Multiple biomarker panel to screen for severe aortic stenosis: results from the CASABLANCA study ================================================================================================ * Sammy Elmariah * Cian McCarthy * Nasrien Ibrahim * Deborah Furman * Renata Mukai * Craig Magaret * Rhonda Rhyne * Grady Barnes * Roland R J van Kimmenade * James L Januzzi, Jr ## Abstract **Objective** Severe aortic valve stenosis (AS) develops via insidious processes and can be challenging to correctly diagnose. We sought to develop a circulating biomarker panel to identify patients with severe AS. **Methods** We enrolled study participants undergoing coronary or peripheral angiography for a variety of cardiovascular diseases at a single academic medical centre. A panel of 109 proteins were measured in blood obtained at the time of the procedure. Statistical learning methods were used to identify biomarkers and clinical parameters that associate with severe AS. A diagnostic model incorporating clinical and biomarker results was developed and evaluated using Monte Carlo cross-validation. **Results** Of 1244 subjects (age 66.4±11.5  years, 28.7% female), 80 (6.4%) had severe AS (defined as aortic valve area (AVA) <1.0  cm2). A final model included age, N-terminal pro-B-type natriuretic peptide, von Willebrand factor and fetuin-A. The model had good discrimination for severe AS (OR=5.9, 95% CI 3.5 to 10.1, p<0.001) with an area under the curve of 0.76 insample and 0.74 with cross-validation. A diagnostic score was generated. Higher prevalence of severe AS was noted in those with higher scores, such that 1.6% of those with a score of 1 had severe AS compared with 15.3% with a score of 5 (p<0.001), and score values were inversely correlated with AVA (r=−0.35; p<0.001). At optimal model cut-off, we found 76% sensitivity, 65% specificity, 13% positive predictive value and 98% negative predictive value. **Conclusions** We describe a novel, multiple biomarker approach for diagnostic evaluation of severe AS. **Trial registration number** [NCT00842868](http://openheart.bmj.com/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT00842868&atom=%2Fopenhrt%2F5%2F2%2Fe000916.atom). * aortic stenosis * biomarkers * valvular heart disease ### Key questions #### What is already known about this subject? * Severe aortic stenosis develops insidiously and is associated with markedly increased morbidity and mortality when not recognised and treated in a timely manner. #### What does this study add? * We developed and evaluated a model including age and three proteins, N-terminal pro-B-type natriuretic peptide, von Willebrand factor and fetuin-A, which identified patients with severe aortic stenosis. #### How might this impact on clinical practice? * A model composed of age and three circulating biomarkers may supplement currently available diagnostic methods to facilitate the diagnosis of severe aortic valve stenosis and may serve as the basis for population screening algorithms. ## Introduction Calcific aortic stenosis (AS) is the most common cause of valvular heart disease in the Western world, present in >20% of older adults.1 As the proportion of elderly Americans grows, so too will the prevalence and socioeconomic burden of AS. AS progresses via an indolent process with symptoms developing at a late stage of the disease. Once symptoms occur, AS portends a dismal prognosis unless the aortic valve is replaced; without aortic valve replacement (AVR), only half of patients will survive 1 year.2 Current clinical practice guidelines recommend deferring AVR until AS severity reaches the severe stage and until onset of clinical symptoms or the occurrence of overt left ventricular (LV) systolic dysfunction.3 Classic teaching suggests that survival normalises after AVR; however, evidence is mounting that survival, symptom improvement and quality of life are diminished when valve replacement is performed late. With progressive valve narrowing, compensatory LV remodelling mechanisms fail and become maladaptive, in turn resulting in irreversible myocardial injury and fibrosis. For example, we have demonstrated that the absence of LV ejection fraction (LVEF) improvement within 1 month after transcatheter AVR is associated with a tripling of the risk of 1-year all-cause mortality and fivefold increase in 1-year cardiac death,4 and persistent LV hypertrophy is similarly associated with increased mortality.5 Myocardial fibrosis, a late sequela of AS, has also been associated with markedly increased risk of mortality, regardless of whether the stenotic aortic valve is replaced.6 Early recognition of severe AS is important for maximising health and survival after valve replacement. While clinical history is important, symptoms caused by severe AS are often incorrectly attributed to comorbid conditions or simply to advanced age, delaying appropriate diagnosis and therapy. Physical examination may be useful to identify AS and grade its severity, yet skills in such evaluation may be limited in clinical practice.7 Echocardiography is the gold-standard diagnostic test for the diagnosis and grading of AS severity, yet several inconsistencies with the echocardiographic assessment of AS exist that may hinder appropriate clinical management decisions. Echocardiography is also time-consuming, potentially costly and requires specialised interpretation. Together, the aforementioned observations highlight the detrimental impact of delayed intervention for severe AS and highlight the need for clinical tools to assist in the rapid identification of severe AS. We therefore sought to develop a biomarker panel to identify patients with severe AS within the Catheter Sampled Blood Archive in Cardiovascular Diseases (CASABLANCA) study. Such a blood-based biomarker panel may serve as a convenient and low-cost method of screening for severe AS. ## Methods The design of the CASABLANCA study has been described previously (ClinicalTrials.gov [NCT00842868](http://openheart.bmj.com/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT00842868&atom=%2Fopenhrt%2F5%2F2%2Fe000916.atom)).8 9 Briefly, the CASABLANCA study was a prospective, single-centre, investigator-initiated, observational cohort study performed at the Massachusetts General Hospital to investigate novel biomarkers in subjects who underwent coronary and peripheral angiography with or without intervention between 2008 and 2011. Patients were referred for these procedures for several reasons, including angiography for acute coronary syndromes, stable angina, heart failure, abnormal cardiac functional test, peripheral arterial disease, and preoperatively before heart valve surgery or transcatheter intervention. All subjects provided informed consent. ### Data acquisition Detailed clinical and historical variables and reason for referral for angiography were recorded from medical record review and subject interviews at the time of the procedure. Echocardiographic data from studies performed within 6 months of enrolment were collected retrospectively. Transthoracic echocardiographic images were obtained using standard views for routine clinical care. Recorded echocardiographic parameters include measures of AS severity (valve area, transvalvular gradients), LV chamber dimensions, hypertrophic remodelling (absolute and relative wall thickness and LV mass), systolic and diastolic function, and other valvular abnormalities. Measures of AS severity were available in patients with at least moderate AS. All measurements were performed as outlined by the American Society of Echocardiography.10 ### Biomarker testing Blood (15 mL) was collected immediately before angiography and after the completion of angiographic procedure(s) through a centrally placed vascular access sheath. Samples were immediately centrifuged for 15 min, and plasma samples were then aliquoted on ice and frozen in a −80°C refrigerator until analysis. After a single freeze–thaw cycle, 200 µL of plasma was analysed using the Luminex 100/200 xMAP technology platform (Luminex, Austin, Texas), which uses multiplexed, microsphere-based assays in a single reaction vessel. Multiplexing was accomplished by assigning each protein-specific assay a microsphere set labelled with a unique fluorescence signature. An assay-specific capture antibody was conjugated covalently to each unique set of microspheres and bound to the protein of interest. Assay-specific, biotinylated detecting antibodies were added, followed by a streptavidin-labelled fluorescent ‘reporter’ molecule. We specifically assayed 109 proteins (online supplementary table 1) using a commercially available kit, known as the Myriad RBM MAP. The panel incorporates biomarkers that reflect a wide variety of pathways associated with plaque rupture/erosion and includes acute phase reactants, inflammatory markers and biomarkers of atherosclerosis. ### Supplementary data [[openhrt-2018-000916-supp1.docx]](pending:yes) ### Statistical analyses For the purposes of this analysis, subjects were considered to have severe AS if a transthoracic echocardiogram within 6 months of enrolment demonstrated severe AS, defined by an aortic valve area (AVA) <1.0 cm2. Since AVA is consistently measured in patients with AS, patients missing AVA measurements were assumed to be negative. Baseline characteristics in those with and without severe AS were compared using Fisher’s exact test for categorical variables, two-sided two-sample t-test for normally distributed continuous variables, and Wilcoxon rank-sum test for continuous variables that were not normally distributed. Protein concentrations were log-transformed to achieve a normal distribution, and to facilitate the analysis they were rescaled to a zero-mean, unit-variance distribution, and outliers (ie, values beyond 3× median absolute deviation) were Winsorised. The case was conducted as a complete case analysis, and one patient was excluded from the analysis for missing data. Candidate panels of proteins and clinical features were generated via least-angle regression (LARS).8 In this method, factors were included in the model one at a time, with their coefficients determined by their correlation with the outcome. This was repeated until all factors were included in the model, and the step at which the performance plateaued resulted in our initial panel of interest. With this panel of interest, predictive analyses were run on the training set using least absolute shrinkage and selection operator (LASSO) with logistic regression, predicting the outcome of severe AS using only the variables in the panel of interest. This model development process was done via Monte Carlo cross-validation, using 400 iterations with an 80:20 (training:test) split. The final panel was used to create a final model with the entire sample, and this model was then evaluated to predict severe AS. Discrimination of the final model was assessed by calculating the area under the receiver operating curve (AUC), and calibration was determined using Akaike or Bayesian information criteria (AIC and BIC, respectively) and the Hosmer-Lemeshow goodness of fit test. The score of the final model was grouped into quintiles to create a five-level clinical risk score. All statistics were performed using R V.3.3 software (R Foundation for Statistical Computing, Vienna, Austria); p values are two-sided, with a value <0.05 considered significant. ## Results ### Patient population We enrolled 1251 subjects who underwent coronary and peripheral angiography with and without intervention between 2008 and 2011. Overall, 981 subjects underwent coronary angiography, 155 peripheral angiography, and 115 both coronary and peripheral angiography. Of these, 1244 had available blood samples and clinical data for analysis. Subjects were on average 66±11 years old and 28.8% (358 of 1244) were female. Table 1 details the characteristics of those subjects with (n=80; 6.4%) and without severe AS; of those without severe AS, moderate stenosis was present in 2.0% (26 of 1244). Sex and many comorbid conditions were similar between those with and without severe AS. However, compared with those without, subjects with severe AS were older (74±11 vs 66±11 years) and more often afflicted with atrial fibrillation/flutter (28.8 vs 18.0%; p=0.03). Patients with severe AS were less likely than those without severe AS to have undergone a prior stent (18.8 vs 32.0%; p=0.01) and were less often receiving a beta-blocker (51.9 vs 72.0%; p<0.001) or antiplatelet agent (aspirin: 65.8 vs 78.5%; p=0.01; clopidogrel: 12.7 vs 25.1%; p=0.01). Loop diuretics were more often prescribed with severe AS (31.7 vs 20.6%; p=0.03). View this table: [Table 1](http://openheart.bmj.com/content/5/2/e000916/T1) Table 1 Baseline patient characteristics ### Model for identification of severe AS LARS was used to identify independent predictors of severe AS. These factors included age and concentrations of three biomarkers: N-terminal pro-B-type natriuretic peptide (NT-proBNP), von Willebrand factor (vWF) and fetuin-A. Concentrations of NT-proBNP demonstrated direct correlation with AS, with a progressive increase in circulating NT-proBNP concentrations with worsening AS severity (figure 1; p<0.001, Kruskal-Wallis rank-sum test), while concentrations of circulating fetuin-A progressively decreased with worse AS (p=0.003). vWF concentrations did not clearly correlate with AS severity (p=0.19), although the direction of vWF’s effect in the final model was negative, with lower concentrations associated with severe AS. Using Monte Carlo cross-validation, this panel achieved a cross-validated AUC of 0.74. ![Figure 1](http://openheart.bmj.com/https://openheart.bmj.com/content/openhrt/5/2/e000916/F1.medium.gif) [Figure 1](http://openheart.bmj.com/content/5/2/e000916/F1) Figure 1 Distribution of NT-proBNP, von Willebrand factor and fetuin-A in patients with and without aortic stenosis. Levels of circulating biomarkers are shown in patients without significant AS, moderate AS and severe AS. Dark lines indicate median values and grey bar the IQR. Error bars indicate the range of the rest of the distribution to 1.5 times the IQR. AS, aortic stenosis; NT-proBNP, N-terminal pro-B-type natriuretic peptide. A final combined model of age, NT-proBNP, vWF and fetuin-A was created using the entire population with LASSO with logistic regression. This model was strongly predictive of severe AS (OR=5.9, 95% CI 3.5 to 10.1, p<0.001). To examine the model’s performance and the relative contribution of each parameter, we assessed discrimination, calibration and Hosmer-Lemeshow goodness of fit for identification of severe AS. Addition of biomarkers to age resulted in improved AUC of the receiver operating characteristic curve, minimisation of AIC and BIC, and with non-significant Hosmer-Lemeshow p values (table 2). This final model demonstrated discrimination with an insample AUC of 0.76 (p<0.001). View this table: [Table 2](http://openheart.bmj.com/content/5/2/e000916/T2) Table 2 Model fit characteristics We examined the distribution of the final model’s diagnostic score for each patient with their status for severe AS. A clinical risk score was derived from the score generated by the model, with the five quintiles of the score corresponding to a five-level risk score. Higher prevalence of severe AS was noted in those with higher scores (figure 2), such that 1.6% of those with a risk score of 1 (lowest risk) had severe AS compared with 15.3% with a risk score of 5 (highest risk) (p<0.001). When modelled as a continuous measure, higher AS scores were directly correlated with the proportion of subjects with severe AS (p<0.001) and inversely correlated with AVA (r=−0.35; p<0.001). ![Figure 2](http://openheart.bmj.com/https://openheart.bmj.com/content/openhrt/5/2/e000916/F2.medium.gif) [Figure 2](http://openheart.bmj.com/content/5/2/e000916/F2) Figure 2 Prevalence of severe aortic stenosis (AS) within a quintile-based five-level diagnostic score. Increasing prevalence of severe AS is noted at higher scores, such that 1.6% of those with a score of 1 (lowest risk) had severe AS compared with 15.3% with a score of 5 (highest risk) (p<0.001). Receiver operator characteristic analyses were performed and operating characteristics calculated (figure 3). Using a score cut-off determined by the optimal Youden’s Index, we found 76% sensitivity, 65% specificity, 13% positive predictive value and 98% negative predictive value. Likelihood ratio for positive results (LR+) was 2.17 and 0.37 for negative results (LR−). ![Figure 3](http://openheart.bmj.com/https://openheart.bmj.com/content/openhrt/5/2/e000916/F3.medium.gif) [Figure 3](http://openheart.bmj.com/content/5/2/e000916/F3) Figure 3 Receiver operating characteristic curve for aortic stenosis (AS) score. The AS score demonstrated good discrimination with an insample area under the curve (AUC) of 0.76. The sensitivity, specificity, positive predictive value and negative predictive value at the optimal cut-off are depicted. We evaluated the levels of circulating biomarkers and the AS score within subtypes of severe AS (table 3). We found marked differences in the levels of NT-proBNP across AS subtype, with the highest levels noted in patients with low LVEF, low aortic valve gradient (AVG) AS, and the lowest levels in those patients with AS with preserved LVEF and low AVG (p<0.001). A similar although non-significant pattern of abundance was noted for circulating vWF (p=0.055). Fetuin-A was highest in the preserved LVEF and low AVG group and lowest in those with preserved LVEF and high AVG (p=0.01). The AS risk score did not differ between AS subtypes. View this table: [Table 3](http://openheart.bmj.com/content/5/2/e000916/T3) Table 3 Biomarkers in subtypes of aortic stenosis ## Discussion The identification of AS may be challenging to recognise and grade. Furthermore, broad application of tools to assess presence or severity of AS such as echocardiography—although useful—may not be practical in some populations. As a consequence, a substantial proportion of valvular heart disease is not diagnosed within the general population.11 Availability of a non-invasive, easily interpreted and cost-effective tool to screen for the presence of AS is therefore of clinical value. Using patients undergoing coronary and peripheral angiography within the CASABLANCA study, we developed and evaluated a robust, non-invasive, clinical plus multimarker approach for the identification of severe AS. This model, which includes age and concentrations of NT-proBNP, vWF and fetuin-A, was adapted into a clinically applicable score, which performed well within the CASABLANCA study cohort with strong sensitivity, specificity and negative predictive value. The score may therefore add a novel tool to our clinical armamentarium for the identification of patients with severe AS, in particular for excluding its presence in a patient who might otherwise be suspected as suffering from the diagnosis. AS is an indolent disease that culminates in clinical decompensation and necessitates valve replacement, without which 1-year mortality is exceedingly high.2 In fact, mortality due to untreated symptomatic severe AS exceeds that associated with several advanced cancer diagnoses. While survival is thought to improve on valve replacement, recent evidence suggests persistently elevated morbidity and mortality risk in patients treated at a late disease state. Adverse outcomes are in part driven by irreversible maladaptive LV remodelling and fibrosis.4–6 In addition, frailty, sarcopaenia and poor functional status worsen with time and contribute to diminished survival and recovery after AVR.12 13 Timely identification of severe AS is therefore of critical importance to the maintenance of health in the vulnerable elderly population. A detailed physical examination with cardiac auscultation is often relied on to screen for valve lesions, including severe AS. When cardiac auscultation reveals an abnormal heart murmur, an echocardiogram is often performed to confirm or exclude a pathological cardiac lesion.3 Unfortunately, limited proficiency with cardiac auscultation among practitioners contributes to the underdiagnosis of AS.14 15 Furthermore, even with comprehensive echocardiography, the gold-standard diagnostic tool, accurate identification and grading of AS severity often prove challenging due to poor imaging windows, inaccurate quantification of LV outflow tract dimensions and discordant metrics of AS severity (ie, AVA, mean gradient and peak velocity).16 17 Inconsistencies regarding the evaluation of AS severity occur due to measurement error and due to the disruption of the physiological relationship between AV area and gradient that occurs as a consequence of diminished transvalvular flow.3 18 19 Together, these factors contribute to the frequent late recognition of severe AS and highlight the need for novel objective metrics that will supplement current diagnostic strategies and aid in the timely recognition of at-risk patients. While biomarkers may be of prognostic value in those with AS, our data add significant understanding regarding the role of biomarkers for the diagnosis of AS. Concentrations of circulating natriuretic peptides rise with AS disease severity due to cardiomyocyte stretch and pressure overload and have been shown to predict survival and clinical outcomes in patients with AS and after transcatheter and surgical AVR.20–22 Clinical practice guidelines articulate a possible role for elevated natriuretic peptide concentrations to inform surgical intervention in asymptomatic patients23; reliance on NT-proBNP has not been universally endorsed, however.3 For example, while NT-proBNP may be elevated in those with AS, LV hypertrophy due to AS may normalise wall stress and therefore attenuate the anticipated rise in the biomarker. Because there is also considerable overlap in natriuretic peptide concentrations across elderly patients with and without AS,24 a strategy of integrating NT-proBNP with orthogonal biomarkers may inform clinical practice better than NT-proBNP alone. Several studies have demonstrated abnormalities in vWF high-molecular-weight multimers in subjects with severe AS.25 26 As blood flows through a severely stenotic valve, shear forces unfold these multimers, resulting in increased susceptibility to proteolytic cleavage.25 The resultant haematological deficiencies form the clinical basis for the acquired type 2A von Willebrand syndrome and gastrointestinal angiodysplasia (Heyde’s syndrome), often noted with severe AS,25 which may resolve with alleviation of AVG.26 Association between circulating vWF concentrations in those with and without severe AS were less direct than NT-proBNP; however, vWF concentrations were selected by LARS during the model development process, and their presence improved calibration of the AS model, supporting the adoption of this biomarker into a multimarker approach for AS discrimination. Fetuin-A is a liver-derived protein that binds and solubilises calcium and phosphate and in turn regulates soft tissue mineralisation. Reduced circulating fetuin-A concentrations have been implicated as a mediator of calcific cardiovascular disease seen in patients with end-stage renal disease and valve disease.27 As shown here, a recent analysis suggested that patients with AS have lower circulating fetuin-A than control subjects.28 Much as with NT-proBNP and vWF, fetuin-A concentrations were selected by LARS to remain in the final aggregate multiple biomarker approach for AS diagnosis, and the presence of this protein in the model improved model discrimination while maintaining equivalent calibration. While the size of the CASABLANCA cohort, the large number of proteins assayed, and the unbiased and methodical statistical approach are strengths of the current study, several limitations warrant attention. First, the CASABLANCA study enrolled subjects undergoing invasive cardiovascular angiography, irrespective of the clinical indication. The cohort is therefore diverse with low prevalence of severe AS. We anticipate that the clinical application of the AS score in select patients with higher pretest probability of having severe AS may result in improved model performance. Second, the AS score possessed low positive predictive value, a consequence of the low prevalence of severe AS within our population. Such performance characteristics are commonly noted for screening tests applied to the general population.29 Third, subjects with severe AS undergoing angiography were largely symptomatic patients being evaluated for AVR. We were consequently not able to examine the interplay between the AS score and clinical symptomatology. Fourth, the relatively small number of AS cases limited the statistical power of our attempts to evaluate the value of the AS score in discriminating the degree of AS severity or AS subtype (preserved LVEF high gradient vs paradoxical preserved LVEF low gradient vs low LVEF low gradient AS). Future efforts will seek to externally validate the AS score with larger patient cohorts and to determine the value of the AS score in aiding the grading of AS severity. In conclusion, we have developed a non-invasive, biomarker-supported strategy for screening for severe AS. This approach includes age and three protein biomarkers indicative of distinct biological pathways previously implicated in the pathophysiology of AS. The model we describe could theoretically be a cost-effective addition to the clinical armamentarium for evaluating those with suspected AS, facilitating the identification of this high-risk patient cohort and fostering timely valve replacement. Further efforts are needed to examine the utility of incorporating the predictive model into clinical practice. ## Footnotes * Contributors Each author has made substantial contributions to the manuscript that warrant authorship. * Funding This work was funded by grants from Prevencio and from Siemens Diagnostics (to the CASABLANCA study), the American Heart Association (14 FTF20440012 to SE), the Dennis and Marilyn Barry Fellowship in Cardiology (to NI), and the Hutter Family Professorship in Cardiology (to JLJ). * Competing interests SE receives institutional research support from Siemens and Boehringer Ingelheim Pharmaceuticals and consulting fees from Medtronic and Edwards Lifesciences. The remaining authors have no relationships to disclose. JLJ has received grant support from Abbott Diagnostics, Roche Diagnostics, Singulex and Prevencio; consulting income from Roche Diagnostics, Critical Diagnostics, Philips and Novartis; and participates in clinical endpoint committees/data safety monitoring boards for Novartis, Amgen, AbbVie, Pfizer, Janssen and Boehringer Ingelheim. CM is a consultant to Prevencio. RR and GB are employees of Prevencio. Siemens Diagnostics had no involvement in the present study design, collection, analysis and interpretation of data; in the writing of the report; and in the decision to submit the article for publication. All other authors have reported that they have no relationships relevant to the content of this paper to disclose. * Patient consent Patient consent obtained. * Ethics approval Study procedures were approved by the Partners Healthcare Institutional Review Board and carried out in accordance with the Declaration of Helsinki. * Provenance and peer review Not commissioned; externally peer reviewed. * Data sharing statement No additional data are available. * Received August 11, 2018. * Revision received October 10, 2018. * Accepted October 12, 2018. * © Author(s) (or their employer(s)) 2018. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: [http://creativecommons.org/licenses/by-nc/4.0/](http://creativecommons.org/licenses/by-nc/4.0/) ## References 1. 1. Osnabrugge RL , Mylotte D , Head SJ , et al . Aortic stenosis in the elderly: disease prevalence and number of candidates for transcatheter aortic valve replacement: a meta-analysis and modeling study. J Am Coll Cardiol 2013;62:1002–12.[doi:10.1016/j.jacc.2013.05.015](http://dx.doi.org/10.1016/j.jacc.2013.05.015) [FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo0OiJhY2NqIjtzOjU6InJlc2lkIjtzOjEwOiI2Mi8xMS8xMDAyIjtzOjQ6ImF0b20iO3M6MjU6Ii9vcGVuaHJ0LzUvMi9lMDAwOTE2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 2. 2. Leon MB , Smith CR , Mack M , et al . Transcatheter aortic-valve implantation for aortic stenosis in patients who cannot undergo surgery. N Engl J Med 2010;363:1597–607.[doi:10.1056/NEJMoa1008232](http://dx.doi.org/10.1056/NEJMoa1008232) [CrossRef](http://openheart.bmj.com/lookup/external-ref?access_num=10.1056/NEJMoa1008232&link_type=DOI) [PubMed](http://openheart.bmj.com/lookup/external-ref?access_num=20961243&link_type=MED&atom=%2Fopenhrt%2F5%2F2%2Fe000916.atom) [Web of Science](http://openheart.bmj.com/lookup/external-ref?access_num=000283242700004&link_type=ISI) 3. 3. Nishimura RA , Otto CM , Bonow RO , et al . 2014 AHA/ACC guideline for the management of patients with valvular heart disease: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 2014;63:e57–185.[doi:10.1016/j.jacc.2014.02.536](http://dx.doi.org/10.1016/j.jacc.2014.02.536) [FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo0OiJhY2NqIjtzOjU6InJlc2lkIjtzOjk6IjYzLzIyL2U1NyI7czo0OiJhdG9tIjtzOjI1OiIvb3BlbmhydC81LzIvZTAwMDkxNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 4. 4. Elmariah S , Palacios IF , McAndrew T , et al . Outcomes of transcatheter and surgical aortic valve replacement in high-risk patients with aortic stenosis and left ventricular dysfunction: results from the Placement of Aortic Transcatheter Valves (PARTNER) trial (cohort A). Circ Cardiovasc Interv 2013;6:604–14.[doi:10.1161/CIRCINTERVENTIONS.113.000650](http://dx.doi.org/10.1161/CIRCINTERVENTIONS.113.000650) [Abstract/FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToiY2lyY2N2aW50IjtzOjU6InJlc2lkIjtzOjc6IjYvNi82MDQiO3M6NDoiYXRvbSI7czoyNToiL29wZW5ocnQvNS8yL2UwMDA5MTYuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 5. 5. Lindman BR , Stewart WJ , Pibarot P , et al . Early regression of severe left ventricular hypertrophy after transcatheter aortic valve replacement is associated with decreased hospitalizations. JACC Cardiovasc Interv 2014;7:662–73.[doi:10.1016/j.jcin.2014.02.011](http://dx.doi.org/10.1016/j.jcin.2014.02.011) [Abstract/FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiamludCI7czo1OiJyZXNpZCI7czo3OiI3LzYvNjYyIjtzOjQ6ImF0b20iO3M6MjU6Ii9vcGVuaHJ0LzUvMi9lMDAwOTE2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 6. 6. Chin CWL , Everett RJ , Kwiecinski J , et al . Myocardial fibrosis and cardiac decompensation in aortic stenosis. JACC Cardiovasc Imaging 2017;10:1320–33.[doi:10.1016/j.jcmg.2016.10.007](http://dx.doi.org/10.1016/j.jcmg.2016.10.007) [Abstract/FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiamltZyI7czo1OiJyZXNpZCI7czoxMDoiMTAvMTEvMTMyMCI7czo0OiJhdG9tIjtzOjI1OiIvb3BlbmhydC81LzIvZTAwMDkxNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 7. 7. Gardezi SKM , Myerson SG , Chambers J , et al . Cardiac auscultation poorly predicts the presence of valvular heart disease in asymptomatic primary care patients. Heart 2018:heartjnl-2018-313082.[doi:10.1136/heartjnl-2018-313082](http://dx.doi.org/10.1136/heartjnl-2018-313082) 8. 8. Ibrahim NE , Januzzi JL , Magaret CA , et al . A clinical and biomarker scoring system to predict the presence of obstructive coronary artery disease. J Am Coll Cardiol 2017;69:1147–56.[doi:10.1016/j.jacc.2016.12.021](http://dx.doi.org/10.1016/j.jacc.2016.12.021) [FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo0OiJhY2NqIjtzOjU6InJlc2lkIjtzOjk6IjY5LzkvMTE0NyI7czo0OiJhdG9tIjtzOjI1OiIvb3BlbmhydC81LzIvZTAwMDkxNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 9. 9. McCarthy CP , van Kimmenade RRJ , Gaggin HK , et al . Usefulness of multiple biomarkers for predicting incident major adverse cardiac events in patients who underwent diagnostic coronary angiography (from the catheter sampled blood archive in cardiovascular diseases [CASABLANCA] study). Am J Cardiol 2017;120:25–32.[doi:10.1016/j.amjcard.2017.03.265](http://dx.doi.org/10.1016/j.amjcard.2017.03.265) 10. 10. Lang RM , Badano LP , Mor-Avi V , et al . Recommendations for cardiac chamber quantification by echocardiography in adults: an update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. J Am Soc Echocardiogr 2015;28:1–39.[doi:10.1016/j.echo.2014.10.003](http://dx.doi.org/10.1016/j.echo.2014.10.003) [CrossRef](http://openheart.bmj.com/lookup/external-ref?access_num=10.1016/j.echo.2014.10.003&link_type=DOI) [PubMed](http://openheart.bmj.com/lookup/external-ref?access_num=25559473&link_type=MED&atom=%2Fopenhrt%2F5%2F2%2Fe000916.atom) 11. 11. d'Arcy JL , Coffey S , Loudon MA , et al . Large-scale community echocardiographic screening reveals a major burden of undiagnosed valvular heart disease in older people: the OxVALVE Population Cohort Study. Eur Heart J 2016;37:3515–22.[doi:10.1093/eurheartj/ehw229](http://dx.doi.org/10.1093/eurheartj/ehw229) [CrossRef](http://openheart.bmj.com/lookup/external-ref?access_num=10.1093/eurheartj/ehw229&link_type=DOI) [PubMed](http://openheart.bmj.com/lookup/external-ref?access_num=27354049&link_type=MED&atom=%2Fopenhrt%2F5%2F2%2Fe000916.atom) 12. 12. Green P , Woglom AE , Genereux P , et al . The impact of frailty status on survival after transcatheter aortic valve replacement in older adults with severe aortic stenosis: a single-center experience. JACC Cardiovasc Interv 2012;5:974–81.[doi:10.1016/j.jcin.2012.06.011](http://dx.doi.org/10.1016/j.jcin.2012.06.011) [Abstract/FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiamludCI7czo1OiJyZXNpZCI7czo3OiI1LzkvOTc0IjtzOjQ6ImF0b20iO3M6MjU6Ii9vcGVuaHJ0LzUvMi9lMDAwOTE2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 13. 13. Afilalo J , Lauck S , Kim DH , et al . Frailty in older adults undergoing aortic valve replacement: the FRAILTY-AVR study. J Am Coll Cardiol 2017;70:689–700.[doi:10.1016/j.jacc.2017.06.024](http://dx.doi.org/10.1016/j.jacc.2017.06.024) [FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo0OiJhY2NqIjtzOjU6InJlc2lkIjtzOjg6IjcwLzYvNjg5IjtzOjQ6ImF0b20iO3M6MjU6Ii9vcGVuaHJ0LzUvMi9lMDAwOTE2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 14. 14. Mangione S , Nieman LZ , Gracely E , et al . The teaching and practice of cardiac auscultation during internal medicine and cardiology training. A nationwide survey. Ann Intern Med 1993;119:47–54.[doi:10.7326/0003-4819-119-1-199307010-00009](http://dx.doi.org/10.7326/0003-4819-119-1-199307010-00009) [CrossRef](http://openheart.bmj.com/lookup/external-ref?access_num=10.7326/0003-4819-119-1-199307010-00009&link_type=DOI) [PubMed](http://openheart.bmj.com/lookup/external-ref?access_num=8498764&link_type=MED&atom=%2Fopenhrt%2F5%2F2%2Fe000916.atom) [Web of Science](http://openheart.bmj.com/lookup/external-ref?access_num=A1993LJ26600009&link_type=ISI) 15. 15. Gaibazzi N , Reverberi C , Ghillani M , et al . Prevalence of undiagnosed asymptomatic aortic valve stenosis in the general population older than 65 years. a screening strategy using cardiac auscultation followed by Doppler-echocardiography. Int J Cardiol 2013;168:4905–6.[doi:10.1016/j.ijcard.2013.07.100](http://dx.doi.org/10.1016/j.ijcard.2013.07.100) 16. 16. Saikrishnan N , Kumar G , Sawaya FJ , et al . Accurate assessment of aortic stenosis: a review of diagnostic modalities and hemodynamics. Circulation 2014;129:244–53.[doi:10.1161/CIRCULATIONAHA.113.002310](http://dx.doi.org/10.1161/CIRCULATIONAHA.113.002310) [FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjk6IjEyOS8yLzI0NCI7czo0OiJhdG9tIjtzOjI1OiIvb3BlbmhydC81LzIvZTAwMDkxNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 17. 17. Michelena HI , Margaryan E , Miller FA , et al . Inconsistent echocardiographic grading of aortic stenosis: is the left ventricular outflow tract important? Heart 2013;99:921–31.[doi:10.1136/heartjnl-2012-302881](http://dx.doi.org/10.1136/heartjnl-2012-302881) [Abstract/FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiaGVhcnRqbmwiO3M6NToicmVzaWQiO3M6OToiOTkvMTMvOTIxIjtzOjQ6ImF0b20iO3M6MjU6Ii9vcGVuaHJ0LzUvMi9lMDAwOTE2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 18. 18. Clavel MA , Messika-Zeitoun D , Pibarot P , et al . The complex nature of discordant severe calcified aortic valve disease grading: new insights from combined Doppler echocardiographic and computed tomographic study. J Am Coll Cardiol 2013;62:2329–38.[doi:10.1016/j.jacc.2013.08.1621](http://dx.doi.org/10.1016/j.jacc.2013.08.1621) [FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo0OiJhY2NqIjtzOjU6InJlc2lkIjtzOjEwOiI2Mi8yNC8yMzI5IjtzOjQ6ImF0b20iO3M6MjU6Ii9vcGVuaHJ0LzUvMi9lMDAwOTE2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 19. 19. Hachicha Z , Dumesnil JG , Bogaty P , et al . Paradoxical low-flow, low-gradient severe aortic stenosis despite preserved ejection fraction is associated with higher afterload and reduced survival. Circulation 2007;115:2856–64.[doi:10.1161/CIRCULATIONAHA.106.668681](http://dx.doi.org/10.1161/CIRCULATIONAHA.106.668681) [Abstract/FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjExOiIxMTUvMjIvMjg1NiI7czo0OiJhdG9tIjtzOjI1OiIvb3BlbmhydC81LzIvZTAwMDkxNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 20. 20. Bergler-Klein J , Klaar U , Heger M , et al . Natriuretic peptides predict symptom-free survival and postoperative outcome in severe aortic stenosis. Circulation 2004;109:2302–8.[doi:10.1161/01.CIR.0000126825.50903.18](http://dx.doi.org/10.1161/01.CIR.0000126825.50903.18) [Abstract/FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjExOiIxMDkvMTkvMjMwMiI7czo0OiJhdG9tIjtzOjI1OiIvb3BlbmhydC81LzIvZTAwMDkxNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 21. 21. Clavel MA , Malouf J , Michelena HI , et al . B-type natriuretic peptide clinical activation in aortic stenosis: impact on long-term survival. J Am Coll Cardiol 2014;63:2016–25.[doi:10.1016/j.jacc.2014.02.581](http://dx.doi.org/10.1016/j.jacc.2014.02.581) [FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo0OiJhY2NqIjtzOjU6InJlc2lkIjtzOjEwOiI2My8xOS8yMDE2IjtzOjQ6ImF0b20iO3M6MjU6Ii9vcGVuaHJ0LzUvMi9lMDAwOTE2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 22. 22. Lindman BR , Breyley JG , Schilling JD , et al . Prognostic utility of novel biomarkers of cardiovascular stress in patients with aortic stenosis undergoing valve replacement. Heart 2015;101:1382–8.[doi:10.1136/heartjnl-2015-307742](http://dx.doi.org/10.1136/heartjnl-2015-307742) [Abstract/FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiaGVhcnRqbmwiO3M6NToicmVzaWQiO3M6MTE6IjEwMS8xNy8xMzgyIjtzOjQ6ImF0b20iO3M6MjU6Ii9vcGVuaHJ0LzUvMi9lMDAwOTE2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 23. 23. Baumgartner H , Falk V , Bax JJ , et al . 2017 ESC/EACTS Guidelines for the management of valvular heart disease. Eur Heart J 2017;38:2739–91.[doi:10.1093/eurheartj/ehx391](http://dx.doi.org/10.1093/eurheartj/ehx391) [CrossRef](http://openheart.bmj.com/lookup/external-ref?access_num=10.1093/eurheartj/ehx391&link_type=DOI) [PubMed](http://openheart.bmj.com/lookup/external-ref?access_num=28886619&link_type=MED&atom=%2Fopenhrt%2F5%2F2%2Fe000916.atom) 24. 24. Gardezi SK , Coffey S , Prendergast BD , et al . Serum biomarkers in valvular heart disease. Heart 2018;104:349–58.[doi:10.1136/heartjnl-2016-310482](http://dx.doi.org/10.1136/heartjnl-2016-310482) [FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiaGVhcnRqbmwiO3M6NToicmVzaWQiO3M6OToiMTA0LzQvMzQ5IjtzOjQ6ImF0b20iO3M6MjU6Ii9vcGVuaHJ0LzUvMi9lMDAwOTE2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 25. 25. Vincentelli A , Susen S , Le Tourneau T , et al . Acquired von Willebrand syndrome in aortic stenosis. N Engl J Med 2003;349:343–9.[doi:10.1056/NEJMoa022831](http://dx.doi.org/10.1056/NEJMoa022831) [CrossRef](http://openheart.bmj.com/lookup/external-ref?access_num=10.1056/NEJMoa022831&link_type=DOI) [PubMed](http://openheart.bmj.com/lookup/external-ref?access_num=12878741&link_type=MED&atom=%2Fopenhrt%2F5%2F2%2Fe000916.atom) [Web of Science](http://openheart.bmj.com/lookup/external-ref?access_num=000184312800007&link_type=ISI) 26. 26. Bander J , Elmariah S , Aledort LM , et al . Changes in von Willebrand factor-cleaving protease (ADAMTS-13) in patients with aortic stenosis undergoing valve replacement or balloon valvuloplasty. Thromb Haemost 2012;108:86–93.[doi:10.1160/TH11-12-0803](http://dx.doi.org/10.1160/TH11-12-0803) [CrossRef](http://openheart.bmj.com/lookup/external-ref?access_num=10.1160/TH11-12-0803&link_type=DOI) [PubMed](http://openheart.bmj.com/lookup/external-ref?access_num=22627470&link_type=MED&atom=%2Fopenhrt%2F5%2F2%2Fe000916.atom) 27. 27. Ix JH , Chertow GM , Shlipak MG , et al . Association of fetuin-A with mitral annular calcification and aortic stenosis among persons with coronary heart disease: data from the Heart and Soul Study. Circulation 2007;115:2533–9.[doi:10.1161/CIRCULATIONAHA.106.682450](http://dx.doi.org/10.1161/CIRCULATIONAHA.106.682450) [Abstract/FREE Full Text](http://openheart.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjExOiIxMTUvMTkvMjUzMyI7czo0OiJhdG9tIjtzOjI1OiIvb3BlbmhydC81LzIvZTAwMDkxNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 28. 28. Di Minno A , Zanobini M , Myasoedova VA , et al . Could circulating fetuin a be a biomarker of aortic valve stenosis? Int J Cardiol 2017;249:426–30.[doi:10.1016/j.ijcard.2017.05.040](http://dx.doi.org/10.1016/j.ijcard.2017.05.040) 29. 29. Imperiale TF , Ransohoff DF , Itzkowitz SH , et al . Multitarget stool DNA testing for colorectal-cancer screening. N Engl J Med 2014;370:1287–97.[doi:10.1056/NEJMoa1311194](http://dx.doi.org/10.1056/NEJMoa1311194) [CrossRef](http://openheart.bmj.com/lookup/external-ref?access_num=10.1056/NEJMoa1311194&link_type=DOI) [PubMed](http://openheart.bmj.com/lookup/external-ref?access_num=24645800&link_type=MED&atom=%2Fopenhrt%2F5%2F2%2Fe000916.atom) [Web of Science](http://openheart.bmj.com/lookup/external-ref?access_num=000333830000007&link_type=ISI)