Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Primer
  • Published:

Calcific aortic stenosis

Abstract

Calcific aortic stenosis (AS) is the most prevalent heart valve disorder in developed countries. It is characterized by progressive fibro-calcific remodelling and thickening of the aortic valve leaflets that, over years, evolve to cause severe obstruction to cardiac outflow. In developed countries, AS is the third-most frequent cardiovascular disease after coronary artery disease and systemic arterial hypertension, with a prevalence of 0.4% in the general population and 1.7% in the population >65 years old. Congenital abnormality (bicuspid valve) and older age are powerful risk factors for calcific AS. Metabolic syndrome and an elevated plasma level of lipoprotein(a) have also been associated with increased risk of calcific AS. The pathobiology of calcific AS is complex and involves genetic factors, lipoprotein deposition and oxidation, chronic inflammation, osteoblastic transition of cardiac valve interstitial cells and active leaflet calcification. Although no pharmacotherapy has proved to be effective in reducing the progression of AS, promising therapeutic targets include lipoprotein(a), the renin–angiotensin system, receptor activator of NF-κB ligand (RANKL; also known as TNFSF11) and ectonucleotidases. Currently, aortic valve replacement (AVR) remains the only effective treatment for severe AS. The diagnosis and staging of AS are based on the assessment of stenosis severity and left ventricular systolic function by Doppler echocardiography, and the presence of symptoms. The introduction of transcatheter AVR in the past decade has been a transformative therapeutic innovation for patients at high or prohibitive risk for surgical valve replacement, and this new technology might extend to lower-risk patients in the near future.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The prevalence of AS as a function of age.
Figure 2: Comparison of tricuspid and bicuspid aortic valve structures.
Figure 3: Macroscopic and histopathological appearance of normal and abnormal aortic valves.
Figure 4: Pathogenesis of calcific AS.
Figure 5: Maladaptive remodelling and impaired function of the left ventricle in response to pressure overload from AS.
Figure 6: Patterns of left ventricular remodelling.
Figure 7: Assessment of AS severity by Doppler echocardiography.
Figure 8: Assessment of aortic valve mineralization activity by PET-CT.
Figure 9: Assessment of flow patterns in the aorta by 4D flow cardiac magnetic resonance according to aortic valve phenotype.
Figure 10: Assessment of myocardial fibrosis by cardiac magnetic resonance in patients with AS.
Figure 11: Algorithm for the management of AS.
Figure 12: Different types of surgical aortic valve replacement.
Figure 13: Different types of transcatheter aortic valve replacement.

Similar content being viewed by others

References

  1. Rajamannan, N. M. et al. Calcific aortic valve disease: not simply a degenerative process: a review and agenda for research from the national heart and lung and blood institute aortic stenosis working group executive summary: calcific aortic valve disease — 2011 update. Circulation 124, 1783–1791 (2011). A good review of the state of knowledge and future research directions for calcific aortic valve disease (in 2011).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Go, A. S. et al. Executive summary: heart disease and stroke statistics — 2013 update: a report from the American Heart Association. Circulation 127, 143–152 (2013).

    Article  PubMed  Google Scholar 

  3. Nishimura, R. A. 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. 63, e57–e185 (2014). The most recent version of the clinical guidelines for the management of valvular heart disease, including AS.

    Article  PubMed  Google Scholar 

  4. Vahanian, A. et al. Guidelines on the management of valvular heart disease Joint Task Force on the management of valvular heart disease of the European Society of Cardiology (ESC); European Association for Cardio-Thoracic Surgery (EACTS). Eur. Heart J. 33, 2451–2496 (2012).

    Article  PubMed  Google Scholar 

  5. Roberts, W. C. & Ko, J. M. Frequency by decades of unicuspid, bicuspid, and tricuspid aortic valves in adults having isolated aortic valve replacement for aortic stenosis, with or without associated aortic regurgitation. Circulation 111, 920–925 (2005).

    Article  PubMed  Google Scholar 

  6. Bosse, Y., Mathieu, P. & Pibarot, P. Genomics: the next step to elucidate the etiology of calcific aortic valve stenosis. J. Am. Coll. Cardiol. 51, 1327–1336 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Michelena, H. I. et al. Incidence of aortic complications in patients with bicuspid aortic valves. JAMA 306, 1104–1112 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Michelena, H. I. et al. Bicuspid aortic valve: dentifying knowledge gaps and rising to the challenge from the international bicuspid aortic valve consortium (BAVCon). Circulation 129, 2691–2704 (2014). A review of the diagnosis, complications and management of bicuspid aortic valve syndrome.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Stewart, B. F. et al. Clinical factors associated with calcific aortic valve disease. Cardiovascular Health Study. J. Am. Coll. Cardiol. 29, 630–634 (1997).

    Article  CAS  PubMed  Google Scholar 

  10. Cosmi, J. E. et al. The risk of the development of aortic stenosis in patients with “benign” aortic valve thickening. Arch. Intern. Med. 162, 2345–2347 (2002).

    Article  PubMed  Google Scholar 

  11. Coffey, S., Cox, B. & Williams, M. J. The prevalence, incidence, progression, and risks of aortic valve sclerosis: a systematic review and meta-analysis. J. Am. Coll. Cardiol. 63, 2852–2861 (2014).

    Article  PubMed  Google Scholar 

  12. Nkomo, V. T. et al. Burden of valvular heart diseases: a population-based study. Lancet 368, 1005–1011 (2006). An epidemiology study reporting the prevalence of valvular heart disease, including calcific AS, in the general population.

    Article  PubMed  Google Scholar 

  13. Eveborn, G. W., Schirmer, H., Heggelund, G., Lunde, P. & Rasmussen, K. The evolving epidemiology of valvular aortic stenosis. The Tromso Study. Heart 99, 396–400 (2013).

    Article  PubMed  Google Scholar 

  14. Lindroos, M., Kupari, M., Heikkila, J. & Tilvis, R. Prevalence of aortic valve abnormalities in the elderly: an echocardiographic study of a random population sample. J. Am. Coll. Cardiol. 21, 1220–1225 (1993).

    Article  CAS  PubMed  Google Scholar 

  15. Danielsen, R., Aspelund, T., Harris, T. B. & 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. 176, 916–922 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Osnabrugge, R. L. 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. 62, 1002–1012 (2013).

    Article  PubMed  Google Scholar 

  17. Probst, V. et al. Familial aggregation of calcific aortic valve stenosis in the western part of France. Circulation 113, 856–860 (2006).

    Article  PubMed  Google Scholar 

  18. Iung, B. et al. A prospective survey of patients with valvular heart disease in Europe: the Euro Heart Survey on valvular heart disease. Eur. Heart J. 24, 1231–1243 (2003). A large European survey describing the clinical management of valvular heart disease and adherence to guidelines in real-life clinical practice.

    Article  PubMed  Google Scholar 

  19. Iung, B. & Vahanian, A. Degenerative calcific aortic stenosis: a natural history. Heart 98 (Suppl. 4), iv7–iv13 (2012).

    Article  PubMed  Google Scholar 

  20. Demirbag, R., Sade, L. E., Aydin, M., Bozkurt, A. & Acarturk, E. The Turkish registry of heart valve disease. Turk Kardiyol. Dern. Ars. 41, 1–10 (2013).

    Article  PubMed  Google Scholar 

  21. Sliwa, K. et al. Incidence and characteristics of newly diagnosed rheumatic heart disease in urban African adults: insights from the heart of Soweto study. Eur. Heart J. 31, 719–727 (2010).

    Article  PubMed  Google Scholar 

  22. Freeman, R. V. & Otto, C. M. Spectrum of calcific aortic valve disease: pathogenesis, disease progression, and treatment strategies. Circulation 111, 3316–3326 (2005).

    Article  PubMed  Google Scholar 

  23. Rajamannan, N. M., Bonow, R. O. & Rahimtoola, S. H. Calcific aortic stenosis: an update. Nat. Clin. Pract. Cardiovasc. Med. 4, 254–262 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Chen, J. H. & Simmons, C. A. Cell–matrix interactions in the pathobiology of calcific aortic valve disease: critical roles for matricellular, matricrine, and matrix mechanics cues. Circ. Res. 108, 1510–1524 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Schoen, F. J. Evolving concepts of cardiac valve dynamics: the continuum of development, functional structure, pathobiology, and tissue engineering. Circulation 118, 1864–1880 (2008).

    Article  PubMed  Google Scholar 

  26. Taylor, P. M., Allen, S. P. & Yacoub, M. H. Phenotypic and functional characterization of interstitial cells from human heart valves, pericardium and skin. J. Heart Valve Dis. 9, 150–158 (2000).

    CAS  PubMed  Google Scholar 

  27. Latif, N., Sarathchandra, P., Chester, A. H. & Yacoub, M. H. Expression of smooth muscle cell markers and co-activators in calcified aortic valves. Eur. Heart J. 36, 1335–1345 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Steiner, I., Kasparova, P., Kohout, A. & Dominik, J. Bone formation in cardiac valves: a histopathological study of 128 cases. Virchows Arch. 450, 653–657 (2007).

    Article  PubMed  Google Scholar 

  29. Cote, N. et al. Inflammation is associated with the remodeling of calcific aortic valve disease. Inflammation 36, 573–581 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Helske, S. et al. Possible role for mast cell-derived cathepsin G in the adverse remodelling of stenotic aortic valves. Eur. Heart J. 27, 1495–1504 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. Mathieu, P. & Boulanger, M. C. Basic mechanisms of calcific aortic valve disease. Can. J. Cardiol. 30, 982–993 (2014).

    Article  PubMed  Google Scholar 

  32. Price, P. A., Toroian, D. & Chan, W. S. Tissue-nonspecific alkaline phosphatase is required for the calcification of collagen in serum: a possible mechanism for biomineralization. J. Biol. Chem. 284, 4594–4604 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Price, J. et al. Prosthesis–patient mismatch is less frequent and more clinically indolent in patients operated for aortic insufficiency. J. Thorac Cardiovasc. Surg. 138, 639–645 (2009).

    Article  PubMed  Google Scholar 

  34. Rattazzi, M. et al. Extracellular pyrophosphate is reduced in aortic interstitial valve cells acquiring a calcifying profile: implications for aortic valve calcification. Atherosclerosis 237, 568–576 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Hinton, R. B. et al. Extracellular matrix remodeling and organization in developing and diseased aortic valves. Circ. Res. 98, 1431–1438 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Satta, J. et al. Progression of human aortic valve stenosis is associated with tenascin-C expression. J. Am. Coll. Cardiol. 39, 96–101 (2002).

    Article  CAS  PubMed  Google Scholar 

  37. Pawade, T. A., Newby, D. E. & Dweck, M. R. Calcification in aortic stenosis: the skeleton key. J. Am. Coll. Cardiol. 66, 561–577 (2015).

    Article  PubMed  Google Scholar 

  38. Abdelbaky, A. et al. Early aortic valve inflammation precedes calcification: a longitudinal FDG-PET/CT study. Atherosclerosis 238, 165–172 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. O'Brien, K. D. et al. Apolipoproteins B, (a), and E accumulate in the morphologically early lesion of ‘degenerative’ valvular aortic stenosis. Arterioscler. Thromb. Vasc. Biol. 16, 523–532 (1996).

    Article  CAS  PubMed  Google Scholar 

  40. Mohty, D. et al. Association between plasma LDL particle size, valvular accumulation of oxidized LDL, and inflammation in patients with aortic stenosis. Arterioscler. Thromb. Vasc. Biol. 28, 187–193 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Olsson, M., Thyberg, J. & Nilsson, J. Presence of oxidized low density lipoprotein in nonrheumatic stenotic aortic valves. Arterioscler. Thromb. Vasc. Biol. 19, 1218–1222 (1999).

    Article  CAS  PubMed  Google Scholar 

  42. Cote, C. et al. Association between circulating oxidised low-density lipoprotein and fibrocalcific remodelling of the aortic valve in aortic stenosis. Heart 94, 1175–1180 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Miller, J. D. et al. Dysregulation of antioxidant mechanisms contributes to increased oxidative stress in calcific aortic valvular stenosis in humans. J. Am. Coll. Cardiol. 52, 843–850 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Liberman, M. et al. Oxidant generation predominates around calcifying foci and enhances progression of aortic valve calcification. Arterioscler. Thromb. Vasc. Biol. 28, 463–470 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Parhami, F. et al. Lipid oxidation products have opposite effects on calcifying vascular cell and bone cell differentiation. A possible explanation for the paradox of arterial calcification in osteoporotic patients. Arterioscler. Thromb. Vasc. Biol. 17, 680–687 (1997).

    Article  CAS  PubMed  Google Scholar 

  46. Tsimikas, S. & Witztum, J. L. The role of oxidized phospholipids in mediating lipoprotein (a) atherogenicity. Curr. Opin. Lipidol. 19, 369–377 (2008).

    Article  CAS  PubMed  Google Scholar 

  47. Dube, J. B., Boffa, M. B., Hegele, R. A. & Koschinsky, M.L. Lipoprotein (a): more interesting than ever after 50 years. Curr. Opin. Lipidol. 23, 133–140 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. Thanassoulis, G. et al. Genetic associations with valvular calcification and aortic stenosis. N. Engl. J. Med. 368, 503–512 (2013). This is the first large genetic study to show an association between the LPA gene and incident AS.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kamstrup, P. R., Tybjaerg-Hansen, A. & Nordestgaard, B. G. Elevated lipoprotein(a) and risk of aortic valve stenosis in the general population. J. Am. Coll. Cardiol. 63, 470–477 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Arsenault, B. J. et al. Lipoprotein(a) levels, genotype and incident aortic valve stenosis: a prospective Mendelian randomization study and replication in a case–control cohort. Circ. Cardiovasc. Genet. 7, 304–310 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Capoulade, R. et al. Oxidized phospholipids, lipoprotein(a) and progression of aortic valve stenosis. J. Am. Coll. Cardiol. 66, 1236–1246 (2015).

    Article  CAS  PubMed  Google Scholar 

  52. Derbali, H. et al. Increased biglycan in aortic valve stenosis leads to the overexpression of phospholipid transfer protein via Toll-like receptor 2. Am. J. Pathol. 176, 2638–2645 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Song, R. et al. Biglycan induces the expression of osteogenic factors in human aortic valve interstitial cells via Toll-like receptor-2. Arterioscler. Thromb. Vasc. Biol. 32, 2711–2720 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mahmut, A. et al. Lipoprotein lipase in aortic valve stenosis is associated with lipid retention and remodelling. Eur. J. Clin. Invest. 43, 570–578 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Osman, N. et al. Smad2-dependent glycosaminoglycan elongation in aortic valve interstitial cells enhances binding of LDL to proteoglycans. Cardiovasc. Pathol. 22, 146–155 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Hung, M. Y., Witztum, J. L. & Tsimikas, S. New therapeutic targets for calcific aortic valve stenosis: the lipoprotein(a)–lipoprotein-associated phospholipase A2-oxidized phospholipid axis. J. Am. Coll. Cardiol. 63, 478–480 (2014).

    Article  PubMed  Google Scholar 

  57. Mahmut, A. et al. Elevated expression of Lp-PLA2 in calcific aortic valve disease: implication for valve mineralization. J. Am. Coll. Cardiol. 63, 460–469 (2014).

    Article  CAS  PubMed  Google Scholar 

  58. Mahmut, A. et al. Lp-PLA2 is associated with structural valve degeneration of bioprostheses. Eur. J. Clin. Invest. 44, 136–145 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Capoulade, R. et al. Impact of plasma Lp-PLA2 activity on the progression of aortic stenosis. JACC Cardiovasc. Imaging 8, 26–33 (2015).

    Article  PubMed  Google Scholar 

  60. Tellis, C. C. & Tselepis, A. D. The role of lipoprotein-associated phospholipase A2 in atherosclerosis may depend on its lipoprotein carrier in plasma. Biochim. Biophys. Acta 1791, 327–338 (2009).

    Article  CAS  PubMed  Google Scholar 

  61. Lehti, S. et al. Modified lipoprotein-derived lipid particles accumulate in human stenotic aortic valves. PLoS ONE 8, e65810 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Bouchareb, R. et al. Autotaxin derived from lipoprotein(a) and valve interstitial cells promotes inflammation and mineralization of the aortic valve. Circulation 132, 677–690 (2015).

    Article  CAS  PubMed  Google Scholar 

  63. Rogers, M. A. & Aikawa, E. Not a-so-little role for lipoprotein(a) in the development of calcific aortic valve disease. Circulation 132, 621–623 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Nagy, E. et al. Upregulation of the 5-lipoxygenase pathway in human aortic valves correlates with severity of stenosis and leads to leukotriene-induced effects on valvular myofibroblasts. Circulation 123, 1316–1325 (2011).

    Article  CAS  PubMed  Google Scholar 

  65. Wirrig, E. E., Gomez, M. V., Hinton, R. B. & Yutzey, K. E. COX2 inhibition reduces aortic valve calcification in vivo. Arterioscler. Thromb. Vasc. Biol. 35, 938–947 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Mathieu, P., Bouchareb, R. & Boulanger, M. C. Innate and adaptive immunity in calcific aortic valve disease. J. Immunol. Res. 2015, 851945 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Venardos, N. et al. Aortic valve calcification is mediated by a differential response of aortic valve interstitial cells to inflammation. J. Surg. Res. 190, 1–8 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. West, X. Z. et al. Oxidative stress induces angiogenesis by activating TLR2 with novel endogenous ligands. Nature 467, 972–976 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Winchester, R. et al. Circulating activated and effector memory T cells are associated with calcification and clonal expansions in bicuspid and tricuspid valves of calcific aortic stenosis. J. Immunol. 187, 1006–1014 (2011).

    Article  CAS  PubMed  Google Scholar 

  70. Yoshioka, M. et al. Chondromodulin-I maintains cardiac valvular function by preventing angiogenesis. Nat. Med. 12, 1151–1159 (2006).

    Article  CAS  PubMed  Google Scholar 

  71. Charest, A. et al. Distribution of SPARC during the neovascularisation of degenerative aortic stenosis. Heart 92, 1844–1849 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Bosse, Y. et al. Refining molecular pathways leading to calcific aortic valve stenosis by studying gene expression profile of normal calcified stenotic human aortic valves. Circ. Cardiovasc. Genet. 2, 489–498 (2009).

    Article  CAS  PubMed  Google Scholar 

  73. Helske, S. et al. Increased expression of elastolytic cathepsins S, K, and V and their inhibitor cystatin C in stenotic aortic valves. Arterioscler. Thromb. Vasc. Biol. 26, 1791–1798 (2006).

    Article  CAS  PubMed  Google Scholar 

  74. Aikawa, E. et al. Arterial and aortic valve calcification abolished by elastolytic cathepsin S deficiency in chronic renal disease. Circulation 119, 1785–1794 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Yu, Y. C., Boston, J. R., Simaan, M. A. & Antak, J. F. Minimally invasive estimation of systemic vascular parameters. Ann. Biomed. Eng. 29, 595–606 (2001).

    Article  CAS  PubMed  Google Scholar 

  76. Hsu, H., Shu, H. B., Pan, M. G. & Goeddel, D. V. TRADD–TRAF2 and TRADD–FADD interactions define two distinct TNF receptor 1 signal transduction pathways. Cell 84, 299–308 (1996).

    Article  CAS  PubMed  Google Scholar 

  77. Lee, H. L., Woo, K. M., Ryoo, H. M. & Baek, J. H. Tumor necrosis factor-α increases alkaline phosphatase expression in vascular smooth muscle cells via MSX2 induction. Biochem. Biophys. Res. Commun. 391, 1087–1092 (2010).

    Article  CAS  Google Scholar 

  78. Isoda, K., Matsuki, T., Kondo, H., Iwakura, Y. & Ohsuzu, F. Deficiency of interleukin-1 receptor antagonist induces aortic valve disease in BALB/c mice. Arterioscler. Thromb. Vasc. Biol. 30, 708–715 (2010).

    Article  CAS  PubMed  Google Scholar 

  79. Lai, C. F. et al. TNFR1-activated reactive oxidative species signals up-regulate osteogenic Msx2 programs in aortic myofibroblasts. Endocrinology 153, 3897–3910 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Galeone, A. et al. Aortic valvular interstitial cells apoptosis and calcification are mediated by TNF-related apoptosis-inducing ligand. Int. J. Cardiol. 169, 296–304 (2013).

    Article  PubMed  Google Scholar 

  81. El Husseini, D. et al. P2Y2 receptor represses IL-6 expression by valve interstitial cells through Akt: implication for calcific aortic valve disease. J. Mol. Cell. Cardiol. 72, 146–156 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Wada, T., Nakashima, T., Hiroshi, N. & Penninger, J. M. RANKL–RANK signaling in osteoclastogenesis and bone disease. Trends Mol. Med. 12, 17–25 (2006).

    Article  CAS  PubMed  Google Scholar 

  83. Kaden, J. J. et al. Receptor activator of nuclear factor κB ligand and osteoprotegerin regulate aortic valve calcification. J. Mol. Cell. Cardiol. 36, 57–66 (2004).

    Article  CAS  PubMed  Google Scholar 

  84. Weiss, R. M. et al. Osteoprotegerin inhibits aortic valve calcification and preserves valve function in hypercholesterolemic mice. PLoS ONE 8, e65201 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Demer, L. L. Vascular calcification and osteoporosis: inflammatory responses to oxidized lipids. Int. J. Epidemiol. 31, 737–741 (2002).

    Article  PubMed  Google Scholar 

  86. Skolnick, A. H., Osranek, M., Formica, P. & Kronzon, I. Osteoporosis treatment and progression of aortic stenosis. Am. J. Cardiol. 104, 122–124 (2009).

    Article  PubMed  Google Scholar 

  87. Osako, M. K. et al. Estrogen inhibits vascular calcification via vascular RANKL system: common mechanism of osteoporosis and vascular calcification. Circ. Res. 107, 466–475 (2010).

    Article  CAS  PubMed  Google Scholar 

  88. Helske, S. et al. Induction of local angiotensin II-producing systems in stenotic aortic valves. J. Am. Coll. Cardiol. 44, 1859–1866 (2004).

    Article  CAS  PubMed  Google Scholar 

  89. O'Brien, K. D. et al. Association of angiotensin-converting enzyme with low-density lipoprotein in aortic valvular lesions and in human plasma. Circulation 106, 2224–2230 (2002).

    Article  CAS  PubMed  Google Scholar 

  90. Cote, N. et al. Oxidized low-density lipoprotein, angiotensin II and increased waist cirumference are associated with valve inflammation in prehypertensive patients with aortic stenosis. Int. J. Cardiol. 145, 444–449 (2010).

    Article  PubMed  Google Scholar 

  91. Fujisaka, T. et al. Angiotensin II promotes aortic valve thickening independent of elevated blood pressure in apolipoprotein-E deficient mice. Atherosclerosis 226, 82–87 (2013).

    Article  CAS  PubMed  Google Scholar 

  92. Arishiro, K. et al. Angiotensin receptor-1 blocker inhibits atherosclerotic changes and endothelial disruption of the aortic valve in hypercholesterolemic rabbits. J. Am. Coll. Cardiol. 49, 1482–1489 (2007).

    Article  CAS  PubMed  Google Scholar 

  93. Cote, N., Couture, C., Pibarot, P., Despres, J. P. & Mathieu, P. Angiotensin receptor blockers are associated with a lower remodelling score of stenotic aortic valves. Eur. J. Clin. Invest. 41, 1172–1179 (2011).

    Article  CAS  PubMed  Google Scholar 

  94. Capoulade, R. et al. Impact of hypertension and renin–angiotensin system inhibitors in aortic stenosis. Eur. J. Clin. Invest. 43, 1262–1272 (2013).

    Article  CAS  PubMed  Google Scholar 

  95. Simmons, C. A., Grant, G. R., Manduchi, E. & Davies, P. F. Spatial heterogeneity of endothelial phenotypes correlates with side-specific vulnerability to calcification in normal porcine aortic valves. Circ. Res. 96, 792–799 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Holliday, C. J., Ankeny, R. F., Jo, H. & Nerem, R. M. Discovery of shear- and side-specific mRNAs and miRNAs in human aortic valvular endothelial cells. Am. J. Physiol. Heart Circ. Physiol. 301, H856–H867 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Garg, V. et al. Mutations in NOTCH1 cause aortic valve disease. Nature 437, 270–274 (2005). This is the first study to report that mutations in NOTCH1 cause both development of bicuspid aortic valve and calcification of aortic valve.

    Article  CAS  PubMed  Google Scholar 

  98. Nigam, V. & Srivastava, D. J. Notch1 represses osteogenic pathways in aortic valve cells. Mol. Cell. Cardiol. 47, 828–834 (2009).

    Article  CAS  Google Scholar 

  99. Nus, M. et al. Diet-induced aortic valve disease in mice haploinsufficient for the Notch pathway effector RBPJK/CSL. Arterioscler. Thromb. Vasc. Biol. 31, 1580–1588 (2011).

    Article  CAS  PubMed  Google Scholar 

  100. Deregowski, V., Gazzerro, E., Priest, L., Rydziel, S. & Canalis, E. Notch 1 overexpression inhibits osteoblastogenesis by suppressing Wnt/β-catenin but not bone morphogenetic protein signaling. J. Biol. Chem. 281, 6203–6210 (2006).

    Article  CAS  PubMed  Google Scholar 

  101. Theodoris, C. V. et al. Human disease modeling reveals integrated transcriptional and epigenetic mechanisms of NOTCH1 haploinsufficiency. Cell 160, 1072–1086 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Caira, F. C. et al. Human degenerative valve disease is associated with up-regulation of low-density lipoprotein receptor-related protein 5 receptor-mediated bone formation. J. Am. Coll. Cardiol. 47, 1707–1712 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Shao, J. S. et al. Msx2 promotes cardiovascular calcification by activating paracrine Wnt signals. J. Clin. Invest. 115, 1210–1220 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Cola, C., Almeida, M., Li, D., Romeo, F. & Mehta, J. L. Regulatory role of endothelium in the expression of genes affecting arterial calcification. Biochem. Biophys. Res. Commun. 320, 424–427 (2004).

    Article  CAS  PubMed  Google Scholar 

  105. Zhang, M. et al. MicroRNA-30b is a multifunctional regulator of aortic valve interstitial cells. J. Thorac Cardiovasc. Surg. 147, 1073–1080 (2014).

    Article  CAS  PubMed  Google Scholar 

  106. Bertazzo, S. et al. Nano-analytical electron microscopy reveals fundamental insights into human cardiovascular tissue calcification. Nat. Mater. 12, 576–583 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Bouchareb, R. et al. Mechanical strain induces the production of spheroid mineralized microparticles in the aortic valve through a RhoA/ROCK-dependent mechanism. J. Mol. Cell. Cardiol. 67, 49–59 (2014).

    Article  CAS  PubMed  Google Scholar 

  108. New, S. E. et al. Macrophage-derived matrix vesicles: an alternative novel mechanism for microcalcification in atherosclerotic plaques. Circ. Res. 113, 72–77 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Hjortnaes, J., New, S.E. & Aikawa, E. Visualizing novel concepts of cardiovascular calcification. Trends Cardiovasc. Med. 23, 71–79 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Cote, N. et al. ATP acts as a survival signal and prevents the mineralization of aortic valve. J. Mol. Cell. Cardiol. 52, 1191–1202 (2012).

    Article  CAS  PubMed  Google Scholar 

  111. Mahmut, A., Boulanger, M. C., Bouchareb, R., Hadji, F. & Mathieu, P. Adenosine derived from ecto-nucleotidases in calcific aortic valve disease promotes mineralization through A2a adenosine receptor. Cardiovasc. Res. 106, 109–120 (2015).

    Article  CAS  PubMed  Google Scholar 

  112. Mathieu, P. et al. Calcification of human valve interstitial cells is dependent on alkaline phosphatase activity. J. Heart Valve Dis. 14, 353–357 (2005).

    PubMed  Google Scholar 

  113. Bouchareb, R. et al. Carbonic anhydrase XII in valve interstitial cells promotes the regression of calcific aortic valve stenosis. J. Mol. Cell. Cardiol. 82, 104–115 (2015).

    Article  CAS  PubMed  Google Scholar 

  114. Lorell, B. H. & Carabello, B. A. Left ventricular hypertrophy: pathogenesis, detection, and prognosis. Circulation 102, 470–479 (2000).

    Article  CAS  PubMed  Google Scholar 

  115. Mewton, N., Liu, C. Y., Croisille, P., Bluemke, D. & Lima, J. A. Assessment of myocardial fibrosis with cardiovascular magnetic resonance. J. Am. Coll. Cardiol. 57, 891–903 (2011).

    Article  PubMed  Google Scholar 

  116. Carroll, J. D. et al. Sex-associated differences in left ventricular function in aortic stenosis of the elderly. Circulation 86, 1099–1107 (1992).

    Article  CAS  PubMed  Google Scholar 

  117. Pagé, A. et al. Metabolic syndrome is associated with more pronounced impairment of LV geometry and function in patients with calcific aortic stenosis: a substudy of the ASTRONOMER trial (Aortic Stenosis Progression Observation Measuring Effects of Rosuvastatin). J. Am. Coll. Cardiol. 55, 1867–1874 (2010).

    Article  PubMed  Google Scholar 

  118. Lund, B. P. et al. Effect of obesity on left ventricular mass and systolic function in patients with asymptomatic aortic stenosis (a Simvastatin Ezetimibe in Aortic Stenosis [SEAS] substudy). Am. J. Cardiol. 105, 1456–1460 (2010).

    Article  PubMed  Google Scholar 

  119. Lindman, B. R. et al. The adverse impact of diabetes mellitus on left ventricular remodeling and function in patients with severe aortic stenosis. Circ. Heart Fail 4, 286–292 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Cramariuc, D. et al. Low-flow aortic stenosis in asymptomatic patients: valvular arterial impedance and systolic function from the SEAS substudy. JACC Cardiovasc. Imaging 2, 390–399 (2009).

    Article  PubMed  Google Scholar 

  121. Cioffi, G. et al. Prognostic value of inappropriately high left ventricular mass in asymptomatic severe aortic stenosis. Heart 97, 301–307 (2011).

    Article  PubMed  Google Scholar 

  122. Duncan, A. I. et al. Influence of concentric left ventricular remodeling on early mortality after aortic valve replacement. Ann. Thorac Surg. 85, 2030–2039 (2008).

    Article  PubMed  Google Scholar 

  123. Rajappan, K. et al. Mechanisms of coronary microcirculatory dysfunction in patients with aortic stenosis and angiographically normal coronary arteries. Circulation 105, 470–476 (2002).

    Article  PubMed  Google Scholar 

  124. Rajappan, K. et al. Functional changes in coronary microcirculation after valve replacement in patients with aortic stenosis. Circulation 107, 3170–3175 (2003).

    Article  PubMed  Google Scholar 

  125. Julius, B. K. et al. Angina pectoris in patients with aortic stenosis and normal coronary arteries. Mechanisms and pathophysiological concepts. Circulation 95, 892–898 (1997).

    Article  CAS  PubMed  Google Scholar 

  126. Mutlak, D. et al. Frequency determinants and outcome of pulmonary hypertension in patients with aortic valve stenosis. Am. J. Med. Sci. 343, 397–401 (2012).

    Article  PubMed  Google Scholar 

  127. Lancellotti, P. et al. Determinants and prognostic significance of exercise pulmonary hypertension in asymptomatic severe aortic stenosis. Circulation 126, 851–859 (2012).

    Article  PubMed  Google Scholar 

  128. Dumesnil, J. G., Shoucri, R. M., Laurenceau, J. L. & Turcot, J. A mathematical model of the dynamic geometry of the intact left ventricle and its application to clinical data. Circulation 59, 1024–1034 (1979).

    Article  CAS  PubMed  Google Scholar 

  129. Lancellotti, P. et al. Risk stratification in asymptomatic moderate to severe aortic stenosis: the importance of the valvular, arterial and ventricular interplay. Heart 96, 1364–1371 (2010). This is a prospective study that shows the prognostic value of valve stenosis severity, valvulo-arterial impedance, left ventricular longitudinal function and left atrial dilation in patients with calcific aortic stenosis.

    Article  PubMed  Google Scholar 

  130. Kusunose, K. et al. Incremental prognostic value of left ventricular global longitudinal strain in patients with aortic stenosis and preserved ejection fraction. Circ. Cardiovasc. Imaging 7, 938–945 (2014).

    Article  PubMed  Google Scholar 

  131. Fernandez, B. et al. Bicuspid aortic valves with different spatial orientations of the leaflets are distinct etiological entities. J. Am. Coll. Cardiol. 54, 2312–2318 (2009).

    Article  PubMed  Google Scholar 

  132. Mathieu, P. et al. The pathology and pathobiology of bicuspid aortic valves: state of the art and novel research perspective. J. Pathol. Clin. Res. 1, 195–206 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  133. Mohty, D. et al. Age-related differences in the pathogenesis of calcific aortic stenosis: the potential role of resistin. Int. J. Cardiol. 142, 126–132 (2010).

    Article  PubMed  Google Scholar 

  134. Aronow, W. S., Schwartz, K. S. & Koenigsberg, M. Correlation of serum lipids, calcium, and phosphorus, diabetes mellitus and history of systemic hypertension with presence or absence of calcified or thickened aortic cusps or root in elderly patients. Am. J. Cardiol. 59, 998–999 (1987).

    Article  CAS  PubMed  Google Scholar 

  135. Katz, R. et al. Features of the metabolic syndrome and diabetes mellitus as predictors of aortic valve calcification in the multi-ethnic study of atherosclerosis. Circulation 113, 2113–2119 (2006).

    Article  PubMed  Google Scholar 

  136. Mohler, E. R., Sheridan, M. J., Nichols, R., Harvey, W. P. & Waller, B.F. Development and progression of aortic valve stenosis: atherosclerosis risk factors — a causal relationship? A clinical morphologic study. Clin. Cardiol. 14, 995–999 (1991).

    Article  CAS  PubMed  Google Scholar 

  137. Otto, C. M. et al. Prospective study of asymptomatic valvular aortic stenosis. Clinical, echocardiographic, and exercise predictors of outcome. Circulation 95, 2262–2270 (1997). This is one of the first prospective studies to describe the clinical and echocardiographic predictors of the progression and outcomes of calcific AS.

    Article  CAS  PubMed  Google Scholar 

  138. Livanainen, A. M., Lindroos, M., Tilvis, R., Heikkila, J. & Kupari, M. Natural history of aortic valve stenosis of varying severity in the elderly. Am. J. Cardiol. 78, 97–101 (1996).

    Article  Google Scholar 

  139. Palta, S., Pai, A. M., Gill, K. S. & Pai, R. G. New insights into the progression of aortic stenosis: implications for secondary prevention. Circulation 101, 2497–2502 (2000).

    Article  CAS  PubMed  Google Scholar 

  140. Ngo, M. V., Gottdiener, J. S., Fletcher, R. D., Fernicola, D. J. & Gersh, B. J. Smoking and obesity are associated with the progression of aortic stenosis. Am. J. Geriatr. Cardiol. 10, 86–90 (2001).

    Article  CAS  PubMed  Google Scholar 

  141. Capoulade, R. et al. Impact of metabolic syndrome on progression of aortic stenosis: influence of age and statin therapy. J. Am. Coll. Cardiol. 60, 216–223 (2012). This is a post hoc analysis of the ASTRONOMER study showing that metabolic syndrome is associated with faster progression of AS and that statins may accelerate the stenosis progression in these patients.

    Article  PubMed  Google Scholar 

  142. Hekimian, G. et al. Progression of aortic valve stenosis is associated with bone remodelling and secondary hyperparathyroidism in elderly patients — the COFRASA study. Eur. Heart J. 34, 1915–1922 (2013).

    Article  CAS  PubMed  Google Scholar 

  143. Briand, M. et al. Reduced systemic arterial compliance impacts significantly on left ventricular afterload and function in aortic stenosis: implications for diagnosis and treatment. J. Am. Coll. Cardiol. 46, 291–298 (2005).

    Article  PubMed  Google Scholar 

  144. Rieck, A. E. et al. Hypertension in aortic stenosis: implications for left ventricular structure and cardiovascular events. Hypertension 60, 90–97 (2012).

    Article  CAS  PubMed  Google Scholar 

  145. Cripe, L., Andelfinger, G., Martin, L. J., Shooner, K. & Benson, D. W. Bicuspid aortic valve is heritable. J. Am. Coll. Cardiol. 44, 138–143 (2004).

    Article  PubMed  Google Scholar 

  146. Laforest, B., Andelfinger, G. & Nemer, M. Loss of Gata5 in mice leads to bicuspid aortic valve. J. Clin. Invest. 121, 2876–2887 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Shi, L. M. et al. GATA5 loss-of-function mutations associated with congenital bicuspid aortic valve. Int. J. Mol. Med. 33, 1219–1226 (2014).

    Article  CAS  PubMed  Google Scholar 

  148. Foffa, I. et al. Sequencing of NOTCH1, GATA5, TGFBR1 and TGFBR2 genes in familial cases of bicuspid aortic valve. BMC Med. Genet. 14, 44 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Ducharme, V., Guauque-Olarte, S., Pibarot, P., Mathieu, P. & Bossé, Y. NOTCH1 genetic variants in patients with tricuspid calcific aortic valve stenosis. J. Heart Valve Dis. 22, 142–149 (2013).

    PubMed  Google Scholar 

  150. Guauque-Olarte, S. et al. Calcium signalings pathway genes RUNX2 and CACNA1C are associated with calcific aortic valve disease. Circ. Cardiovasc. Genet. 8, 812–822 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Smith, J. G. et al. Association of low-density lipoprotein cholesterol-related genetic variants with aortic valve calcium and incident aortic stenosis. JAMA 312, 1764–1771 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Cowell, S. J. et al. A randomized trial of intensive lipid-lowering therapy in calcific aortic stenosis. N. Engl. J. Med. 352, 2389–2397 (2005).

    Article  CAS  PubMed  Google Scholar 

  153. Rossebo, A. B. et al. Intensive lipid lowering with simvastatin and ezetimibe in aortic stenosis. N. Engl. J. Med. 359, 1343–1356 (2008). This is a randomized clinical trial that reports no effect of statins on the progression and the outcomes of calcific AS.

    Article  PubMed  Google Scholar 

  154. Chan, K. L., Teo, K., Dumesnil, J. G., Ni, A. & Tam, J. 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 121, 306–314 (2010).

    Article  CAS  PubMed  Google Scholar 

  155. Baumgartner, H. et al. Echocardiographic assessment of valve stenosis: EAE/ASE recommendations for clinical practice. Eur. J. Echocardiogr. 10, 1–25 (2009).

    Article  PubMed  Google Scholar 

  156. Pibarot, P. & Dumesnil, J. G. Low-flow, low-gradient aortic stenosis with normal and depressed left ventricular ejection fraction. J. Am. Coll. Cardiol. 60, 1845–1853 (2012). This is a review of the diagnosis and the management of low-flow, low-gradient AS, which is one of the most challenging entities in patients with valvular heart diseases.

    Article  PubMed  Google Scholar 

  157. Nishimuram, R. A. & Carabello, B. A. Hemodynamics in the cardiac catheterization laboratory of the 21st century. Circulation 125, 2138–2150 (2012).

    Article  Google Scholar 

  158. Omran, H. et al. Silent and apparent cerebral embolism after retrograde catheterisation of the aortic valve in valvular stenosis: a prospective, randomised study. Lancet 361, 1241–1246 (2003).

    Article  PubMed  Google Scholar 

  159. Otto, C. M., Lind, B. K., Kitzman, D. W., Gersh, B. J. & Siscovick, D. S. Association of aortic-valve sclerosis with cardiovascular mortality and morbidity in the elderly. N. Engl. J. Med. 341, 142–147 (1999).

    Article  CAS  PubMed  Google Scholar 

  160. Owens, D. S. et al. Aortic valve calcium independently predicts coronary and cardiovascular events in a primary prevention population. J. Am. Coll. Cardiol. 5, 619–625 (2012).

    Article  Google Scholar 

  161. Lancellotti, P. et al. Prognostic importance of quantitative exercise Doppler echocardiography in asymptomatic valvular aortic stenosis. Circulation 112, I377–I382 (2005).

    PubMed  Google Scholar 

  162. Maréchaux, S. et al. Usefulness of exercise stress echocardiography for risk stratification of true asymptomatic patients with aortic valve stenosis. Eur. Heart J. 31, 1390–1397 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  163. Monin, J. L. et al. Low-gradient aortic stenosis: operative risk stratification and predictors for long-term outcome: a multicenter study using dobutamine stress hemodynamics. Circulation 108, 319–324 (2003).

    Article  PubMed  Google Scholar 

  164. Hachicha, Z., Dumesnil, J. G., Bogaty, P. & Pibarot, P. Paradoxical low flow low gradient severe aortic stenosis despite preserved ejection fraction is associated with higher afterload and reduced survival. Circulation 115, 2856–2864 (2007).

    Article  PubMed  Google Scholar 

  165. Blais, C. et al. Projected valve area at normal flow rate improves the assessment of stenosis severity in patients with low flow, low-gradient aortic stenosis: the multicenter TOPAS (Truly or Pseudo Severe Aortic Stenosis) study. Circulation 113, 711–721 (2006).

    Article  PubMed  Google Scholar 

  166. Clavel, M. A. et al. Validation of conventional and simplified methods to calculate projected valve area at normal flow rate in patients with low flow, low gradient aortic stenosis: the multicenter TOPAS (True or Pseudo Severe Aortic Stenosis) study. J. Am. Soc. Echocardiogr. 23, 380–386 (2010).

    Article  PubMed  Google Scholar 

  167. Tribouilloy, C. et al. Outcome after aortic valve replacement for low-flow/low-gradient aortic stenosis without contractile reserve on dobutamine stress echocardiography. J. Am. Coll. Cardiol. 53, 1865–1873 (2009).

    Article  PubMed  Google Scholar 

  168. Clavel, M. A. et al. Stress echocardiography to assess stenosis severity and predict outcome in patients with paradoxical low-flow, low-gradient aortic stenosis and preserved LVEF. JACC Cardiovasc. Imaging 6, 175–183 (2013).

    Article  PubMed  Google Scholar 

  169. Clavel, M. A. 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. 62, 2329–2338 (2013). This is a multicentre study showing the usefulness of aortic valve calcium scoring by MDCT to corroborate the stenosis severity in patients with discordant findings at echocardiography.

    Article  CAS  PubMed  Google Scholar 

  170. Clavel, M. A. et al. Impact of aortic valve calcification, as measured by MDCT, on survival in patients with aortic stenosis: results of an international registry study. J. Am. Coll. Cardiol. 64, 1202–1213 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  171. Rosenhek, R. et al. Predictors of outcome in severe, asymptomatic aortic stenosis. N. Engl. J. Med. 343, 611–617 (2000).

    Article  CAS  PubMed  Google Scholar 

  172. Jenkins, W. S. et al. Valvular 18F-fluoride and 18F-fluorodeoxyglucose uptake predict disease progression and clinical outcome in patients with aortic stenosis. J. Am. Coll. Cardiol. 66, 1200–1201 (2015).

    Article  PubMed  Google Scholar 

  173. Minners, J. et al. Inconsistent grading of aortic valve stenosis by current guidelines: haemodynamic studies in patients with apparently normal left ventricular function. Heart 96, 1463–1468 (2010).

    Article  PubMed  Google Scholar 

  174. Dayan, V. et al. Outcome and impact of aortic valve replacement in patients with preserved LVEF and low-gradient aortic stenosis. J. Am. Coll. Cardiol. 66, 2594–2603 (2015).

    Article  PubMed  Google Scholar 

  175. Hyafil, F. et al. Detection of 18fluoride sodium accumulation by positron emission tomography in calcified stenotic aortic valves. Am. J. Cardiol. 109, 1194–1196 (2012).

    Article  PubMed  Google Scholar 

  176. Dweck, M. R. et al. Assessment of valvular calcification and inflammation by positron emission tomography in patients with aortic stenosis. Circulation 125, 76–86 (2012).

    Article  CAS  PubMed  Google Scholar 

  177. Dweck, M. R. et al. 18F-sodium fluoride uptake is a marker of active calcification and disease progression in patients with aortic stenosis. Circ. Cardiovasc. Imaging 7, 371–378 (2014). This study shows the use of sodium fluoride PET to detect the mineralization activity in the aortic valve and to predict the rapidity of aortic stenosis progression.

    Article  PubMed  Google Scholar 

  178. Le Ven, F. et al. Valve tissue characterization by magnetic resonance imaging in calcific aortic valve disease. Can. J. Cardiol. 30, 1676–1683 (2014).

    Article  PubMed  Google Scholar 

  179. Hope, M. D. et al. Bicuspid aortic valve: four-dimensional MR evaluation of ascending aortic systolic flow patterns. Radiology 255, 53–61 (2010).

    Article  PubMed  Google Scholar 

  180. Hope, M. D. et al. 4D flow CMR in assessment of valve-related ascending aortic disease. JACC Cardiovasc. Imaging 4, 781–787 (2011).

    Article  PubMed  Google Scholar 

  181. Bartko, P. E. et al. Two-dimensional strain for the assessment of left ventricular function in low flow-low gradient aortic stenosis, relationship to hemodynamics and outcome: a substudy of the multicenter TOPAS study. Circ. Cardiovasc. Imaging 6, 268–276 (2012).

    Article  PubMed  Google Scholar 

  182. Dahou, A. et al. Usefulness of global left ventricular longitudinal strain for risk stratification in low ejection fraction, low-gradient aortic stenosis: results from the multicenter true or pseudo-severe aortic stenosis study. Circ. Cardiovasc. Imaging 8, e002117 (2015).

    Article  PubMed  Google Scholar 

  183. Lancellotti, P. et al. Impact of global left ventricular afterload on left ventricular function in asymptomatic severe aortic stenosis: a two-dimensional speckle-tracking study. Eur. J. Echocardiogr. 11, 537–543 (2010).

    Article  PubMed  Google Scholar 

  184. Dahl, J. S. et al. Effect of left ventricular ejection fraction on postoperative outcome in patients with severe aortic stenosis undergoing aortic valve replacement. Circ. Cardiovasc. Imaging 8, e002917 (2015).

    Article  PubMed  Google Scholar 

  185. Lancellotti, P. et al. Prognostic effect of long-axis left ventricular dysfunction and B-type natriuretic peptide levels in asymptomatic aortic stenosis. Am. J. Cardiol. 105, 383–388 (2010).

    Article  CAS  PubMed  Google Scholar 

  186. Ozkan, A., Kapadia, S., Tuzcu, M. & Marwick, T. H. Assessment of left ventricular function in aortic stenosis. Nat. Rev. Cardiol. 8, 494–501 (2011).

    Article  PubMed  Google Scholar 

  187. Clavel, M. A. et al. B-type natriuretic peptide clinical activation in aortic stenosis: impact on long-term survival. J. Am. Coll. Cardiol. 63, 2016–2025 (2014). This study shows the prognostic value of plasma BNP in patients with calcific AS. It also shows the importance of standardizing the measured level of BNP against the normal reference value for the patient's age and sex.

    Article  CAS  PubMed  Google Scholar 

  188. Weidemann, F. et al. Impact of myocardial fibrosis in patients with symptomatic severe aortic stenosis. Circulation 120, 577–584 (2009).

    Article  CAS  PubMed  Google Scholar 

  189. Azevedo, C. F. et al. Prognostic significance of myocardial fibrosis quantification by histopathology and magnetic resonance imaging in patients with severe aortic valve disease. J. Am. Coll. Cardiol. 56, 278–287 (2010). This is one of the first studies to show the prognostic value of myocardial fibrosis measured by CMR in patients with calcific AS.

    Article  PubMed  Google Scholar 

  190. Dweck, M. R. et al. Midwall fibrosis is an independent predictor of mortality in patients with aortic stenosis. J. Am. Coll. Cardiol. 58, 1271–1279 (2011).

    Article  PubMed  Google Scholar 

  191. Milano, A. D. et al. Prognostic value of myocardial fibrosis in patients with severe aortic valve stenosis. J. Thorac Cardiovasc. Surg. 144, 830–837 (2012).

    Article  PubMed  Google Scholar 

  192. Herrmann, S. et al. Low-gradient aortic valve stenosis: myocardial fibrosis and its influence on function and outcome. J. Am. Coll. Cardiol. 58, 402–412 (2011).

    Article  PubMed  Google Scholar 

  193. Nazarian, S. Is ventricular arrhythmia a possible mediator of the association between aortic stenosis-related midwall fibrosis and mortality? J. Am. Coll. Cardiol. 58, 1280–1282 (2011).

    Article  PubMed  Google Scholar 

  194. Chin, C. W. et al. High-sensitivity troponin I concentrations are a marker of an advanced hypertrophic response and adverse outcomes in patients with aortic stenosis. Eur. Heart J. 35, 2312–2321 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Rosjo, H., Andreassen, J., Edvardsen, T. & Omland, T. Prognostic usefulness of circulating high-sensitivity troponin T in aortic stenosis and relation to echocardiographic indexes of cardiac function and anatomy. Am. J. Cardiol. 108, 88–91 (2011).

    Article  PubMed  Google Scholar 

  196. Chen, Z. et al. Circulating level of miR-378 predicts left ventricular hypertrophy in patients with aortic stenosis. PLoS ONE 9, e105702 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Rosjo, H. et al. Prognostic value of circulating microRNA-210 levels in patients with moderate to severe aortic stenosis. PLoS ONE 9, e91812 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Lindman, B. R. et al. Prognostic utility of novel biomarkers of cardiovascular stress in patients with aortic stenosis undergoing valve replacement. Heart 101, 1382–1388 (2015).

    Article  CAS  PubMed  Google Scholar 

  199. Lindman, B. R., Bonow, R. O. & Otto, C. M. Current management of calcific aortic stenosis. Circ. Res. 113, 223–237 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Iung, B. et al. Decision-making in elderly patients with severe aortic stenosis: why are so many denied surgery? Eur. Heart J. 26, 2714–2720 (2005).

    Article  PubMed  Google Scholar 

  201. Leon, M. B. et al. Transcatheter aortic-valve implantation for aortic stenosis in patients who cannot undergo surgery. N. Eng. J. Med. 363, 1597–1607 (2010). This randomized clinical trial shows the major superiority of TAVR compared with conservative management in patients who cannot undergo surgical AVR.

    Article  CAS  Google Scholar 

  202. Lindman, B. R., Alexander, K. P., O'Gara, P. T. & Afilalo, J. Futility, benefit, and transcatheter aortic valve replacement. JACC Cardiovasc. Interv. 7, 707–716 (2014). This review describes the challenges and strategies for decision-making in patients with severe calcific AS who are potential candidates for TAVR.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Holmes, D. R. Jr, Rich, J. B., Zoghbi, W. A. & Mack, M. J. The heart team of cardiovascular care. J. Am. Coll. Cardiol. 61, 903–907 (2013).

    Article  PubMed  Google Scholar 

  204. Holmes, D. R. Jr, Mohr, F., Hamm, C. W. & Mack, M. J. Venn diagrams in cardiovascular disease: the Heart Team concept. Eur. J. Cardiothorac Surg. 43, 255–257 (2013).

    Article  PubMed  Google Scholar 

  205. Holmes, D. R. et al. 2012 ACCF/AATS/SCAI/STS expert consensus document on transcatheter aortic valve replacement. J. Am. Coll. Cardiol. 59, 1200–1254 (2012).

    Article  PubMed  Google Scholar 

  206. Levy, F. et al. Aortic valve replacement for low-flow/low-gradient aortic stenosis: operative risk stratification and long-term outcome: a European multicenter study. J. Am. Coll. Cardiol. 51, 1466–1472 (2008).

    Article  PubMed  Google Scholar 

  207. Harken, D. E. et al. Partial and complete prostheses in aortic insufficiency. J. Thorac Cardiovasc. Surg. 40, 744–762 (1960).

    CAS  PubMed  Google Scholar 

  208. Lee, R. et al. Fifteen-year outcome trends for valve surgery in North America. Ann. Thorac Surg. 91, 677–684 (2011).

    Article  PubMed  Google Scholar 

  209. Brown, J. M. et al. Isolated aortic valve replacement in North America comprising 108,687 patients in 10 years: changes in risks, valve types, and outcomes in the Society of Thoracic Surgeons National Database. J. Thorac Cardiovasc. Surg. 137, 82–90 (2009).

    Article  PubMed  Google Scholar 

  210. Hamm, C. W. et al. The German Aortic Valve registry (GARY): in-hospital outcome. Eur. Heart J. 35, 1588–1598 (2014).

    Article  PubMed  Google Scholar 

  211. David, T. E., Woo, A., Armstrong, S. & Maganti, M. When is the Ross operation a good option to treat aortic valve disease? J. Thorac Cardiovasc. Surg. 139, 68–73 (2010).

    Article  PubMed  Google Scholar 

  212. Stulak, J. M. et al. Spectrum and outcome of reoperations after the Ross procedure. Circulation 122, 1153–1158 (2010).

    Article  PubMed  Google Scholar 

  213. David, T. E. Reoperations after the Ross procedure. Circulation 122, 1139–1140 (2010).

    Article  PubMed  Google Scholar 

  214. Chiang, Y. P. et al. Survival and long-term outcomes following bioprosthetic versus mechanical aortic valve replacement in patients aged 50 to 69 years. JAMA 312, 1323–1329 (2014).

    Article  CAS  PubMed  Google Scholar 

  215. Brown, M. L., McKellar, S. H., Sundt, T. M. & Schaff, H. V. Ministernotomy versus conventional sternotomy for aortic valve replacement: a systematic review and meta-analysis. J. Thorac Cardiovasc. Surg. 137, 670–679 (2009).

    Article  PubMed  Google Scholar 

  216. Astor, B. C., Kaczmarek, R. G., Hefflin, B. & Daley, W. R. Mortality after aortic valve replacement: results from a nationally representative database. Ann. Thorac Surg. 70, 1939–1945 (2000).

    Article  CAS  PubMed  Google Scholar 

  217. Ambler, G. et al. Generic, simple risk stratification model for heart valve surgery. Circulation 112, 224–231 (2005).

    Article  PubMed  Google Scholar 

  218. Ashikhmina, E. A. et al. Aortic valve replacement in the elderly: determinants of late outcome. Circulation 124, 1070–1078 (2011).

    Article  PubMed  Google Scholar 

  219. Bach, D. S. et al. Evaluation of patients with severe symptomatic aortic stenosis who do not undergo aortic valve replacement: the potential role of subjectively overestimated operative risk. Circ. Cardiovasc. Qual. Outcomes 2, 533–539 (2009).

    Article  PubMed  Google Scholar 

  220. Smith, C. R. et al. Transcatheter versus surgical aortic-valve replacement in high-risk patients. N. Engl. J. Med. 364, 2187–2198 (2011).

    Article  CAS  PubMed  Google Scholar 

  221. Adams, D. H. et al. Transcatheter aortic-valve replacement with a self-expanding prosthesis. N. Engl. J. Med. 370, 1790–1798 (2014). This randomized clinical trial shows the superiority of TAVR compared with surgical AVR in patients with intermediate or high surgical risk.

    Article  CAS  PubMed  Google Scholar 

  222. Kapadia, S. R. et al. 5-year outcomes of transcatheter aortic valve replacement compared with standard treatment for patients with inoperable aortic stenosis (PARTNER 1): a randomised controlled trial. Lancet 385, 2485–2491 (2015).

    Article  PubMed  Google Scholar 

  223. Mack, M. J. et al. 5-year outcomes of transcatheter aortic valve replacement or surgical aortic valve replacement for high surgical risk patients with aortic stenosis (PARTNER 1): a randomised controlled trial. Lancet 385, 2477–2484 (2015).

    Article  PubMed  Google Scholar 

  224. Popma, J. J. et al. Transcatheter aortic valve replacement using a self-expanding bioprosthesis in patients with severe aortic stenosis at extreme risk for surgery. J. Am. Coll. Cardiol. 63, 1972–1981 (2014).

    Article  PubMed  Google Scholar 

  225. Biere, L. et al. Influence of sex on mortality and perioperative outcomes in patients undergoing TAVR: insights from the FRANCE 2 registry. J. Am. Coll. Cardiol. 65, 755–757 (2015).

    Article  PubMed  Google Scholar 

  226. Gilard, M. et al. Registry of transcatheter aortic-valve implantation in high-risk patients. N. Eng. J. Med. 366, 1705–1715 (2012).

    Article  CAS  Google Scholar 

  227. Ludman, P. F. et al. Transcatheter aortic valve implantation in the United Kingdom: temporal trends, predictors of outcome, and 6-year follow-up: a report from the UK Transcatheter Aortic Valve Implantation (TAVI) registry, 2007 to 2012. Circulation 131, 1181–1190 (2015).

    Article  PubMed  Google Scholar 

  228. Holmes, D. R. et al. Clinical outcomes at 1 year following transcatheter aortic valve replacement. JAMA 313, 1019–1028 (2015).

    Article  CAS  PubMed  Google Scholar 

  229. Blackstone, E. H. et al. Propensity-matched comparisons of clinical outcomes after transapical or transfemoral TAVR: a PARTNER-I trial substudy. Circulation 131, 1989–2000 (2015).

    Article  PubMed  Google Scholar 

  230. Martinez-Clark, P. O. et al. Transcaval retrograde transcatheter aortic valve replacement for patients with no other access: first-in-man experience with CoreValve. JACC Cardiovasc. Interv 7, 1075–1077 (2014).

    Article  PubMed  Google Scholar 

  231. Moat, N. E. et al. Long-term outcomes after transcatheter aortic valve implantation in high-risk patients with severe aortic stenosis the U. K. TAVI (United Kingdom Transcatheter Aortic Valve Implantation) registry. J. Am. Coll. Cardiol. 58, 2130–2138 (2011).

    Article  PubMed  Google Scholar 

  232. Walther, T. et al. Perioperative results and complications in 15,964 transcatheter aortic valve replacements: prospective data from the GARY registry. J. Am. Coll. Cardiol. 65, 2173–2180 (2015).

    Article  PubMed  Google Scholar 

  233. Rodes-Cabau, J. Transcatheter aortic valve implantation: current and future approaches. Nat. Rev. Cardiol. 9, 15–29 (2012).

    Article  Google Scholar 

  234. Athappan, G. et al. Incidence, predictors, and outcomes of aortic regurgitation after transcatheter aortic valve replacement: meta-analysis and systematic review of literature. J. Am. Coll. Cardiol. 61, 1585–1595 (2013).

    Article  PubMed  Google Scholar 

  235. Genereux, P. et al. Paravalvular leak after transcatheter aortic valve replacement: the new Achilles’ heel? A comprehensive review of the literature. J. Am. Coll. Cardiol. 61, 1125–1136 (2013).

    Article  PubMed  Google Scholar 

  236. Pibarot, P., Hahn, R. T., Weissman, N. J. & Monaghan, M. J. Assessment of paravalvular regurgitation following TAVR: a proposal of unifying grading scheme. JACC Cardiovasc. Imaging 8, 340–360 (2015).

    Article  PubMed  Google Scholar 

  237. Kodali, S. et al. Paravalvular regurgitation after transcatheter aortic valve replacement with the Edwards sapien valve in the PARTNER trial: characterizing patients and impact on outcomes. Eur. Heart J. 36, 449–456 (2015).

    Article  PubMed  Google Scholar 

  238. Van Belle, E. et al. Postprocedural aortic regurgitation in balloon-expandable and self-expandable TAVR procedures: analysis of predictors and impact on long-term mortality: insights from the FRANCE2 registry. Circulation 129, 1415–1427 (2014).

    Article  PubMed  Google Scholar 

  239. Babaliaros, V. et al. Comparison of transfemoral transcatheter aortic valve replacement performed in the catheterization laboratory (minimalist approach) versus hybrid operating room (standard approach): outcomes and cost analysis. JACC Cardiovasc. Interv. 7, 898–904 (2014).

    Article  PubMed  Google Scholar 

  240. Dvir, D. et al. Transcatheter aortic valve implantation in failed bioprosthetic surgical valves. JAMA 312, 162–170 (2014).

    Article  CAS  PubMed  Google Scholar 

  241. Makkar, R. R. et al. Stratification of outcomes after transcatheter aortic valve replacement according to surgical inoperability for technical versus clinical reasons. J. Am. Coll. Cardiol. 63, 901–911 (2014).

    Article  PubMed  Google Scholar 

  242. Toggweiler, S. et al. Transcatheter aortic valve replacement: outcomes of patients with moderate or severe mitral regurgitation. J. Am. Coll. Cardiol. 59, 2068–2074 (2012).

    Article  PubMed  Google Scholar 

  243. Lindman, B. R. et al. Effect of tricuspid regurgitation and the right heart on survival after transcatheter aortic valve replacement: insights from the placement of aortic transcatheter valves II inoperable cohort. Circ. Cardiovasc. Interv. 8, e002073 (2015).

    Article  PubMed  Google Scholar 

  244. Reynolds, M. R. et al. Health-related quality of life after transcatheter aortic valve replacement in inoperable patients with severe aortic stenosis. Circulation 124, 1964–1972 (2011).

    Article  PubMed  Google Scholar 

  245. Reynolds, M. R. et al. Health-related quality of life after transcatheter or surgical aortic valve replacement in high-risk patients with severe aortic stenosis: results from the PARTNER (Placement of AoRTic TraNscathetER Valve) trial (cohort A). J. Am. Coll. Cardiol. 60, 548–558 (2012).

    Article  PubMed  Google Scholar 

  246. Svensson, L. G. et al. Comprehensive analysis of mortality among patients undergoing TAVR: results of the PARTNER trial. J. Am. Coll. Cardiol. 64, 158–168 (2014).

    Article  PubMed  Google Scholar 

  247. Goel, S. S. et al. Severe aortic stenosis and coronary artery disease — implications for management in the transcatheter aortic valve replacement era: a comprehensive review. J. Am. Coll. Cardiol. 62, 1–10 (2013).

    Article  PubMed  Google Scholar 

  248. Stefanini, G. G. et al. Coronary artery disease severity and aortic stenosis: clinical outcomes according to SYNTAX score in patients undergoing transcatheter aortic valve implantation. Eur. Heart J. 35, 2530–2540 (2014).

    Article  PubMed  Google Scholar 

  249. Paradis, J. M. et al. Aortic stenosis and coronary artery disease: What do we know? What don't we know? A comprehensive review of the literature with proposed treatment algorithms. Eur. Heart J. 35, 2069–2082 (2014).

    Article  PubMed  Google Scholar 

  250. Otto, C. M. et al. Three-year outcome after balloon aortic valvuloplasty. Insights into prognosis of valvular aortic stenosis. Circulation 89, 642–650 (1994).

    Article  CAS  PubMed  Google Scholar 

  251. Kapadia, S. et al. Outcomes of inoperable symptomatic aortic stenosis patients not undergoing aortic valve replacement: insight into the impact of balloon aortic valvuloplasty from the PARTNER trial (Placement of AoRtic TraNscathetER valve trial). JACC Cardiovasc. Interv. 8, 324–333 (2015).

    Article  PubMed  Google Scholar 

  252. Otto, C. M. & Prendergast, B. Aortic-valve stenosis — from patients at risk to severe valve obstruction. N. Engl. J. Med. 371, 744–756 (2014).

    Article  CAS  PubMed  Google Scholar 

  253. Schwarz, F. et al. The effect of aortic valve replacement on survival. Circulation 66, 1105–1110 (1982).

    Article  CAS  PubMed  Google Scholar 

  254. Green, 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. 5, 974–981 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  255. Schoenenberger, A. W. et al. Predictors of functional decline in elderly patients undergoing transcatheter aortic valve implantation (TAVI). Eur. Heart J. 34, 692 (2013).

    Article  CAS  Google Scholar 

  256. Stortecky, S. et al. Evaluation of multidimensional geriatric assessment as a predictor of mortality and cardiovascular events after transcatheter aortic valve implantation. JACC Cardiovasc. Interv. 5, 489–496 (2012).

    Article  PubMed  Google Scholar 

  257. Mihaljevic, T. et al. Survival after valve replacement for aortic stenosis: implications for decision making. J. Thorac Cardiovasc. Surg. 135, 1270–1278 (2008).

    Article  PubMed  Google Scholar 

  258. Kupari, M., Turto, H. & Lommi, J. Left ventricular hypertrophy in aortic valve stenosis: preventive or promotive of systolic dysfunction and heart failure? Eur. Heart J. 26, 1790–1796 (2005).

    Article  PubMed  Google Scholar 

  259. Dweck, M. R., Boon, N. A. & Newby, D. E. Calcific aortic stenosis: a disease of the valve and the myocardium. J. Am. Coll. Cardiol. 60, 1854–1863 (2012).

    Article  PubMed  Google Scholar 

  260. Biner, S. et al. Prognostic value of E/E' ratio in patients with unoperated severe aortic stenosis. JACC Cardiovasc. Imaging 3, 899–907 (2010).

    Article  PubMed  Google Scholar 

  261. Dewey, T. M. et al. Reliability of risk algorithms in predicting early and late operative outcomes in high-risk patients undergoing aortic valve replacement. J. Thorac Cardiovasc. Surg. 135, 180–187 (2008).

    Article  PubMed  Google Scholar 

  262. Lindman, B. R. et al. Transcatheter versus surgical aortic valve replacement in patients with diabetes and severe aortic stenosis at high risk for surgery: an analysis of the PARTNER trial (placement of aortic transcatheter valve). J. Am. Coll. Cardiol. 63, 1090–1099 (2014).

    Article  PubMed  Google Scholar 

  263. Herrmann, H. C. et al. Predictors of mortality and outcomes of therapy in low flow severe aortic stenosis: a PARTNER trial analysis. Circulation 127, 2316–2326 (2013).

    Article  PubMed  Google Scholar 

  264. Dvir, D. et al. Outcomes of patients with chronic lung disease and severe aortic stenosis treated with transcatheter versus surgical aortic valve replacement or standard therapy: insights for the PARTNER trial. J. Am. Coll. Cardiol. 63, 269–279 (2014).

    Article  PubMed  Google Scholar 

  265. Mok, M. et al. Chronic obstructive pulmonary disease in patients undergoing transcatheter aortic valve implantation: insights on clinical outcomes, prognostic markers, and functional status changes. JACC Cardiovasc. Interv. 6, 1072–1084 (2013).

    Article  PubMed  Google Scholar 

  266. Rodes-Cabau, J. et al. Impact of aortic annulus size on valve hemodynamics and clinical outcomes after transcatheter and surgical aortic valve replacement: insights from the PARTNER Trial. Circ. Cardiovasc. Interv 7, 701–711 (2014).

    Article  PubMed  Google Scholar 

  267. Gjesdal, O., Bluemke, D. A. & Lima, J. A. Cardiac remodeling at the population level — risk factors, screening, and outcomes. Nat. Rev. Cardiol. 8, 673–685 (2011).

    Article  PubMed  Google Scholar 

  268. Bull, S. et al. Human non-contrast T1 values and correlation with histology in diffuse fibrosis. Heart 99, 932–937 (2013).

    Article  PubMed  Google Scholar 

  269. Makkar, R. R. et al. Transcateter aortic-valve replacement for inoperable severe aortic stenosis. N. Engl. J. Med. 366, 1696–1704 (2012).

    Article  CAS  PubMed  Google Scholar 

  270. Kodali, S. K. et al. Two-year outcomes after transcatheter or surgical aortic-valve replacement. N. Engl. J. Med. 366, 1686–1695 (2012). This article reports the 2-year outcomes of the first randomized trial comparing transcatheter versus surgical AVR. This study shows that paravalvular regurgitation following TAVR is associated with increased risk of mortality.

  271. Webb, J. G. et al. A randomized evaluation of the SAPIEN XT transcatheter heart valve system in patients with aortic stenosis who are not candidates for surgery. JACC Cardiovasc. Interv. 8, 1797–1806 (2015).

    Article  PubMed  Google Scholar 

  272. Kodali, S. & The PARTNER 3 TAVR Investigators. Clinical and echocardiographic outcomes at 30 days with the SAPIEN 3 TAVR system in inoperable, high-risk and intermediate-risk AS patients. J. Am. Coll. Cardiol. Abstr. press release [online] (2015).

  273. Herrmann, H. C. Evaluation of a balloon-expandable transcatheter aortic valve in high-risk and inoperable patients with aortic stenosis — one-year outcomes. J. Am. Coll. Cardiol. Abstr. [online] (2015).

  274. Reardon, M. J. et al. 2-year outcomes in patients undergoing surgical or self-expanding transcatheter aortic valve replacement. J. Am. Coll. Cardiol. 66, 113–121 (2015).

    Article  PubMed  Google Scholar 

  275. Abdel-Wahab, M. et al. Comparison of balloon-expandable versus self-expandable valves in patients undergoing transcatheter aortic valve replacement: the CHOICE randomized clinical trial. JAMA 311, 1503–1514 (2014).

    Article  CAS  PubMed  Google Scholar 

  276. Thyregod, H. G. et al. Transcatheter versus surgical aortic valve replacement in patients with severe aortic valve stenosis: one-year results from the all-comers nordic aortic valve intervention (NOTION) randomized clinical trial. J. Am. Coll. Cardiol. 65, 2184–2194 (2015).

    Article  PubMed  Google Scholar 

  277. US National Library of Science. The PARTNER II trial: Placement of AoRTic TraNscathetER Valves. ClinicalTrials.gov [online], (2011).

  278. US National Library of Science. Safety and efficacy study of the Medtronic CoreValve® system in the treatment of severe, symptomatic aortic stenosis in intermediate risk subjects who need aortic valve replacement (SURTAVI). ClinicalTrials.gov [online], (2012).

Download references

Acknowledgements

B.R.L. is supported by the NIH K23 HL116660 and was a Gilead Sciences Research Scholars Program in Cardiovascular Disease Award Recipient. P.P. holds the Canada Research Chair in Valvular Heart Disease and his research programme is funded by the Canadian Institutes of Health Research (Grant numbers FDN-143225, MOP 126072, MOP 114997 and MOP 102737; Ottawa, Ontario, Canada). P.M. is a research scholar from the Fonds de Recherche du Québec-Santé (FRQS) and his research programme is supported by the Canadian Institutes of Health Research (MOP114893, MOP245048 and MOP341860), the Heart and Stroke Foundation of Canada and the Fonds nature et Technologies-Québec.

Author information

Authors and Affiliations

Authors

Contributions

Introduction (P.P.); Epidemiology (B.I. and P.P.); Mechanisms/pathophysiology (P.M. and P.P.); Diagnosis, screening and prevention (B.R.L., M.-A.C., P.L., P.M. and P.P.); Management (B.R.L., C.M.O., P.L., P.M. and P.P.); Quality of life (B.R.L., C.M.O. and P.P.); Outlook (B.L. and P.P.); Overview of Primer (P.P.).

Corresponding author

Correspondence to Philippe Pibarot.

Ethics declarations

Competing interests

B.R.L. has received research support from and served on the scientific advisory board for Roche Diagnostics. B.I. has received consultant fees from Abbott and Boehringer Ingelheim and speaker's fees from Edwards Lifesciences. P.P. has Core Lab contracts with Edwards Lifesciences, for which he receives no direct compensation. The other authors declare no competing interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lindman, B., Clavel, MA., Mathieu, P. et al. Calcific aortic stenosis. Nat Rev Dis Primers 2, 16006 (2016). https://doi.org/10.1038/nrdp.2016.6

Download citation

  • Published:

  • DOI: https://doi.org/10.1038/nrdp.2016.6

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing