantation
.
Eur Heart J
2020
;
41
:
933
9
.

72

Vieceli Dalla Sega
F
,
Palumbo
D
,
Fortini
F
,
D'Agostino
Y
,
Cimaglia
P
,
Marracino
L
, et al.
Transcriptomic profiling of calcified aortic valves in clonal hematopoiesis of indeterminate potential carriers
.
Sci Rep
2022
;
12
:
20400
.

73

Simard
L
,
Côté
N
,
Dagenais
F
,
Mathieu
P
,
Couture
C
,
Trahan
S
, et al.
Sex-Related discordance between aortic valve calcification and hemodynamic severity of aortic stenosis: is valvular fibrosis the explanation?
Circ Res
2017
;
120
:
681
91
.

74

Voisine
M
,
Hervault
M
,
Shen
M
,
Boilard
AJ
,
Filion
B
,
Rosa
M
, et al.
Age, sex, and valve phenotype differences in fibro-calcific remodeling of calcified aortic valve
.
J Am Heart Assoc
2020
;
9
:
e015610
.

75

Aggarwal
SR
,
Clavel
MA
,
Messika-Zeitoun
D
,
Cueff
C
,
Malouf
J
,
Araoz
PA
, et al.
Sex differences in aortic valve calcification measured by multidetector computed tomography in aortic stenosis
.
Circ Cardiovasc Imaging
2013
;
6
:
40
7
.

76

Horitani
K
,
Chavkin
NW
,
Arai
Y
,
Wang
Y
,
Ogawa
H
,
Yura
Y
, et al.
Disruption of the Uty epigenetic regulator locus in hematopoietic cells phenocopies the profibrotic attributes of Y chromosome loss in heart failure
.
Nat Cardiovasc Res
2024
;
3
:
343
55
.

77

Haitjema
S
,
Kofink
D
,
van Setten
J
,
van der Laan
SW
,
Schoneveld
AH
,
Eales
J
, et al.
Loss of Y chromosome in blood is associated with major cardiovascular events during follow-up in men after carotid endarterectomy
.
Circ Cardiovasc Genet
2017
;
10
:
e001544
.

78

Mas-Peiro
S
,
Abplanalp
WT
,
Rasper
T
,
Berkowitsch
A
,
Leistner
DM
,
Dimmeler
S
, et al.
Mosaic loss of Y chromosome in monocytes is associated with lower survival after transcatheter aortic valve replacement
.
Eur Heart J
2023
;
44
:
1943
52
.

79

Aguado
BA
,
Walker
CJ
,
Grim
JC
,
Schroeder
ME
,
Batan
D
,
Vogt
BJ
, et al.
Genes that escape X chromosome inactivation modulate sex differences in valve myofibroblasts
.
Circulation
2022
;
145
:
513
30
.

80

Sarajlic
P
,
Plunde
O
,
Franco-Cereceda
A
,
Bäck
M
.
Artificial intelligence models reveal sex-specific gene expression in aortic valve calcification
.
JACC Basic Transl Sci
2021
;
6
:
403
12
.

81

Myasoedova
VA
,
Massaiu
I
,
Moschetta
D
,
Chiesa
M
,
Songia
P
,
Valerio
V
, et al.
Sex-specific cell types and molecular pathways indicate fibro-calcific aortic valve stenosis
.
Front Immunol
2022
;
13
:
747714
.

82

Carracedo
M
,
Pawelzik
SC
,
Artiach
G
,
Pouwer
MG
,
Plunde
O
,
Saliba-Gustafsson
P
, et al.
The tyrosine kinase inhibitor nilotinib targets the discoidin domain receptor DDR2 in calcific aortic valve stenosis
.
Br J Pharmacol
2022
;
179
:
4709
21
.

83

Han
Y
,
Zhang
J
,
Yang
Z
,
Jian
W
,
Zhu
Y
,
Gao
S
, et al.
Palmdelphin deficiency evokes NF-κB signaling in valvular endothelial cells and aggravates aortic valvular remodeling
.
JACC Basic Transl Sci
2023
;
8
:
1457
72
.

84

Chignon
A
,
Rosa
M
,
Boulanger
MC
,
Argaud
D
,
Devillers
R
,
Bon-Baret
V
, et al.
Enhancer-associated aortic valve stenosis risk locus 1p21.2 alters NFATC2 binding site and promotes fibrogenesis
.
iScience
2021
;
24
:
102241
.

85

Goettsch
C
,
Hutcheson
JD
,
Aikawa
M
,
Iwata
H
,
Pham
T
,
Nykjaer
A
, et al.
Sortilin mediates vascular calcification via its recruitment into extracellular vesicles
.
J Clin Invest
2016
;
126
:
1323
36
.

86

Iqbal
F
,
Schlotter
F
,
Becker-Greene
D
,
Lupieri
A
,
Goettsch
C
,
Hutcheson
JD
, et al.
Sortilin enhances fibrosis and calcification in aortic valve disease by inducing interstitial cell heterogeneity
.
Eur Heart J
2023
;
44
:
885
98
.

87

Gersony
WM
,
Hayes
CJ
,
Driscoll
DJ
,
Keane
JF
,
Kidd
L
,
O'Fallon
WM
, et al.
Bacterial endocarditis in patients with aortic stenosis, pulmonary stenosis, or ventricular septal defect
.
Circulation
1993
;
87
:
I121
126
. https://pubmed.ncbi.nlm.nih.gov/8425318/

88

Kolltveit
KM
,
Geiran
O
,
Tronstad
L
,
Olsen
I
.
Multiple bacteria in calcific aortic valve stenosis
.
Microb Ecol Health Dis
2002
;
14
:
110
7
.

89

Cohen
DJ
,
Malave
D
,
Ghidoni
JJ
,
Iakovidis
P
,
Everett
MM
,
You
S
, et al.
Role of oral bacterial flora in calcific aortic stenosis: an animal model
.
Ann Thorac Surg
2004
;
77
:
537
43
.

90

Passos
LSA
,
Jha
PK
,
Becker-Greene
D
,
Blaser
MC
,
Romero
D
,
Lupieri
A
, et al.
Prothymosin alpha: a novel contributor to estradiol receptor alpha-mediated CD8(+) T-cell pathogenic responses and recognition of type 1 collagen in rheumatic heart valve disease
.
Circulation
2022
;
145
:
531
48
.

91

Chen
HY
,
Cairns
BJ
,
Small
AM
,
Burr
HA
,
Ambikkumar
A
,
Martinsson
A
, et al.
Association of FADS1/2 locus variants and polyunsaturated fatty acids with aortic stenosis
.
JAMA Cardiol
2020
;
5
:
694
702
.

92

Plunde
O
,
Larsson
SC
,
Artiach
G
,
Thanassoulis
G
,
Carracedo
M
,
Franco-Cereceda
A
, et al.
FADS1 (fatty acid desaturase 1) genotype associates with aortic valve FADS mRNA expression, fatty acid content and calcification
.
Circ Genom Precis Med
2020
;
13
:
e002710
.

93

Bäck
M
,
Michel
JB
.
From organic and inorganic phosphates to valvular and vascular calcifications
.
Cardiovasc Res
2021
;
117
:
2016
29
.

94

Yang
X
,
Fullerton
DA
,
Su
X
,
Ao
L
,
Cleveland
JC
Jr
,
Meng
X
.
Pro-osteogenic phenotype of human aortic valve interstitial cells is associated with higher levels of Toll-like receptors 2 and 4 and enhanced expression of bone morphogenetic protein 2
.
J Am Coll Cardiol
2009
;
53
:
491
500
.

95

Gollmann-Tepeköylü
C
,
Graber
M
,
Hirsch
J
,
Mair
S
,
Naschberger
A
,
Pölzl
L
, et al.
Toll-like receptor 3 mediates aortic stenosis through a conserved mechanism of calcification
.
Circulation
2023
;
147
:
1518
33
.

96

Niepmann
ST
,
Willemsen
N
,
Boucher
AS
,
Stei
M
,
Goody
P
,
Zietzer
A
, et al.
Toll-like receptor-3 contributes to the development of aortic valve stenosis
.
Basic Res Cardiol
2023
;
118
:
6
.

97

Karadimou
G
,
Plunde
O
,
Pawelzik
S-C
,
Carracedo
M
,
Eriksson
P
,
Franco-Cereceda
A
, et al.
TLR7 expression is associated with M2 macrophage subset in calcific aortic valve stenosis
.
Cells
2020
;
9
:
1710
.

98

Yuan
S
,
Bäck
M
,
Bruzelius
M
,
Mason
AM
,
Burgess
S
,
Larsson
S
.
Plasma phospholipid fatty acids, FADS1 and risk of 15 cardiovascular diseases: a Mendelian randomisation study
.
Nutrients
2019
;
11
:
3001
.

99

Cayer
LGJ
,
Surendran
A
,
Karakach
T
,
Aukema
HM
,
Ravandi
A
.
Valvular prostaglandins are elevated in severe human aortic valve stenosis
.
Arterioscler Thromb Vasc Biol
2024
;
44
:
e131
44
.

100

Artiach
G
,
Carracedo
M
,
Plunde
O
,
Wheelock
CE
,
Thul
S
,
Sjövall
P
, et al.
Omega-3 polyunsaturated fatty acids decrease aortic valve disease through the resolvin E1 and ChemR23 axis
.
Circulation
2020
;
142
:
776
89
.

101

Laguna-Fernandez
A
,
Checa
A
,
Carracedo
M
,
Artiach
G
,
Petri
MH
,
Baumgartner
R
, et al.
ERV1/ChemR23 signaling protects against atherosclerosis by modifying oxidized low-density lipoprotein uptake and phagocytosis in macrophages
.
Circulation
2018
;
138
:
1693
705
.

102

Blaser
MC
,
Aikawa
E
.
Differential miRNA loading underpins dual harmful and protective roles for extracellular vesicles in atherogenesis
.
Circ Res
2019
;
124
:
467
9
.

103

Raju
S
,
Botts
SR
,
Blaser
MC
,
Abdul-Samad
M
,
Prajapati
K
,
Khosraviani
N
, et al.
Directional endothelial communication by polarized extracellular vesicle release
.
Circ Res
2024
;
134
:
269
89
.

104

Hosen
MR
,
Goody
PR
,
Zietzer
A
,
Xiang
X
,
Niepmann
ST
,
Sedaghat
A
, et al.
Circulating microRNA-122-5p is associated with a lack of improvement in left ventricular function after transcatheter aortic valve replacement and regulates viability of cardiomyocytes through extracellular vesicles
.
Circulation
2022
;
146
:
1836
54
.

105

Hutcheson
JD
,
Goettsch
C
,
Bertazzo
S
,
Maldonado
N
,
Ruiz
JL
,
Goh
W
, et al.
Genesis and growth of extracellular-vesicle-derived microcalcification in atherosclerotic plaques
.
Nat Mater
2016
;
15
:
335
43
.

106

Kawakami
R
,
Katsuki
S
,
Travers
R
,
Romero
DC
,
Becker-Greene
D
,
Passos
LSA
, et al.
S100A9-RAGE axis accelerates formation of macrophage-mediated extracellular vesicle microcalcification in diabetes mellitus
.
Arterioscler Thromb Vasc Biol
2020
;
40
:
1838
53
.

107

Aikawa
E
,
Blaser
MC
.
2020 Jeffrey M. Hoeg award lecture: calcifying extracellular vesicles as building blocks of microcalcifications in cardiovascular disorders
.
Arterioscler Thromb Vasc Biol
2021
;
41
:
117
27
.

108

van der Valk
D
,
van der Ven
C
,
Blaser
M
,
Grolman
J
,
Wu
P-J
,
Fenton
O
, et al.
Engineering a 3D-bioprinted model of human heart valve disease using nanoindentation-based biomechanics
.
Nanomaterials
2018
;
8
:
296
.

109

Clift
CL
,
Blaser
MC
,
Gerrits
W
,
Turner
ME
,
Sonawane
A
,
Pham
T
, et al.
Intracellular proteomics and extracellular vesiculomics as a metric of disease recapitulation in 3D-bioprinted aortic valve arrays
.
Sci Adv
2024
;
10
:eadj9793.

110

Xu
K
,
Xie
S
,
Huang
Y
,
Zhou
T
,
Liu
M
,
Zhu
P
, et al.
Cell-type transcriptome atlas of human aortic valves reveal cell heterogeneity and endothelial to mesenchymal transition involved in calcific aortic valve disease
.
Arterioscler Thromb Vasc Biol
2020
;
40
:
2910
21
.

111

Hulin
A
,
Hortells
L
,
Gomez-Stallons
MV
,
O'Donnell
A
,
Chetal
K
,
Adam
M
, et al.
Maturation of heart valve cell populations during postnatal remodeling
.
Development
2019
;
146
:
dev173047
.

112

Shu
S
,
Fu
M
,
Chen
X
,
Zhang
N
,
Zhao
R
,
Chang
Y
, et al.
Cellular landscapes of nondiseased human cardiac valves from end-stage heart failure-explanted heart
.
Arterioscler Thromb Vasc Biol
2022
;
42
:
1429
46
.

113

Lee
SH
,
Kim
N
,
Kim
M
,
Woo
SH
,
Han
I
,
Park
J
, et al.
Single-cell transcriptomics reveal cellular diversity of aortic valve and the immunomodulation by PPARγ during hyperlipidemia
.
Nat Commun
2022
;
13
:
5461
.

114

Huang
Y
,
Wang
C
,
Zhou
T
,
Xie
F
,
Liu
Z
,
Xu
H
, et al.
Lumican promotes calcific aortic valve disease through H3 histone lactylation
.
Eur Heart J
2024
;
45
:
3871
85
.

115

Queen
R
,
Crosier
M
,
Eley
L
,
Kerwin
J
,
Turner
JE
,
Yu
J
, et al.
Spatial transcriptomics reveals novel genes during the remodelling of the embryonic human arterial valves
.
PLoS Genet
2023
;
19
:
e1010777
.

116

Lindman
BR
,
Sukul
D
,
Dweck
MR
,
Madhavan
MV
,
Arsenault
BJ
,
Coylewright
M
, et al.
Evaluating medical therapy for calcific aortic stenosis: JACC state-of-the-art review
.
J Am Coll Cardiol
2021
;
78
:
2354
76
.

117

Rossebø
AB
,
Pedersen
TR
,
Boman
K
,
Brudi
P
,
Chambers
JB
,
Egstrup
K
, et al.
Intensive lipid lowering with simvastatin and ezetimibe in aortic stenosis
.
N Engl J Med
2008
;
359
:
1343
56
.

118

Chan
KL
,
Teo
K
,
Dumesnil
JG
,
Ni
A
,
Tam
J
,
Investigators
A
.
Effect of lipid lowering with rosuvastatin on progression of aortic stenosis: results of the aortic stenosis progression observation: measuring effects of rosuvastatin (ASTRONOMER) trial
.
Circulation
2010
;
121
:
306
14
.

119

Cowell
SJ
,
Newby
DE
,
Prescott
RJ
,
Bloomfield
P
,
Reid
J
,
Northridge
DB
, et al.
A randomized trial of intensive lipid-lowering therapy in calcific aortic stenosis
.
N Engl J Med
2005
;
352
:
2389
97
.

120

Gerdts
E
,
Rossebø
AB
,
Pedersen
TR
,
Boman
K
,
Brudi
P
,
Chambers
JB
, et al.
Impact of baseline severity of aortic valve stenosis on effect of intensive lipid lowering therapy (from the SEAS study)
.
Am J Cardiol
2010
;
106
:
1634
9
.

121

Greve
AM
,
Bang
CN
,
Boman
K
,
Egstrup
K
,
Kesäniemi
YA
,
Ray
S
, et al.
Relation of lipid-lowering therapy to need for aortic valve replacement in patients with asymptomatic mild to moderate aortic stenosis
.
Am J Cardiol
2019
;
124
:
1736
40
.

122

Bergmark
BA
,
O'Donoghue
ML
,
Murphy
SA
,
Kuder
JF
,
Ezhov
MV
,
Ceška
R
, et al.
An exploratory analysis of proprotein convertase subtilisin/kexin type 9 inhibition and aortic stenosis in the FOURIER trial
.
JAMA Cardiol
2020
;
5
:
709
13
.

123

A Randomized Trial of PCSK9 Inhibitors in Calcific Aortic Valve Stenosis. https://clinicaltrials.gov/study/NCT04968509. Date accessed 10 July 2024.

124

Proprotein Convertase Subtilisin Kexin Type 9 Inhibitor in Aortic Stenosis
. https://clinicaltrials.gov/study/NCT03051360. Date accessed 10 July 2024.

125

Perrot
N
,
Valerio
V
,
Moschetta
D
,
Boekholdt
SM
,
Dina
C
,
Chen
HY
, et al.
Genetic and in vitro inhibition of PCSK9 and calcific aortic valve stenosis
.
JACC Basic Transl Sci
2020
;
5
:
649
61
.

126

A Pilot, Randomized Controlled-trial of Lipoprotein(a) Lowering for the Prevention of Aortic Valve Disease-translating Genomic Knowledge for Cardiovascular Prevention.
https://clinicaltrials.gov/study/NCT02109614. Date accessed 10 July 2024.

127

Lamina
C
,
Kronenberg
F
.
Estimation of the required lipoprotein(a)-lowering therapeutic effect size for reduction in coronary heart disease outcomes: a Mendelian randomization analysis
.
JAMA Cardiol
2019
;
4
:
575
9
.

128

A Randomized Double-blind, Placebo-controlled, Multicenter Trial Assessing the Impact of Lipoprotein(a) Lowering with Pelacarsen (TQJ230) on the Progression of Calcific Aortic Valve Stenosis [Lp(a)FRONTIERS CAVS]
. https://clinicaltrials.gov/study/NCT05646381. Date accessed 10 July 2024.

129

Rogers
MA
,
Bartoli-Leonard
F
,
Zheng
KH
,
Small
AM
,
Chen
HY
,
Clift
CL
, et al.
Major facilitator superfamily domain containing 5 inhibition reduces lipoprotein(a) uptake and calcification in valvular heart disease
.
Circulation
2024
;
149
:
391
401
.

130

Bergmark
BA
,
Marston
NA
,
Prohaska
TA
,
Alexander
VJ
,
Zimerman
A
,
Moura
FA
, et al.
Olezarsen for hypertriglyceridemia in patients at high cardiovascular risk
.
N Engl J Med
2024
;
390
:
1770
80
.

131

Bhatt
DL
,
Steg
PG
,
Miller
M
,
Brinton
EA
,
Jacobson
TA
,
Ketchum
SB
, et al.
Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia
.
N Engl J Med
2019
;
380
:
11
22
.

132

Das Pradhan
A
,
Glynn
RJ
,
Fruchart
JC
,
MacFadyen
JG
,
Zaharris
ES
,
Everett
BM
, et al.
Triglyceride lowering with pemafibrate to reduce cardiovascular risk
.
N Engl J Med
2022
;
387
:
1923
34
.

133

Tardif
JC
,
Kouz
S
,
Waters
DD
,
Bertrand
OF
,
Diaz
R
,
Maggioni
AP
, et al.
Efficacy and safety of low-dose colchicine after myocardial infarction
.
N Engl J Med
2019
;
381
:
2497
505
.

134

Effect of Colchicine on the Progression of Aortic Valve Stenosis—A Pilot Study. https://clinicaltrials.gov/study/NCT05253794. Date accessed 10 July 2024.

135

Does Colchicine Reduce Progression of Aortic Valve Stenosis? https://clinicaltrials.gov/study/NCT05162742. Date accessed 10 July 2024.

136

Chu
Y
,
Lund
DD
,
Weiss
RM
,
Brooks
RM
,
Doshi
H
,
Hajj
GP
, et al.
Pioglitazone attenuates valvular calcification induced by hypercholesterolemia
.
Arterioscler Thromb Vasc Biol
2013
;
33
:
523
32
.

137

Effect of Pioglitazone Treatment in Patient's Calcific Aortic Valve Disease with Mild Aortic Valve Stenosis
. https://clinicaltrials.gov/study/NCT05875675. Date accessed 10 July 2024.

138

Choi
B
,
Lee
S
,
Kim
SM
,
Lee
EJ
,
Lee
SR
,
Kim
DH
, et al.
Dipeptidyl peptidase-4 induces aortic valve calcification by inhibiting insulin-like growth factor-1 signaling in valvular interstitial cells
.
Circulation
2017
;
135
:
1935
50
.

139

An Adaptive Phase 2/3 Multicenter, Double-Blind, Placebo-Controlled, Randomized, Parallel, 3 Arm Study to Evaluate the Efficacy and Safety of DA-1229 (Evogliptin) in Patient's Calcific Aortic Valve Disease with Mild to Moderate Aortic Stenosis (EVOID-AS)
. https://clinicaltrials.gov/study/NCT05143177. Date accessed 10 July 2024.

140

A Multicenter, Double-blind, Placebo-controlled, Stratified-randomized, Parallel, Therapeutic Exploratory Clinical Study to Evaluate the Efficacy and Safety of DA-1229 in Patients with Calcific Aortic Valve Disease
. https://clinicaltrials.gov/study/NCT04055883. Date accessed 10 July 2024.

141

Song
J-K
,
Lee
S
,
Kim
Y-J
,
Kim
H-K
,
Ha
J-W
,
Choi
E-Y
, et al.
Effect of evogliptin on the progression of aortic valvular calcification
.
J Am Coll Cardiol
2024
;
84
:
1064
75
.

142

Lincoff
AM
,
Brown-Frandsen
K
,
Colhoun
HM
,
Deanfield
J
,
Emerson
SS
,
Esbjerg
S
, et al.
Semaglutide and cardiovascular outcomes in obesity without diabetes
.
N Engl J Med
2023
;
389
:
2221
32
.

143

Xiao
F
,
Zha
Q
,
Zhang
Q
,
Wu
Q
,
Chen
Z
,
Yang
Y
, et al.
Decreased glucagon-like peptide-1 is associated with calcific aortic valve disease: GLP-1 suppresses the calcification of aortic valve interstitial cells
.
Front Cardiovasc Med
2021
;
8
:
709741
.

144

Zhou
Y
,
Yuan
Z
,
Wang
M
,
Zhang
Z
,
Tan
C
,
Yu
J
, et al.
Liraglutide attenuates aortic valve calcification in a high-cholesterol-diet-induced experimental calcific aortic valve disease model in apolipoprotein E-deficient mice
.
J Cardiovasc Dev Dis
2023
;
10
:
386
.

145

Majumdar
U
,
Manivannan
S
,
Basu
M
,
Ueyama
Y
,
Blaser
MC
,
Cameron
E
, et al.
Nitric oxide prevents aortic valve calcification by S-nitrosylation of USP9X to activate NOTCH signaling
.
Sci Adv
2021
;
7
:eabe3706.

146

Yip
CY
,
Blaser
MC
,
Mirzaei
Z
,
Zhong
X
,
Simmons
CA
.
Inhibition of pathological differentiation of valvular interstitial cells by C-type natriuretic peptide
.
Arterioscler Thromb Vasc Biol
2011
;
31
:
1881
9
.

147

A Phase II Randomized, Placebo-Controlled, Double-Blinded Study Evaluating the Effects of Ataciguat (HMR1766) on Aortic Valve Calcification in Patients with Moderate Calcific Aortic Valve Stenosis
. https://clinicaltrials.gov/study/NCT02481258. Date accessed 10 July 2024.

148

Zhang
B
,
Roos
C
,
Hagler
M
,
Verzosa
G
,
Zhang
H
,
Schaff
H
, et al.
Abstract 123: activation of oxidized soluble guanylate cyclase slows progression of aortic valve calcification
.
Arterioscler Thromb Vasc Biol
2019
;
39
:
A123
. https://www.ahajournals.org/doi/10.1161/atvb.39.suppl_1.123

149

SALTIRE II: Bisphosphonates and RANKL Inhibition in Aortic Stenosis
. https://clinicaltrials.gov/study/NCT02132026. Date accessed 10 July 2024.

150

Pawade
TA
,
Doris
MK
,
Bing
R
,
White
AC
,
Forsyth
L
,
Evans
E
, et al.
Effect of denosumab or alendronic acid on the progression of aortic stenosis: a double-blind randomized controlled trial
.
Circulation
2021
;
143
:
2418
27
.

151

Ferrer
MD
,
Ketteler
M
,
Tur
F
,
Tur
E
,
Isern
B
,
Salcedo
C
, et al.
Characterization of SNF472 pharmacokinetics and efficacy in uremic and non-uremic rats models of cardiovascular calcification
.
PLoS One
2018
;
13
:
e0197061
.

152

Zabirnyk
A
,
Perez
MDM
,
Blasco
M
,
Stensløkken
KO
,
Ferrer
MD
,
Salcedo
C
, et al.
A novel ex vivo model of aortic valve calcification. A preliminary report
.
Front Pharmacol
2020
;
11
:
568764
.

153

Zabirnyk
A
,
Ferrer
MD
,
Bogdanova
M
,
Pérez
MM
,
Salcedo
C
,
Kaljusto
ML
, et al.
SNF472, a novel anti-crystallization agent, inhibits induced calcification in an in vitro model of human aortic valve calcification
.
Vascul Pharmacol
2019
;
122–123
:
106583
.

154

A Double-blind, Randomised, Placebo-controlled Study to Assess the Effect of SNF472 on Progression of Cardiovascular Calcification on Top of Standard of Care in End-stage-renal-disease (ESRD) Patients on Hemodialysis (HD)
. https://clinicaltrials.gov/study/NCT02966028. Date accessed 10 July 2024.

155

Raggi
P
,
Bellasi
A
,
Bushinsky
D
,
Bover
J
,
Rodriguez
M
,
Ketteler
M
, et al.
Slowing progression of cardiovascular calcification with SNF472 in patients on hemodialysis: results of a randomized phase 2b study
.
Circulation
2020
;
141
:
728
39
.

156

Schantl
AE
,
Verhulst
A
,
Neven
E
,
Behets
GJ
,
D'Haese
PC
,
Maillard
M
, et al.
Inhibition of vascular calcification by inositol phosphates derivatized with ethylene glycol oligomers
.
Nat Commun
2020
;
11
:
721
.

157

A First-in-human, Randomized, Double-blind, Placebo-controlled, Single and Multiple Ascending Dose Study to Assess the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of INS-3001 in Healthy Subjects and Patients with Moderate Aortic Valve Stenosis
. https://onderzoekmetmensen.nl/en/trial/52051. Date accessed 10 July 2024.

158

Chiyoya
M
,
Seya
K
,
Yu
Z
,
Daitoku
K
,
Motomura
S
,
Imaizumi
T
, et al.
Matrix Gla protein negatively regulates calcification of human aortic valve interstitial cells isolated from calcified aortic valves
.
J Pharmacol Sci
2018
;
136
:
257
65
.

159

Liu
C
,
Liu
H
,
Xie
T
.
Impact of Fetuin-A, Lp(a), matrix Gla protein and macrophage density on calcific aortic valve disease: a clinical study
.
Lipids Health Dis
2022
;
21
:
14
.

160

Decalcification of the Aortic Valve by Vitamin K2 (Menaquinone-7). https://clinicaltrials.gov/study/NCT03305536. Date accessed 10 July 2024.

161

The Effects of Menaquinone-7 Supplementation in Patients with Aortic Valves Calcification
. https://clinicaltrials.gov/study/NCT03243890. Date accessed 10 July 2024.

162

Bicuspid Aortic Valve Stenosis and the Effect of vItamin K2 on Calciummetabolism on 18F-NaF PET/MRI (BASIK2): a Pilot Study
. https://clinicaltrials.gov/study/NCT02917525. Date accessed 10 July 2024.

163

Feldmann
A
,
Nitschke
Y
,
Linß
F
,
Mulac
D
,
Stücker
S
,
Bertrand
J
, et al.
Improved reversion of calcifications in porcine aortic heart valves using elastin-targeted nanoparticles
.
Int J Mol Sci
2023
;
24
:
16471
.

164

Heuschkel
MA
,
Skenteris
NT
,
Hutcheson
JD
,
van der Valk
DD
,
Bremer
J
,
Goody
P
, et al.
Integrative multi-omics analysis in calcific aortic valve disease reveals a link to the formation of amyloid-like deposits
.
Cells
2020
;
9
:
2164
.

165

Audet
A
,
Côté
N
,
Couture
C
,
Bossé
Y
,
Després
JP
,
Pibarot
P
, et al.
Amyloid substance within stenotic aortic valves promotes mineralization
.
Histopathology
2012
;
61
:
610
9
.

166

Shah
SJ
,
Fine
N
,
Garcia-Pavia
P
,
Klein
AL
,
Fernandes
F
,
Weissman
NJ
, et al.
Effect of tafamidis on cardiac function in patients with transthyretin amyloid cardiomyopathy: a post hoc analysis of the ATTR-ACT randomized clinical trial
.
JAMA Cardiol
2024
;
9
:
25
34
.

167

Urbano Pagan
L
,
Gomes
MJ
,
Damatto
FC
,
Oliveira
JPG
,
Gatto
M
,
Souza
LM
, et al.
Effects of SGLT2 inhibition on cardiac remodeling and heart failure development in rats with aortic stenosis
.
Eur Heart J
2023
;
44
:ehad655.1661.

168

Capoulade
R
,
Clavel
MA
,
Mathieu
P
,
Côté
N
,
Dumesnil
JG
,
Arsenault
M
, et al.
Impact of hypertension and renin-angiotensin system inhibitors in aortic stenosis
.
Eur J Clin Invest
2013
;
43
:
1262
72
.

169

Safety and Efficacy of Angiotensin Receptor Blocker, Fimasartan, on Patients with Aortic Stenosis
. https://clinicaltrials.gov/study/NCT01589380. Date accessed 10 July 2024.

170

Impact of Angiotensin Receptor Blockers in Aortic Stenosis—a Randomized Controlled Trial
. https://clinicaltrials.gov/study/NCT04913870. Date accessed 10 July 2024.

171

O'Brien
KD
,
Shavelle
DM
,
Caulfield
MT
,
McDonald
TO
,
Olin-Lewis
K
,
Otto
CM
, et al.
Association of angiotensin-converting enzyme with low-density lipoprotein in aortic valvular lesions and in human plasma
.
Circulation
2002
;
106
:
2224
30
.

172

Helske
S
,
Lindstedt
KA
,
Laine
M
,
Mäyränpää
M
,
Werkkala
K
,
Lommi
J
, et al.
Induction of local angiotensin II-producing systems in stenotic aortic valves
.
J Am Coll Cardiol
2004
;
44
:
1859
66
.

173

Arishiro
K
,
Hoshiga
M
,
Negoro
N
,
Jin
D
,
Takai
S
,
Miyazaki
M
, et al.
Angiotensin receptor-1 blocker inhibits atherosclerotic changes and endothelial disruption of the aortic valve in hypercholesterolemic rabbits
.
J Am Coll Cardiol
2007
;
49
:
1482
9
.

174

Xie
C
,
Shen
Y
,
Hu
W
,
Chen
Z
,
Li
Y
.
Angiotensin II promotes an osteoblast-like phenotype in porcine aortic valve myofibroblasts
.
Aging Clin Exp Res
2016
;
28
:
181
7
.

175

Carter-Storch
R
,
Le Nezet
E
,
Ali
M
,
Powers
A
,
Haujir
A
,
Demers
K
, et al.
Angiotensin II receptor blockers are associated with reduced valvular fibrosis in women with aortic stenosis
.
Can J Cardiol
2024
;
40
:
1690
9
.

176

Shelbaya
K
,
Arthur
V
,
Yang
Y
,
Dorbala
P
,
Buckley
L
,
Claggett
B
, et al.
Large-scale proteomics identifies novel biomarkers and circulating risk factors for aortic stenosis
.
J Am Coll Cardiol
2024
;
83
:
577
91
.

177

Lin
R
,
Zhu
Y
,
Chen
W
,
Wang
Z
,
Wang
Y
,
Du
J
.
Identification of circulating inflammatory proteins associated with calcific aortic valve stenosis by multiplex analysis
.
Cardiovasc Toxicol
2024
;
24
:
499
512
.

178

Wenzl
FA
,
Kraler
S
,
Ambler
G
,
Weston
C
,
Herzog
SA
,
Räber
L
, et al.
Sex-specific evaluation and redevelopment of the GRACE score in non-ST-segment elevation acute coronary syndromes in populations from the UK and Switzerland: a multinational analysis with external cohort validation
.
Lancet
2022
;
400
:
744
56
.

179

Pekayvaz
K
,
Losert
C
,
Knottenberg
V
,
Gold
C
,
van Blokland
IV
,
Oelen
R
, et al.
Multiomic analyses uncover immunological signatures in acute and chronic coronary syndromes
.
Nat Med
2024
;
30
:
1696
710
.

180

Schroeder
ME
,
Batan
D
,
Gonzalez Rodriguez
A
,
Speckl
KF
,
Peters
DK
,
Kirkpatrick
BE
, et al.
Osteopontin activity modulates sex-specific calcification in engineered valve tissue mimics
.
Bioeng Transl Med
2023
;
8
:
e10358
.

181

Shroff
GR
,
Bangalore
S
,
Bhave
NM
,
Chang
TI
,
Garcia
S
,
Mathew
RO
, et al.
Evaluation and management of aortic stenosis in chronic kidney disease: a scientific statement from the American Heart Association
.
Circulation
2021
;
143
:
e1088
114
.

182

Perpétuo
L
,
Barros
AS
,
Dalsuco
J
,
Nogueira-Ferreira
R
,
Resende-Gonçalves
P
,
Falcão-Pires
I
, et al.
Coronary artery disease and aortic valve stenosis: a urine proteomics study
.
Int J Mol Sci
2022
;
23
:
13579
.

183

Xu
R
,
Huang
Y
,
Zhu
D
,
Guo
J
.
Iron promotes Slc7a11-deficient valvular interstitial cell osteogenic differentiation: a possible mechanism by which ferroptosis participates in intraleaflet hemorrhage-induced calcification
.
Free Radic Biol Med
2022
;
184
:
158
69
.

184

Morvan
M
,
Arangalage
D
,
Franck
G
,
Perez
F
,
Cattan-Levy
L
,
Codogno
I
, et al.
Relationship of iron deposition to calcium deposition in human aortic valve leaflets
.
J Am Coll Cardiol
2019
;
73
:
1043
54
.

185

Laguna-Fernandez
A
,
Carracedo
M
,
Jeanson
G
,
Nagy
E
,
Eriksson
P
,
Caligiuri
G
, et al.
Iron alters valvular interstitial cell function and is associated with calcification in aortic stenosis
.
Eur Heart J
2016
;
37
:
3532
5
.

186

Tastet
L
,
Shen
M
,
Capoulade
R
,
Arsenault
M
,
Bédard
É
,
Côté
N
, et al.
Bone mineral density and progression rate of calcific aortic valve stenosis
.
J Am Coll Cardiol
2020
;
75
:
1725
6
.

187

Hjortnaes
J
,
Butcher
J
,
Figueiredo
JL
,
Riccio
M
,
Kohler
RH
,
Kozloff
KM
, et al.
Arterial and aortic valve calcification inversely correlates with osteoporotic bone remodelling: a role for inflammation
.
Eur Heart J
2010
;
31
:
1975
84
.

188

Mehta
CK
,
Liu
TX
,
Bonnell
L
,
Habib
RH
,
Kaneko
T
,
Flaherty
JD
, et al.
Age-stratified surgical aortic valve replacement for aortic stenosis
.
Ann Thorac Surg
2024
;
118
:
430
8
.

189

Zhu
Z
,
Liu
Z
,
Zhang
D
,
Li
L
,
Pei
J
,
Cai
L
.
Models for calcific aortic valve disease in vivo and in vitro
.
Cell Regen
2024
;
13
:
6
.

190

Duan
B
,
Kapetanovic
E
,
Hockaday
LA
,
Butcher
JT
.
Three-dimensional printed trileaflet valve conduits using biological hydrogels and human valve interstitial cells
.
Acta Biomater
2014
;
10
:
1836
46
.

191

Duan
B
,
Yin
Z
,
Hockaday Kang
L
,
Magin
RL
,
Butcher
JT
.
Active tissue stiffness modulation controls valve interstitial cell phenotype and osteogenic potential in 3D culture
.
Acta Biomater
2016
;
36
:
42
54
.

192

Tandon
I
,
Woessner
AE
,
Ferreira
LA
,
Shamblin
C
,
Vaca-Diez
G
,
Walls
A
, et al.
A three-dimensional valve-on-chip microphysiological system implicates cell cycle progression, cholesterol metabolism and protein homeostasis in early calcific aortic valve disease progression
.
Acta Biomater
2024
;
186
:
167
84
.

193

Goto
S
,
Rogers
MA
,
Blaser
MC
,
Higashi
H
,
Lee
LH
,
Schlotter
F
, et al.
Standardization of human calcific aortic valve disease in vitro modeling reveals passage-dependent calcification
.
Front Cardiovasc Med
2019
;
6
:
49
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)