/  Part II.6 – Translational Pathway for Tissue Engineered Aortic Valves

 

II.6

Translational Pathway for Tissue Engineered Aortic Valves

Jerome Sohier PhD, Adrian H Chester PhD
and Sir Magdi H Yacoub MD

A. The Need for Translational Platforms

The Renaissance era redefined scientific methods through the convergence of observation, engineering and mechanistic approaches. Modern sciences have evolved and matured into basic and applied research. As a result, applying breakthroughs of fundamental research into meaningful outcomes in practice is seldom a seamless process.

In medicine, translational research proposes a paradigm shift to bridge basic and applied research by establishing a multidisciplinary vision that combines disparate fields of laboratory science with the applicability of clinical research. In other words, translational research aims to identify the most effective path from laboratory bench to patient bed.

To effectively develop strategies and solve clinical problems, translational research needs multi-skilled ecosystems in which experts of different fields generate and cross-analyze data through various methods and techniques and through feedback cycles between basic, applied and clinical scientists. By its structure and panel of assets, such a translational platform defines and implements the necessary actions, process or methods.

In the context of aortic valve replacement, specific aspects have to be considered to efficiently link clinical needs and expertise with basic and applied sciences. The aim of this chapter is to provide an overview of such aspects, related to the specificity of the aortic valve tissue, its pathologies and diseases, and the resulting clinical need for replacement. The different replacement approaches that have been investigated and applied in recent decades, from mechanical valves to the most recent and promising valve tissue engineering strategy will be introduced in relation to their original clinical objectives and resulting assets and liabilities. Finally, the specific components of a translational platform for aortic valve replacement will be detailed, from assessment criteria, markers and techniques, to preclinical testing.

B. Aortic Valve Characteristics: Structure and Function

Aortic valves perform an extremely complex functional role. Not only do the valves ensure the unidirectional flow of blood out of the left ventricle into the ascending aorta, but they are also crucial for optimising coronary blood flow and preserving myocardial function. It is now widely accepted that the function of the aortic valve is regulated by complex mechanisms that are initially evident during embryonic development and during adaptation of the valve after birth and growth into adulthood. Clinical experience with the Ross Procedure illustrated the importance of a ‘living valve’ (1). The sophisticated function of valve and its durability depend largely on the biological properties of the valve at cellular and molecular level (2). An elegant example of this can be seen in the observations of Hashadigate et al. who shown that the valve anticipates the movement of blood out the left ventricle by beginning to open prior to the forward flow of blood through the valve (3). The role of living cells with the valve has recently been extensively reviewed (4).

Similarly to the three other types of heart valves in the human heart, aortic valves provide the crucial function of ensuring a unidirectional blood flow through the cardiovascular system. However, unlike the right-sided valves of the heart (tricuspid and pulmonary) that direct venous blood to the lungs at low pressure (less than 20 mm Hg), the aortic valve conveys oxygenated blood to the body at a high pressure (~ 120 mm Hg). Considering that heart valves open and close over 30 million times per year and that 3 to 5 litres of blood are delivered by the heart each minute, the aortic valve is subjected to important and repetitive tissue strains and surface shear stresses (5). To cope with such mechanical requirements, the aortic valve possesses a very specific and adaptive architecture that is continuously remodelled by the valve constituting cells.

Anatomic structure

Aortic valves are macroscopically composed of supporting structures in the aortic root, a fibrous annulus connected to the left ventricular outflow tract, and three semilunar cusps (or leaflets) (Fig. 1-A).

figure 1Figure 1. macro- and microscopic structure of the aortic valve. A: the three flexible cusps (C) attached to the aortic root (R). Adapted from (5). B: the three-layered structure of the aortic cusps composed of the fibrosa (F, facing the aortic root), the spongiosa (S) and the ventricularis (V, facing the left ventricle) stained with miller’s elastic (elastic fibers in dark blue), alcian blue (glycosaminoglycans in pale blue) and sirius red (collagen in pink).

Microscopic structure

The aortic root is a fibrous tissue on which are attached the aortic leaflets. Directly above each cusps are three depressions (sinuses of Valsalva), two of which connect to the coronary arteries. The tri-layered structure of the root is akin to large arteries, with an intima, media and adventitia. Similarly, the cell population is comprised of intimal endothelial cells, medial smooth muscle cells and adventitial fibroblasts.

The aortic valve leaflets are thin and compliant tissues that join during diastole to close the valve. They are attached to the annulus of the aortic root in a parabolic fashion, meeting each other at the commissure. They are composed of three layers of distinct organisations and compositions (Fig. 1-B). Facing the aortic root is a fibrous layer, the fibrosa, mainly composed of collagen bundles organized anisotropically in the circumferential direction, and a less organized elastin structure (6). The opposite side of the leaflet (towards the left ventricular chamber) is the ventricularis fibrous layer characterized by a richer content of radially orientated elastin, and a less organized collagen structure. A loose and gelatinous third layer, the spongiosa, separates the two fibrous layers and is rich in glycosaminoglycans.

Each layer plays an important role during the systole/diastole cycle of opening/closing of the valve. Through its collagen content and circumferential organization, the fibrosa provides the strength and stiffness of the aortic valve during diastole, minimizing flopping of the cusp centers, preventing regurgitation and allowing to withstand repetitive biomechanical loads (7). The spongiosa acts as a shock absorber by accommodating the shape changes of the cusp during the valve cycle while the elastin content of the ventricularis allows for valve extension in diastole to maximize coaptation area and valve recoil during systole to minimize surface area (8).

Cellular components

Two principal types of cells are present in the healthy aortic valve: valve interstitial cells (VICs) that are disseminated throughout the entire valve and valve endothelial cells (VECs). Small populations of smooth muscle cells and nerve cells are also present (9-11).

Interstitial cells are responsible for the synthesis and reorganization of the valve extracellular matrix (ECM), allowing to continuously cope with mechanical strains applied on the aortic valve. Although different sub-cell populations have been described (fibroblast, smooth muscle cells and myofibroblast) (12) VICs generally exhibit a quiescent fibroblastic phenotype in the healthy valve. In response to injury, or during growth and remodeling, VIC phenotype changes to an activated myofibroblastic state characterized by an increased production of ECM proteins as well as ECM degrading enzymes (matrix metalloproteinases, cathepsins and tissue inhibitor of metalloproteinases) (8,13,14). Myofibroblast activation can be as well triggered by mechanical stimulations (15,16) and valve stiffness (17). The important proliferation, ECM protein and glycosaminoglycan turnover of VICs in comparison to other cell types in the body (18) tend to confirm that VICs are recurrently activated to repair mechanically induced tissue damages and ensure valve durability.

Endothelial cells, in direct contact with the blood flow, ensure non-thrombogenic properties while regulating immune and inflammatory responses (19,20). VECs are constantly exposed to shear stress of different natures, which have been related to different cellular behaviors (21). Indeed, the venticular side is exposed to a unidirectional shear stress of an averaged 20 dynes/cm2 while the fibrosa surface is subjected to an oscillatory stress of lower magnitude (22,23).

The homeostasis of the leaflets seems to be regulated by a crosstalk between interstitial and endothelial valve cells. For instance, neuronal stimulation of aortic cusps has been shown to be effective only if the endothelial layer was intact (10). Furthermore, studies in vitro have shown the VECs had an effect on VICs proliferation, protein synthesis and decrease in myofibroblast activation (24). Accordingly, loss of VECs by mechanical damage or pathology could result in myofibroblastic activation and ECM remodeling.

Function

During the cardiac cycle, the aortic root and leaflets act in coordination to efficiently close and open the aortic valve and convey a unidirectional blood flow into the aorta (25). During diastole, the aortic cusps are closed by apposition and ensure the filling of the left ventricle. During filling, the fibrous annulus is dilated, which results in a supplementary strain on the leaflets and prevents a prolapse. At the end of diastole, the isometric contraction of the ventricle further increases the annulus expansion that pulls further the leaflets from their commissure. The valve then quickly opens during systole into a circular pattern while annulus retracts and the root resumes a cylindrical shape to ensure an optimal ejection of blood. When the leaflets open, eddy currents form in the sinuses of Valsalva (26) that prevent the leaflets striking the aortic wall and hold them away from the aortic wall in a position promoting valve closure and helping transfer blood in the coronary arteries (27). The closing of the valve is slower than its opening, possibly due to the vortexes that create a pressure gradient across the leaflets and promote a smooth closure (5).

From a mechanical point of view, stresses on the leaflets are most important during diastole and early systole. Bidirectional measurements in vitro of the cusps mechanical properties have shown that they were strongly anisotropic (circumferential strains being much smaller than radial strains) and compliant until a critical strain where they become rigid (28) (Fig. 2-A). This behavior can be related to the structure of the different cusps layers (6,29-31). At lower strain during systole, non-elastic collagen fibers of the fibrosa are crimpled while elastic fibers of the ventricularis layer provide the leaflets elastic response. During diastole, however, collagen fibers unfold and stiffly resist the pressure load (Fig. 2).

figure 2Figure 2. Aortic valve functional biomechanics. A: anisotropic behaviour of the valves illustrated by biaxial mechanical testing of fresh and glutaraldehyde fixed leaflets. Adapted from (28). B: schematic representation of the role of collagen and elastin fibers in mediating leaflets deformations during heart cycle. Adapted from (29). C: Illustration of the mechanical role of elastic fibers in the ventricularis, measured after removal of all connective tissue but elastin from the fibrosa and ventricularis layers. Adapted from (6). D:uncramping of collagen fibers during diastole, sketched and visualized by picrosirius staining. Adapted from (32).

The complex coordination of the aortic valve elements and microstructures during the cardiac cycle is a strong remainder of the necessity to consider multiple fields of research in view of its replacement and proper recapitulation of its function.

C. Aortic Valve Disease and Clinical Needs for Replacement

Although aortic valve diseases (AVD) are responsible for a limited fraction of the deaths caused by cardiovascular diseases (about 2%), their prevalence is important, notably during aging, as 29 % of people over 65 have aortic valve sclerosis (33,34). The presence of a sclerotic aortic valve is not benign and is associated with an increase of the risk of death from other cardiovascular diseases by 50 % (35). In Western countries, valve diseases are mostly degenerative and related to aging. Conversely, in developing countries, valve diseases mostly affect children and young adults (36). In the latter, even if surgical interventions can save lives, pathological remodeling usually occurs, leading to premature valve failure.

The most common effects of valve pathologies are stenosis, insufficiency (regurgitation or incompetence), or both (8). Aortic stenosis, the most common pathology, is characterized by a thickening and stiffening of the valve, leading to a narrowing of the valve and an increased workload of the heart. Aortic stenosis is usually the result of calcification of the cusp tissue. The mechanisms of calcifications are not fully understood, but could be linked with inflammation, lipids accumulation, trans-differentiation of VICs towards osteoblastic phenotype and the presence of specific spherical calcium phosphate particles (37,38-40). Aortic insufficiency is usually the result of a dilatation of the ascending aorta, related to hypertension, aging, or can be caused by rheumatic fever (41).

AVD diagnostic is usually a two-step process. A first hint of valve dysfunction is obtained during physical examination by the presence of a high-pitched murmur (42,43). A confirmation is then established by ultrasound echocardiography, which measures hemodynamic values (peak ejection velocity, valve opening area and transvalvular pressure gradient) and determines the severity of the dysfunction (44,45).

Once diagnostic and severity have been confirmed, the two only therapies available to aortic valve disease are valvuloplasty (repair) and valve replacement. However, valvuloplasty usually relies on the replacement of the aortic root or cusps (46). Without replacement, the development of aortic stenosis symptoms shorten life expectancy to about 3 years (40).

D. Approaches to Reproduce and Replace Aortic Valves

Historical paradigm: reproduce function

Historically, the objective sought through valve replacement was to reproduce and restore the mechanical function of healthy valves. Logically, the first attempts to reach this goal have concentrated on the use of prosthetic devices using the heart cycle and blood flow pattern to act as mechanical valves. As early as 1951, Hufnagel reported his precursor design, consisting in a polyethylene ball entrapped in a methyl methacrylate tube (47), which he implanted in 1952 in a patient thoracic aorta to prevent aortic backflow and treat aortic insufficiency. With the advent of heart and lung machine and open heart surgery in the 50’s, more designs were successfully evaluated as replacement of the aortic valve such as caged-ball (Starr-Edwards valve, 1960 (48), tilting-disc (Björk-Shiley Delrin, 1969 (49) and pivoting bileaflet devices (St. Jude Medical valve, 1976 (50) (Fig. 3).

Nowadays, mechanical prosthetic valves are frequently used in clinic. However, although they are effective, structurally sound and durable, they suffer from diverse shortcomings (51). Their components are recognized by the body as foreign material while their geometry in the blood flow results in non-physiological shear stresses that induce platelet lysis and give rise to thromboembolic complications. As a result, and despite more than 50 years of research, patients require life-long anticoagulation therapy, which are limiting and increase risk of serious bleeding events (52-54)

In parallel to the development of mechanical valves, tissue valves homografts from cadaveric aortic valve were investigated as implants by Dr. Ross in 1962 (55,56) followed by xenografts from porcine (Hancock glutaraldehyde-fixed valve, 1969 (57) and bovine (Carpentier Edwards pericardial valve, 1976 (58) origin (Fig. 3). These tissue valves advantageously solved the hemocompatibility issues of mechanical valves by providing more physiological hemodynamics and cancelled the need for anticoagulant therapies. However, being composed of fixed tissues, without any living cells, they have a strong risk of structural deterioration. In addition, the lack of living cells prevents valve calcium homeostasis and favours calcium nucleation, nodule formation and calcification (59) that ultimately lead to more re-operations (60). In fact, the tissue valves are now increasingly considered by clinicians to “replace one disease with another” (41, 61).

figure 3Figure 3. Historical overview of mechanical and tissue valves. A: Hufnagel design (1951). Adapted from (48). B: Starr-Edwards caged-ball design (1960). C: Björk-Shiley Delrin tilting-disc design (1969). D: St. Jude Medical pivoting bileaflet design (1976). Adapted from (62).E: Homograft redissected after 8 months (1962). Adapted from (55). F: Hancock aortic porcine valve (1969). G: Carpentier Edwards pericardial bovine valve(1976). Adapted from (54).

Another critical drawback of either mechanical or tissue valve is the passive role that they exert in restoring the valve function. Since they are not living tissue, they lack the ability to grow, repair or remodel. While this passivity is acceptable for sedentary or older patients, it becomes problematic for young and active adults or for infants and children. To compensate for the growth of children conduits, the only solution for valve replacement is to implant larger prosthesis than ideal and/or re-operate frequently to resize the conduit (63). Furthermore, as xenogenic tissue valves in children are notorious for rapid calcification and resulting early reoperation, mechanical valves are used instead together with anticoagulation therapy and the associated risks for children (Fig. 4).

figure 4Figure 4. Drawbacks of prosthetic aortic valve replacements, especially for young patients. A: Mechanical prostheses perform better than tissue valves with regard to mortality after implantation, contrarily to bleeding events, which are lower for tissue valves. Adapted from (53). B: Risks of reoperation and major bleeding events oppositely vary with age at implantation for mechanical and tissue valves. C: Structural deterioration of tissue valves is related to the age at implantation. In young people, use of a biological valve will most probably result in multiple reoperations. Adapted from (61).

Paradigm shift: reproduce function with living tissues

The drawbacks and difficulties associated with mechanical or decellularized tissue prosthesis, especially in paediatric patients, have motivated the development of other strategies aiming not only to restore proper aortic valve function, but as well its living regulatory and adaptive properties.

Autografts

Surgically, a technique has been proposed as early as 1967 by Dr. Ross, consisting in replacing the aortic valve with the patient’s own pulmonary valve, which can in turn be replaced by a decellularized tissue valve (homograft) (64). This pulmonary autograft, commonly termed “Ross procedure”, circumvent the limitations of prosthetics in the aortic position by advantageously transposing them to the less demanding pulmonary position (Fig. 5-A). This operation has been performed for decades and although clinical studies indicate its superiority to homografts in adults (1) (Fig. 5-B), its use still remains controversial (65). For infants and children, the advantages of the Ross procedure as cell-containing living tissue being able to grow with the patient have made it the operation of choice.

Nevertheless, clinical acceptance of the Ross procedure is still not consensual due to the technical difficulty of the operation and a risk of aortic root dilatation instead of growth leading to valve insufficiency(66-68), especially in young patients with bicuspid aortic valve (69).

figure 5Figure 5. The Ross autograft procedure circumvents the limitations of prosthetics in the aortic position. A: The surgical procedure as sketched by the Professor Ross in 1967. Adapted from (64). B: Survival and freedom from need of aortic valve reoperation after autograft procedure on patients (18 to 60 years old) is enhanced when compared to homograft. Adapted from (1).

Aortic valve tissue engineering

To overcome the limitations of prosthetic and surgical approaches, a multidisciplinary strategy has emerged in the past two decades that aims to combine the beneficial aspects of living transplants (biocompatibility, growth, repair and remodeling) with a proper and controlled reproduction of the aortic valve structure and function. Heart valve tissue engineering follows the general paradigm of tissue engineering that was first defined in 1988: “the application of principles and methods of engineering and life sciences towards the fundamental understanding of structure-function relationships in normal and pathological mammalian tissues and the development of biological substitutes that restore, maintain or improve tissue function” (70). In theory, aortic valve tissue engineering could be particularly beneficial for young patients who require valve growth and cannot tolerate the side effects of non-living prosthesis (71). If this population is mostly confined to birth malformations in the western world (about 2 % of life birth), it could amount millions in developing countries due to higher incidence of rheumatic fever (52).

Although different methodologies have been applied and evaluated for aortic valve tissue engineering, an accepted paradigm comprises a natural or synthetic scaffolds that is pre-seeded with cells, followed by an in vitro step of tissue formation conducted in a bioreactor, and subsequent in vivo implantation allowing for tissue growth and remodeling (72). This global approach allows for extensive variations of parameters such as scaffolds properties, architecture and degradation, cell type and in vitro stimulations. The resulting key physiological processes evaluated are cell proliferation and migration, ECM production and organization, tissue remodeling and recruitment of host cells, either inflammatory or precursor, after implantation. Interestingly, the recruitment of host cells by unseeded scaffolds, designed to attract endothelial and mesenchymal circulating precursor cells in vivo and orchestrate tissue formation is as well considered as tissue engineering (Fig. 6).

figure 6Figure 6. The aortic tissue engineering paradigms. A conventional approach (pathway A) associates cells, scaffolds, in vitro maturation and in vivo remodeling; while a modified strategy recruits host cells through a functionalized scaffold (pathway B). Reproduced from (72) (Open Access).

Although this chapter will introduce the different aspects of aortic valve tissue engineering in the following paragraphs, the reader will find more details in extensive reviews on this subject (73-76).

Cell supportive matrices

The first important component in aortic valve tissue engineering is the development of a cell supportive scaffold which shape mimics either a valve leaflet (77-79) or a full conduit comprising leaflets and aortic root (80). Both approaches have advantages and limitations. A semi-lunar valve leaflet shape is certainly of easier conception and realisation but forecasting exact dimension for a given patient is difficult and might result in improper coaptation and closure after suturing to the existing aortic root. Conversely, a full conduit simplifies hemodynamic issues, but is technically difficult considering that the aortic root and leaflets are of different organizations and mechanical properties (81). Regardless of the approach considered, the tissue engineered valve must resume a perfect hemodynamic function immediately upon implantation and keep relatively constant mechanical properties over time despite tissue remodeling and eventual scaffold resorption.

The role of the scaffold is therefore complex. It must possess a number of qualities that will allow and guide the formation of a new tissue such as biocompatibility, elevated and interconnected porosity (to allow cell growth, nutrient diffusion and waste removal), cellular attachment, migration, proliferation and extracellular matrix formation; while providing an initial mechanical support after implantation that must gradually be compensated by the neo-tissue as the scaffold is degraded and resorbed.

Intuitively, the best scaffold to comply with these requirements would be the native aortic valve extracellular matrix itself. In theory, using detergents washings, cellular content can be removed from valve tissue to obtain an intact extracellular matrix structure of low immunogenicity. However, despite experimentation of various decellularization protocols using detergents, enzymes or nucleases (82) (83), some features of the extracellular matrix such as collagen pattern, distribution and glycosaminoglycan content can be modified (84) and lead to altered biomechanical and hydrodynamic properties (85,86). The balance between extent of decellularisation and conservation of mechanical properties is further complicated as remaining cellular debris are known to elicit a strong immune response and promote calcification (87). Despite these difficulties, decellularized valves in pulmonary (88,89) or aortic (90) positions have reached clinical evaluations in adults and children. Although recellularization of the grafts was marginal, the outcome for adults was positive in regards of valve function. Conversely, it resulted in the tragic death of 3 out of 4 children who were treated in the study, due to a swift and strong inflammatory reaction (87). As a result, the prospective use of decellularized valves has been set back and current strategies focus on improving recellularization in vivo to enhance biointegration (91-93). Without minimizing the potential of such an approach, the use of decellularized matrices will nonetheless always rely on a strong supply of human valves of good quality, which is lacking (Table 1).

Table 1. Comparison of cell supportive matrices used in aortic tissue engineering.

table 1

In view of overcoming the limited access to native valve extracellular matrix while controlling a wide range of properties, synthetic or natural biodegradable polymers have been as well investigated. Synthetic polymers have the advantages of limitless supply, well controlled and tailored intrinsic properties and formulation ease towards highly porous and interconnected structures through various manufacturing processes such as salt leaching (94), knitting (95) stereolithography (96), electrospinning (97) or jet-spraying (98, 99). On the other hand, they must be biocompatible, resorbable without major local side effects, allow cell attachment, migration and proliferation, differentiation, ECM formation and remodeling and exhibit mechanical and structural properties comparable to aortic valves, which narrows the list of potential candidates (100). As a result, various polyesters (PGA, PLLA, PLGA, P4HB, PHA and PCL) have been used, without consensus on a unique polymer presenting all necessary requirements (83,101-107). Indeed, each polymer presents different interactions with cells through intrinsic surface properties. Additionally, sponge-like or fibrillar and nanofibrillar formulations of varying porosities influence cells migration, deep colonization of the structures and synthesis of a homogeneous tissue (99, 108). As degradation occurs through hydrolysis at various rates (from weeks for PGA to years for PCL), acidic molecules are released that can influence cell fate over time and induce local tissue inflammation. Concurringly with degradation, polymer matrices mechanical strength and rigidity are decreased while valve function must remain constant over time. This central requirement implies to find the perfect balance between tissue formation, remodeling and polymer resorption. So far, the most advanced results in large animals in vivo have been obtained with fast degrading polymers (PGA and P4HB) cultured in vitro during 2 weeks with myofibroblasts and endothelial cells or associated with bone marrow cells prior implantation as pulmonary valve replacement (95,109).

As an alternative to synthetic molecules, natural polymers issued from ECM have the advantages of being inherently biocompatible and degradable through enzymes, which allows for a cell-driven remodeling. The most commonly used proteins are type 1 collagen and fibrin, which form a fibrillar hydrogel spontaneously at neutral pH or through thrombin polymerisation, respectively. The direct encapsulation of cells within the hydrogels is another advantage over synthetic polymers, which rely on cellular invasion to achieve cellularity of the constructs. Furthermore, all mesenchymal cells seem to flourish within such structures (110-112) that can easily be shaped as aortic valves by injection moulding (113,114). However, these hydrogels lack the mechanical strength required for valve replacement. As a result, dynamic conditioning has been successfully evaluated to induce sufficient strength prior implantation (115). However, severe gel contraction induced by the cells resulted in a quasi-disappearance of the cusps after three months (116). In the future, natural proteins might find a suitable role in valve replacement in combination with synthetic polymers, to improve cellularity while maintaining mechanical strength (117, 118). Nevertheless, for the body, a fibrin matrix still represents a wound granulation tissue that needs to be remodelled in fine in a scar tissue dissimilar to the native one; therefore, the biological rationale of using fibrin gels as substrate for functional tissue formation in vivo remains to be demonstrated. A promising alternative could be to use fibrin matrices as a template for cells to form a controlled collagen matrix that can be subsequently implanted after decellularisation (119).

Although the vast majority of valve tissue engineering approaches associate cells with supportive structures in vitro and stimulates them through chemical and/or mechanical conditioning prior in vivo implantation, cell-free strategies are as well investigated to design smart materials that could attract and retain host circulating progenitor cells and form a new valve in situ. As synthetic polymers lack the appropriate signalling molecules naturally present in the ECM for cell recruitment, the challenge here is to integrate this bioactivity into the materials. Various bioactive cues and moieties can be envisioned to capture, recruit and guide differentiation of progenitor cells, such as antibodies (CD34 and vascular endothelial-cadherin (120,121), aptamers (122), and growth factors (123) for endothelial progenitor cells. Similarly, coatings with mimetic peptides or isolated ECM proteins are known to promote cellular adhesion (124), but could as well direct biological events such as apoptosis (125) or cell differentiation (126). It should be noted that in the context of in situ valve engineering, the polymeric matrices will require appropriate mechanical properties and slower degradation rates as compared to in vitro cellularized constructs, to allow for cell recruitment, differentiation and tissue formation.

As promising as these precursor strategies might be to bypass the in vitro stage of cellularisation and conditioning, they are still in their infancy and the in vitro association of cells with supportive structures remains the most employed approach.

Cell sources and Challenges

Optimally, the cells used for association with supportive structures and development of a new valve tissue should be non-immunogenic, proliferative, easy to harvest and either keep their specialized function or be able to gain such a specialization through differentiation. The most logical cells to harvest are autologous cells from the patient to be treated, to remove any risk of immune rejection. The use of autologous aortic valve cells being impossible, other cell sources have been evaluated, such as fibroblasts, myofibroblasts and endothelial cells isolated from blood vessels or subdermis (77,80,95,127-129). However, autologous material from patient requiring heart valve replacement is usually too sparse or with diminished proliferative capacity, especially for elderly patients. As a result, the use of autologous mesenchymal stem cells (MSCs) has been actively investigated, either from bone marrow or adipose tissue origin (99,130-133). Additionally, specific cell sources for pediatric patients, such as Wharton jelly, umbilical cord blood, peripheral blood, (129,134,135) and prenatal cells issued from chorionic villi (136) and amniotic fluid (137,138) have shown proof of concept results.

Important problematic linked to ce llularization of constructs in vitro are cell retention and endothelial function. Retention of cells after implantation has not been extensively investigated, although a study indicated that canine bone marrow cells associated with decellularized porcine pulmonary valve were still present after 3 weeks of implantation and could have therefore contributed to the regeneration process (139). Endothelial function of the engineered valve is critical to provide anti-thrombotic properties and ensure survival of recipients. Isolation of endothelial cells from blood vessels or differentiation towards endothelial phenotype of MSCs by chemical and cytokine cocktails (140) is possible while re-endothelialisation of supportive structures (141) seem to improve endothelium coverage and prevention of thrombus formation as was shown within 3 months after implantation in pulmonary position in sheep (142). In humans, reconstruction of right ventricular outflow tract of adults undergoing the Ross procedure performed with re-endothelialized pulmonary allografts showed no calcification and thrombogenesis after 10 years (143,144). More importantly, re-endothelialisation with endothelial progenitor cells from peripheric blood of decellularized pulmonary valve allografts implanted in 2 children seemed to have prevented valve degeneration over 3.5 years of implantation (135).

Another important aspect of the in vitro expansion and association of cells with scaffolds is the conservation or acquisition of a phenotype similar to native valve cells in vivo. While cultured MSCs display a spindle morphology characteristic of fibroblasts and myofibroblasts, express markers shared by VICs (72) and have the ability after implantation to form endothelial and interstitial-like layers (145-148), concerns remain regarding the stability of the acquired phenotype and potential unwanted fibrotic overgrowth (149). Similarly, while myofibroblastic and endothelial cells can be maintained in culture, lack of phenotypic control once implanted can result in formation of fibrotic-like tissues causing retraction and regurgitation of the engineered valves (128,129,150).

With regard to cell origin, it should be noted that animal cells are often used in prevision of proof-of-concept animal experiments. However, such cells do not always behave similarly to human ones. For instance, ovine vascular-derived cells show different sizes, proliferation rates and contractile behaviour as compared to human cells of similar source, which result in a stronger scaffold contraction (73). As a result, specific protocols have to be developed for each cell type and origin, and should always be validated again with human cells.

Regardless of cell sources, the favoured way to induce a strong cell commitment toward a specific phenotype and monitor undesired orientations consist in mimicking the natural valve mechanical and flow environment and pre-conditioning the tissue engineered constructs in vitro prior implantation.

Bioreactors and conditioning

The design and use of bioreactors for conditioning heart valve engineered tissue find multiple rationales in biological mechanisms. Indeed, during heart valve adaptation or repair after injury, VICs are known to change phenotype and become activated in myofibroblasts (151). Such activation has been shown to be regulated by environmental stimuli such as dynamic loading of the valve (14,152), profibrotic cytokines (transforming growth factor-beta 1) (153) and elevated matrix stiffness (13). Furthermore, mechanical conditioning can influence extracellular production as was demonstrated with native porcine aortic valve leaflets exposed to hypertensive cyclic pressure (154) or isolated circumferential cyclic stretch (155). The former study indicated an increase of collagen and GAGs synthesis related to magnitude and frequency while the later showed an increase of collagen and a decrease of GAGs after stimulation.

Bioreactors allowing a variety of conditioning strategies in terms of cyclic flow and pressure changes (95,156-159), strain (132,160,161) and shear stress (162) have been developed (Fig. 7) and many studies have highlighted the benefit of mechanical and flow conditioning. For instance, a moderate pulsatile flow reached in small increments over endothelial cells seeded on decellularized pulmonary valves revealed of better quality for cellular proliferation than a rapid increase to physiological flow (141). Straining of tissue engineered valves (163) and collagen cross-linked constructs seeded with smooth muscle cells (164) resulted in more pronounced and organized tissue formation with superior mechanical properties as compared to unstrained controls. Similarly, collagen synthesis by porcine VICs was shown to be dependent on degree and duration of stretching (165) while stretching of adipose and bone-derived MSCs cultures on substrates stretched at 14 % for 3 days showed an increase of collagen synthesis (132). Dynamic straining (4% for up to 10 days) of myofibroblasts-seeded polymer constructs indicated a decrease of collagen production but an increase of collagen cross-links, GAGs production and remodeling markers (166). Prolonged dynamic straining (4% for 2 weeks) were shown to induce the production of collagen with a higher degree of crosslinks while inducing a specific cell orientation parallel to strain direction and a layered collagen organization (167). Interestingly, excessive dynamic strain magnitudes (8%) resulted in a decrease of the mechanical properties of the constructs while intermittent dynamic straining seemed to improve tissue formation and reduce the risk of deterioration (168).

Although the optimal conditioning protocol will depend on different parameters linked to the cell phenotype, the type of scaffold used and the magnitude and type of mechanical cues, the length of stimulation that has been generally applied in various studies is 3 to 4 weeks (80,136,145,160).

figure 7Figure 7. Examples of the various bioreactor designs that have been created to replicate heart valves hemodynamics and biomechanics and employed to condition tissue engineering heart valves. Adapted from (158-160,169).

Validation of tissue engineered valves?

Various animal experiments performed mostly on sheep have clearly demonstrated the proof of concept of tissue engineered valves (Fig. 8) (72,77-79,145). However, although most of these experiments have been conducted in the pulmonary position, which is biomechanically less demanding and therefore more attainable than the aortic, translation to human has not yet been achieved. The few experiments performed in human with decellularized allo- or xenograft (170) have indeed showed either promising results (135,143,144) or catastrophic outcomes (87).

figure 8Figure 8. Examples of tissue engineered valves that were implanted in pulmonary position in sheep. A: polyglycolic acid and poly-lactic acid fibrillar valves seeded with ovine mesenchymal stem cells prior implantation (top) and echocardiogram along the short axis after surgical implantation showing a triangular orifice during valve opening (bottom). B: polyglycolic acid and poly-4-hydroxybutyrate fibrillar scaffold seeded with ovine myofibroblasts and endothelial cells and conditioned in a bioreactor for 2 weeks (top left); macroscopic view of the tissue engineered valves after 6 weeks of implantation (top right) and echocardiography along the long axis after 8 weeks of implantation showing the valves closed and opened positions (bottom). Adapted from (129, 145).

This discrepancy underlines the need for integrated translational tools and platforms. Many unanswered questions remain in regards of biological mechanisms, interactions of cells with supportive materials, animal models and discrepancy between animal and human cells behaviour, before successful human implantation can be achieved. Furthermore, criteria and quality markers need to be established to determine when a tissue engineered heart valve can be implanted in a patient, with a satisfactory prognosis for long-term survival. Similarly, predictive models issued from animal experiments and from in silico models of tissue growth, remodeling and clinical epidemiology remain to be developed.

In this context, it is vital to find and establish suitable criteria to assess the efficacy of engineered aortic valves and translate in vitro and animal experiments to human prognostic, as will be presented in the following part of this chapter.

E. Translational Platform: Establish Criteria and Quality Markers

The need of a translational platform for aortic valve replacement is strongly linked to the valve tissue engineering approach. While mechanical and biological cardiac valve prosthesis design, production and risk management can be evaluated through International Standard Organization (ISO) guidelines (ISO 5840:2005, “Cardiovascular implants — Cardiac valve prostheses”), tissue engineered valves have to comply with more requirements, being living tissues. The need exists for engineered valve to evaluate their functionality prior implantation through quality criteria indicative of a positive clinical outcome.

So far, the path to evaluate aortic tissue engineered valve has been to primarily focus on pulmonary valve engineering and replacement in conjunction with Ross procedure, as pulmonary valves share the same structure as aortic valves but are less hemodynamically demanding (5). The constructs are first evaluated in vitro for biological and biomechanical properties, often in bioreactors allowing hemodynamic stimulations (116,146,171). They are then applied in small animal models, such as rabbit or rats to provide information on biocompatibility, or in large animals such as sheep for functionality assessment. The next evaluation is in adult humans, as replacement of the pulmonary valve during the Ross procedure, prior to the final evaluation in children that will require valve growth.

Before reaching evaluation in infants and child, many steps must therefore be validated. But the question remains as to the validation criteria and markers allowing to indicate a satisfactory prognosis for heart valve replacement in patients. From the previous studies outlined in this chapter, the function of aortic valve is correlated to its structure; therefore, validation criteria of engineered valves must be orientated towards physiological structure and function, and assessed always before and after animal implantation. Accordingly, a set of major criteria to consider for the clinical relevance of engineered construct can be defined as follows:

  • shape and structure
  • biocompatibility, biodegradation
  • cellular biomarkers, tissue formation and quality
  • remodeling towards native structures, structural integrity and endothelial function
  • hemodynamic performances and biomechanical properties
  • functional assessment and durability

These criteria and markers must be established both in vitro and in vivo, in long term follow-up preclinical studies. The provided data can then be used to derive predictive models in humans in regards of:

  • hemodynamic and functionality
  • in vivo remodeling, repair and growth
  • lack of pathological behaviour of the implanted valve (calcification, stenosis)

Finally, the collected data and model predictions must be compared to the function observed in native aortic valves and with mechanical or biological prosthetic valves (172). A visual summary of such a translational approach is presented in Fig. 9.

figure 9Figure 9. Proposed translational approach for tissue engineered aortic valves. Various criteria to consider can be defined in vitro and in vivo to ultimately provide predictive models of the engineered valves functionality in humans and therefore evaluate clinical performances.

To provide an overview of this translational approach, the last part of this chapter will describe in more details the different quality criteria/markers and the corresponding invasive and non-invasive tools that can be applied for the development of a translational platform for aortic valves replacement.

Evaluation Criteria in vitro: supportive matrices and cells

Structural characterization

With regards to parameters and criteria of the cell supportive structures that can be evaluated at early stage of development in vitro, shape and structural organisation are the most straight forward. While shape and exact dimension of the constructs can be easily measured directly or through macroscopy, microscopic structure and organization are commonly characterized with scanning electron microscopy (SEM) followed by image analysis (98). Such analyses are useful in developing matrices that aim to mimic the native anisotropic organization of aortic valves ECM (99,173).

Porosity, pores sizes and pores interconnection of the matrices directly relate to cellular colonization and tissue in-growth and as such can be used as predictive tools. A pore size value of 10 micrometres has indeed been proposed as threshold permitting cellular infiltration (174,175). While gravimetric measures allow to calculate porosity, SEM images analysis (176) or mercury intrusion porosimetry (99,177) can be used to evaluate pores sizes and interconnection. However, results obtained with these methods should be analysed with caution as they do not provide fully accurate results. Image analysis is rendered imprecise by the depth of field of scanning electron microscopes that prevents to isolate pores in a single plane, while the pressure applied on soft matrices to force mercury intrusion will induce structural deformations. As such, these methods remain useful for crossed comparison of various supportive structures and should always be confirmed by cellular infiltration assays.

Biocompatibility

Biocompatibility of manufacturing materials is a mandatory criterion that is usually evaluated both in vitro and in vivo. Direct cell toxicity of the materials and leached out or degradation products can be determined using in vitro cell viability assays based on the quantification of cellular metabolic activity (MTT, MTS assays or alamar blue) (176,178). Direct visualization of cell mortality is as well possible through life/dead assays that clearly differentiate metabolically active cells from dead cells with ruptured cytoplasm under fluorescence microscopy (92,179).

In vivo studies provide further information on the foreign body reaction of the native immune system. Small animal models, such as rabbits or rats, are implanted with the materials to minimize the costs and ethical issues associated with the use of large animal models. Subcutaneous implantations during various times allow to observe capsule formation and identify infiltrating cells such as inflammatory cells, macrophages or fibroblasts by mean of histology and immunohistochemistry (91). Systemic toxicity is indicated by weight changes of the animals after implantation while remote organ toxicity can be evaluated by collecting liver, heart, and kidney samples from each animal and controlling their histological structures (92). Aside from subcutaneous models, alternatives have been proposed to assess biocompatibility in environments closer to the heart valve such as implantation of materials in the blood flow of rats abdominal aorta (180).

Degradation

Similarly to biocompatibility, materials biodegradation is an important parameter to take into account during development of cell supportive structures, in prevision of its replacement by neo-tissue after implantation.

For synthetic polymers, in vitro assays can be performed by incubating the structures in buffers either mimicking physiologic conditions or allowing an accelerated hydrolytic degradation (178). Mass loss, molecular weight decrease, measure of degradation products, crystallinity, and evolution of macro- and microscopic morphology by SEM over time provide valuable information of polymer degradation rates. Variations of mechanical properties over time indicate degradation as well and are particularly useful when combined with in vitro cell culture to infer that ECM synthesis, and hence tissue formation, is sufficient to maintain the constructs mechanical properties. However, degradation rates obtained in vitro cannot be readily translated to the in vivo situation and should always be confirmed.

For matrices made of natural materials, biodegradability is mostly assessed directly in vivo in subcutaneous small animal models (91,181). Histological evaluations of cellular infiltration and type of cells associated with quantification of the remaining implant area over time are then reliable indicators of biodegradation.

It is worth noting that the characterization of polymers biocompatibility and biodegradation are especially valid for novel compounds or compositions. Over the past decades many biodegradable polyesters have already been validated for human use and approved by the FDA (100), such as poly(glycolide), poly(lactic acid), poly(lactic-co-glycolic acid) and poly(ε-caprolactone), which facilitates their use in aortic valve engineering.

Mechanical properties

Although the most common strategy for aortic tissue engineering relies on in vitro cell culture and bioreactor conditioning to improve mechanical properties prior implantation, the gross properties of the supportive structure should optimally mimic some of the valve features such as anisotropy. Moreover, in the specific in situ approach where the implants are colonized by the patient own cells after implantation, the supportive structures should be sufficient to withstand the ranges of stresses and pressures applied on a native aortic valve immediately after implantation and remain until complete tissue formation and remodeling.

Gross mechanical properties of supportive structures are most commonly evaluated by tensile assays. The force necessary to produce a given deformation rate of the sample is recorded and stress/strain curves calculated from the measured force and the sample cross-sectional dimension. From this data, the tissue strength, stiffness, relaxation and elongation before rupture can be calculated and compared to native tissues measured in a similar way (99,182). The material stretching can be applied in one direction (uniaxial), or simultaneously in perpendicular directions (biaxial). Anisotropic properties of the tested structures can be calculated using both approaches, but biaxial tests are closer to physiological situations and as such should be preferred (183, 184). For materials derived from natural ECM, additional information concerning fibre alignment and deformation during stretching can be obtained by combining tensile tests with the quantification of polarized light angles (184,185).

As a guideline, computational modelling can as well be employed to determine specific range of mechanical properties, anisotropy and shape that would allow proper radial stretch and coaptation of the supportive structure in either pulmonary or aortic conditions for instance (186).

Aside from measure of gross mechanical properties, local stiffness can as well be measured by atomic force microscopy. This is particularly relevant to compare different structures to the local mechanical properties of the different valve layers and hence mimic the multifactorial properties of the ECM (176).

The in vitro association and culture of cells with supportive matrices is crucial to identify cellular interactions with the supportive structure, allow the formation of ECM, and provide endothelial functions and sufficient hemodynamic/biomechanical performances.

Cellular behavior

The ability of cells to recognize the provided structures as suitable for their proliferation is usually assessed by classical biochemical techniques. MTT/MTS assays (132) or DNA quantification (99) are commonly used to measure proliferation.

Cellular penetration and colonization of the constructs is an important criterion to assess as it will reflect the possibility of forming a homogenous tissue prior and after implantation. Histological preparations and sectioning of the constructs at different times followed by image analysis are effective to determine the rate of cellular invasion. Cell nuclei are stained using either fluorescent (DAPI) or colorimetric (Movat pentachrome) dyes and cell distributions within the matrices can be deduced (99,132,187). To further evaluate that cellular colonization is homogeneous throughout all available volume, three dimensional reconstructions can be performed by assembly of multiple contiguous cross sections (Fig. 10-A) (188).

Cellular morphology and organisation can be guided by mechanical stimulations (189) and by the type and structural features of supportive matrices, such as anisotropy (99,173). Scanning electron microscopy analyses provide convenient information of cells located at the surface of the matrices, but not of cells within (7). To visualize these cells, confocal microscopy can be used after fluorescent labelling of the cytoskeleton with phalloidin for instance (Fig. 10-B). However, the attainable depth depends strongly on the scaffold material and remains confined to hundreds of micrometres at best.

figure 10Figure 10. Examples of techniques used to characterize cellular behavior. A: analysis of cell colonization of tissue engineered heart valve by contiguous histological cross sections and 3-dimensional reconstruction. B: Phalloidin staining and confocal observations of mesenchymal stem cells cultured on nanofibrillar matrices of increasing anisotropy. Adapted from (99,188).

Evolution of the cell phenotype during culture with the supportive structures should be evaluated over time, to assess uncontrolled differentiation or transdifferentiation when stem or mesenchymal cells are used. The expression of stem or differentiated cell surface markers can be followed over time by immunohistochemistry on cross sections (99), or by flow cytometry (fluorescence-activated cell sorting, FACS) when cells can easily be extracted from the supportive matrices (179). The acquisition of a myofibroblast phenotype is of particular importance as VICs are naturally activated into myofibroblasts during valve growth or remodeling (13,190). Myofibroblast activation of fibroblastic cells in general and VICs in particular is known to be mediated through physical (substrate rigidity) cues in their microenvironment (13,191,192,193) and as well through mechanical conditioning (155). However, excessive and uncontrolled activation of various cell types can lead to excessive deposition of unorganized ECM of fibrotic properties (194,195). Immunohistochemical stainings of sections for alpha-smooth muscle actin (α-SMA), desmin and vimentin indicate myofibroblast-like cells (99,106,196), while western blotting of α-SMA can be used in a semi-quantitative fashion (155).

ECM synthesis and remodeling

The composition and organization of the cell-produced ECM is fundamental for the structural integrity and biomechanical properties of aortic tissue engineered valves after implantation and for its long-term performance, similarly to native valves. It is therefore of high interest to quantitatively and qualitatively assess ECM components deposited by the cells during culture and compare them to the native tissue.

Cross-sections of the constructs at different culture times and histological and immunohistological methods are commonly used to evaluate the deposited ECM within the supportive structures. Classical colorimetric stains such as hematoxilin and eosin are useful for the general ECM distribution and morphology for instance (80, 106, 155) while ECM composition can be discriminated using specific antibodies directed against collagen, elastin and chondroitin sulfates (99, 146). Particular staining procedures such as picrosirius red (155, 196) and Masson’s trichrome stains (80) or alcian blue (182) are also useful to stain collagen or glycosaminoglycan content, respectively. Movat pentachrome allows one to visualize proteoglycans (in green), collagen (in yellow), and elastin (in black) on the same section (14, 145, 196). ECM-related gene expression can also be detected using in situ hybridization (197). With regard to matrix remodeling, information is provided by immunohistological techniques targeting matrix metalloproteinases (MMPs), cysteine endoproteases (cathepsins) and tissue inhibitors of MMPs (TIMPs) (196-198).

All these characterizations require the destruction of the analysed sample by embedding, sectioning and staining. In prevision of quality control of the constructs prior implantation, non-destructive methods for ECM formation analysis have been developed. For instance, multiphoton excitation microscopy allows to visualize collagen structures and elastin fibers with great details at high resolution (Fig. 11) (199, 200). This technique based on the absorption of multiple photons carrying approximately less energy than necessary to excite a molecule results in the subsequent emission of a fluorescence photon at a higher energy. The infrared excitation used results in deeper penetration in the sample than confocal microscopy while being of low cytotoxicity (201). Collagen-specific fluorescent probes have as well been developed to evidence collagen synthesis and organization under confocal microscopy (202, 203).

figure 11Figure 11. Application of multiphoton microscopy to visualize collagen (in red) and elastin (in green) in a tissue engineered heart leaflet. The serial images obtained at different depth can be stacked to reconstruct a 3-dimensional view. Adapted from (199).

It is worth noting that the use of polymer matrices often limits histological, immunohistological, confocal and multi-photon techniques due to polymer dissolution in histological solvents, antibodies retention, light diffraction and auto-fluorescence that will interfere with samples preparation and examination.

Quantitative assessment of ECM formation can be obtained by biochemical assays specific for collagen, GAGs and elastin followed by normalization per cell using DNA content (80, 84, 106, 134, 146). Quantification of different collagens, lysyl oxydase and tropoelastin mRNA by real-time polymerase chain reaction provides as well information on ECM proteins gene expressions levels during in vitro culture (132). Alternatively, collagen formation can be precisely quantified by radio-isotopic detection of (3H)-proline incorporation (132, 165). Aside from the amount of ECM proteins, collagen and elastin crosslinks can as well be monitored by respectively measuring pyridinoline and desmosine in protein hydrolysates using liquid chromatography and mass spectrometry (132). The amount of crosslinks of collagen have been demonstrated to be directly correlated with the resulting tissue strength and stiffness (7) and as such is an important criteria to consider.

The formation of an ECM comparable to the native aortic valve is crucial for the biomechanical performances of the constructs prior and after implantation. Furthermore, not only the repartition and amount of ECM proteins are of importance, but as well their maturation and architecture as a whole. Although tissue engineered constructs in vitro can present a tissue formation of composition similar to native heart valve leaflets (106, 136), different aspects are still not fully satisfactory. For instance, anisotropic organisation of collagen fibres is not yet achieved. It was once thought that once implanted in the aortic valve hemodynamic environment, collagen will be able to mature and gain increased alignment (13). Elastin content is also problematic and not sufficiently synthesized in engineered valves (73, 80), even though its critical role in providing a proper biomechanical function, ensuring optimal valve closure and preventing valve replacement failure is acknowledged (204, 205).

Endothelial function

Endothelialisation of engineered aortic valves is a crucial parameter to assess prior implantation, in view of decreasing thrombogenicity after implantation. Immunohistochemical detection of CD31, vascular endothelial grow factor receptor FLK-1 and von Willebrand factor (vWF) is useful to determine the presence of an endothelial layer at the surface of the engineered constructs (Fig. 12-A) (135, 141, 142). The detection of endothelial nitric oxide synthase (eNOS) production by immunohistochemistry provides as well an indication of the endothelial layer functionality (134, 141, 142).

In conjunction with immunohistochemical techniques, SEM microscopy allows to determine the homogeneity of the endothelial coverage and its density (Fig. 12-B) (134).

figure 12Figure 12. Detection of endothelial cells on tissue engineered heart valve. A: immunohistochemical staining targeting CD31 (top left, red), vascular endothelial grow factor receptor FLK-1 (top right, red) and von Willebrand factor (bottom left, green), compared to a pulmonary artery wall stained for von Willebrand factor (bottom right, green). B: Scanning electron microscopy of the endothelial surface of tissue engineered leaflets submitted to different mechanical conditionings during in vitro culture. Adapted from (134, 135).

Evaluation Criteria in vivo

Biomechanical performance

Considering the critical and vital mechanical and hemodynamic characteristics of aortic heat valves, it is mandatory to validate that the implanted constructs are able to provide functional closing and opening motions and cope with the aortic valve environment, at least immediately after implantation and prior to a complete tissue formation and remodeling.

The same tensile tests as for the characterization of acellular supportive structures are commonly used to determine tissue strength, extensibility, stiffness and anisotropic properties (80, 136, 182). In addition, the evolution of the constructs mechanical properties over culture time is a good indicator of ECM production and quality (7, 99). In this regard, measure of anisotropic properties of the tissue engineered aortic valves should preferentially be performed through biaxial testing to ensure that mechanical behaviour in radial and circumferential direction are not inconsistent with those of native valves (80, 183).

The evolution of the constructs mechanical properties combined with the characterization of ECM production at different culture times could eventually lead to models of tissue formation and biomechanical properties in function of scaffold and cell type (81, 206). Although those approaches are still in their infancy, the prospects are high to allow the preparation of constructs perfectly matching the aortic valve.

Hemodynamic performance

To validate hemodynamic performances, bioreactors have been developed that simulate the pressure cycles applied on the aortic valve (159) or replicate the complete physiological heart cycle and pulsed aortic flow (80, 134, 160). Aside from the validation of closing and opening motions, other parameters such as transvalvular flow velocity, pressure gradients and effective orifice area can be assessed (80, 207, 208). The constructs can be tested over many weeks in such systems to provide information of durability, aging of the constructs and compliance with heart pressure cycles (209). Long test periods always increase the risk of bacterial and fungi contaminations of the samples.

F. Preclinical Testing

Animal models

Functionality assessment of living engineered valves needs to be performed in pre-clinical models prior human to implantation. Although many animal models have been developed to assess function, integrity, thrombogenicity, hemodynamics and pathological behaviors, no standard model is accepted for aortic valve replacement (210). The ISO guideline 5840:2005 mentions that evaluation length should be no less than 90 days, but it does not indicate a preferential animal, possibly as each model possesses inherent advantages/limitations and does not completely replicate the human anatomy, biochemistry and physiology.

Sheep is by far the most employed animal model for pre-clinical valve studies (210, 211). Lambs have cardiac size and output similar to humans under 20 years old (5) and attain full growth within 2 years, which provides fast and valuable information in regards of growth and remodeling (212). Furthermore, they tolerate well cardiothoracic surgery and their elevated calcium metabolism allows to evidence degenerative processes in a short time (213, 214). However, sheep endothelialisation of implanted constructs is more rapid than humans, which renders extrapolation of the results to humans difficult (215).

Porcine models are less employed but present several advantages over sheep. Pigs have a fast growth, possess cardiac sizes and outputs similar to humans and do not readily endothelialize implants, which make them more human-like models. However, the surgical intervention for valve replacement in pigs is technically as stringent and difficult as for humans, and is therefore challenging. Furthermore, the costs associated with animals (especially Yucatan mini-pigs), husbandry and surgery are very high. As a results, pig models are preferred for acute and not long term studies (210).

Other models include dogs, calves and primates. Dogs have a convenient size, are easy to work with and do not have a rapid growth, which would make them interesting for long-term studies. However, animal rights concern are important for dogs (211) and they present a cardiac output much lower than humans. Conversely, calves rapid growth is accompanied by a dramatic increase of cardiac output, well above human values, which makes them more suited for very acute studies or for studying patient-prosthesis mismatch (210). Primates, aside from ethical concerns and very elevated costs (216), present annulus diameters of lower dimension than humans. Therefore, implants of relevant size for humans cannot be directly evaluated. However, the model has be successfully used as proof of concept for transapical tissue engineered pulmonary valve implantation (109).

Evaluation Criteria

Regardless of the animal model employed, the strategy followed consists in comparing post-operative measurements over time to reference data obtained immediately prior implantation and to control valves (mechanical or biological) (217). To obtain information of the implants structure and function during implantation, the animals are either euthanized at different time points and the explants analysed in a destructive fashion. Alternatively, noninvasive techniques can be used instead. The latter option is preferred for ethical and cost reasons but cannot be applied for all necessary characterizations.

An important parameter to consider in pre-clinical studies is site-specificity, since implants placed in positions other than the aortic one may behave differently due to the different biomechanic and hemodynamic environments. As a matter of fact, heart valve replacement pre-clinical studies have indeed been conducted preferentially in the pulmonary position (77, 95, 102, 105, 109, 129, 139, 145, 150, 171, 218). These studies provide valuable information but which cannot be readily extrapolated to the systemic pressure environment of the aortic valve. However, they can be used as guidelines for the necessary validation of aortic valve replacement strategies.

Tissue formation, remodeling and endothelial function

The different techniques employed during in vitro testing can similarly be employed to evaluate tissue formation, structure, organisation and remodeling over the course of implantation. Hematoxilin/eosin or Movat pentachrome staining procedures are useful to determine cellular ingrowth and tissue formation and structure (95, 129, 142, 150, 171, 218). Elastin and collagen deposition are more specifically visualized with van Gieson (129, 139) and Masson’s trichrome stainings (129), respectively, or using immunohistochemical techniques (142, 150). Quantification of the different ECM proteins can be performed with biochemical assays normalized by cell number (DNA quantification) (105, 129, 218)

Tissue remodeling within the implanted constructs can similarly be followed histologically, by comparing volume, ECM composition and cell phenotype of explants retrieved at different times. The presence of myofibroblastic-like cells similar to activated VICs is an important marker of tissue remodeling and should be actively sought. Immunohistochemistry directed against αSMA provides valuable information of the presence and distribution of such activated cells over time (129, 139, 142, 150).

The presence or restoration of a functional endothelial layer at the implant surface can be evaluated by SEM analysis evaluating cell morphology and the presence of adherent thrombocytes (142). Alternatively, immunohistological detection of CD31 (139), vWF (129, 142, 150) and eNOS (129, 142) are commonly employed.

Retention of the implanted cells after implantation is an important parameter to consider for understanding of their role in tissue formation. However, only few studies have sought to discriminate cell origin, using fluorescent labelling prior implantation (139).

The ultimate goal of these characterizations is to evidence the formation of a neotissue structurally and physiologically similar to native aortic valves. The formation of a tri-layered structure rapidly after implantation will undeniably be beneficial for the long term stability and function of the implant (95, 145). This criterion is nevertheless not sufficient. The consistent synthesis of elastin is as well a very important criterion for the proper function and longevity of the replaced valve (204, 205). Finally, the presence of activated myofibroblastic-like cells should not remain for too long periods, to prevent the excessive deposition of unorganized ECM and cause compaction and retraction of the constructs within the first 20 weeks of implantation, resulting in blood regurgitation and impaired function (Fig. 13) (14, 106, 129, 160, 219-221).

figure 13Figure 13. Illustration of the importance of controlling tissue remodeling after implantation in vivo. A: distal and proximal view of a minimally invasive tissue engineered heart valve mounted on a self-expanding stent prior implantation. B: thickened and retracted tissue remodeling 4 weeks after implantation. Adapted from (129).

Biomechanical properties

Tensile tests after explantation provide valuable information of the improvement of biomechanical properties linked to tissue formation and remodeling in the systemic flow. As in vitro characterizations, tensile tests are commonly used (95, 129, 218) and should preferentially be biaxial.

The evolution of anisotropic mechanical properties over implantation time should be particularly evaluated to evidence the structural maturation of the deposited collagen (13) and determine inconsistency with native valves and incoherent biomechanical behaviour (80).

Pathological behaviors

Pre-clinical studies undeniably provide important information regarding the replaced valve functionality. They are as well of high interest to evidence pathological behaviors induces by the replaced valve and resembling natural valve pathologies (8).

Signs of thrombosis induced at the implant surface can be evidenced by SEM analysis (Fig. 14-A) (142) while inflammatory reaction due to the implant material or its degradation products can be assessed by immunostaining of macrophages (92).

Calcification and stenosis of the replaced leaflets can be investigated using various techniques. SEM combined with energy-dispersive X-ray spectroscopy (SEM–EDS) has been showed to confirm the presence of calcification nodules, isolated with focused ion beam, on valves (Fig. 14-B) (38). Raman microscopy can as well be employed to evidence the presence of calcified nodules (222) as well as classical histological stains targeting calcium (alizarin red) (92, 222) or phosphate (von Kossa) crystals (142).

figure 14Figure 14. Identification of pathological behaviors after implantation in vivo. A: electronic microscopy observations of thrombosis at the implant surface is demonstrated by the aggregation of adherent thrombocytes (arrows) at the surface of decellularized ovine pulmonary valve implanted in lamb for 3 months. B: aortic valve calcification can be determined by electronic microscopy. When coupled to focused ion beam and energy-dispersive X-ray spectroscopy, this technique allows to precisely characterize the mineral phase. Adapted from (38, 142).

The determination and measure of circulating biomarkers of aortic valve disorders would be highly beneficial to closely monitor the apparition of pathological behaviors due to the replaced valve. However, from the multiple early and late AVD markers candidates evaluated in clinical studies, only fetuin-A and osteopontin seem interesting as late marker for calcification (5). So far, these markers have never been monitored during pre-clinical studies of aortic valve replacement.

Non-invasive, in situ approaches for functionality, growth and remodeling evaluation

The aforementioned evaluation methods are endpoints that require euthanizing the animals. The development and use of non-invasive methods undeniably provides a better way of monitoring functionality and growth of the replaced aortic valve.

The most useful and straight forward method to non-invasively assess functionality and biomechanical/hemodynamic performance of replaced valves is Doppler echocardiography. This approach provides hemodynamic information over time on peak velocity across the valve, transvalvular systolic pressure gradient, left ventricular outflow tract velocity and insufficiency (223) as well as aortic valve area, effective orifice area, root dimensions, prosthesis size, morphology and function during growth of an animal (142, 224). Doppler is routinely used in post-operative follow up of patients after heart valve replacement to monitor the functional development of prosthetic heart valves and assess the severity of aortic regurgitation (225). It can therefore be used as well in pre-clinical studies to measure the intrinsic gradient after valve replacement and compare with subsequent measurements. From the orifice area obtained from Doppler, the presence and severity of stenosis can be assessed. Furthermore, the calculated orifice area and pressure gradients can be compared to the ones reported from commonly accepted biological prostheses (207). Two methods are currently used to perform echocardiographies: transoesophageal and transthoracic. Transoesophageal echocardiography is the most clinically used and more suited for diagnosing valvular diseases (226). However, transthoracic echocardiography is especially suitable for examination of replaced valves because of the proximity of the oesophagus to the heart and absence of interference with lungs and ribs (224). In summary, Doppler echocardiography is practical, cost effective, readily available and non-invasive. As a result it is a method of choice to study mechanical function and hemodynamic performance of replaced aortic heart valves. Nevertheless, some of the results obtained by echocardiography are underestimated and inter-operator variability is high (227).

Alternatives have been proposed to assess replaced valve functionality and hemodynamic performances such as magnetic resonance imaging (MRI) and multi–detector row CT (Fig. 15) (227). Multi-detector row CT has a superior resolution of aortic valve function and can quantify aortic valve calcification and stenosis (228, 229) while MRI can examine mechanical and hemodynamic properties of heart valves (blood flow velocity, valve opening and closing, valvular dysfunction, and especially regurgitation volumes by phase contrast MR). Other methods have been developed based on velocimetric techniques and computational analysis (laser Doppler anemometry, LDA, and particle image velocimetry) (230). However, these non-invasive techniques are so far limited to testing in vitro.

figure 15Figure 15. Evaluation of valve functionality and hemodynamic performances with non-invasive multi–detector row CT and magnetic resonance imaging. A: multi–detector row CT provides excellent resolution of aortic valve function (top row) and allows calcification quantification (bottom row, arrows indicate moderate to severe calcification from left to right). B: Velocity phase-contrast magnetic resonance imaging allows for hemodynamic properties of aortic valves(measured here at the level of the pulmonary bifurcation, circled in red). Adapted from (227).

In addition to the evolution of valve leaflet over implantation time, the functionality and hemodynamic compliance of the aortic root should as well be monitored to determine the performance of replaced aortic valves. Indeed, flow pattern in the aortic root can affect valve function by allowance of load and stress sharing between leaflet and aortic wall. As a matter of fact, reduced aortic compliance and increased aortic stress lead to thickening of aortic valve leaflet (231). MRI combined with time-resolved particle traces and velocity vector fields can assess aortic root functionality and opening/closing motions of the leaflets simultaneously with morphological data (232, 233).

Valve growth and remodeling during implantation is more difficult to assess through non-invasive techniques although doppler echocardiography could possibly be used to deduce valve growth by monitoring of valve diameter and apparition of regurgitation over time (217). To determine ECM remodeling and organisation in situ, development of multi-photonic endoscope is currently investigated (234, 235). Collagen probes might as well be improved in the future to provide in situ markers of collagen formation (200, 202, 203). Other options relying on the use of activatable molecular imaging agent (near infrared fluorescence, NIRF) have been shown promising to follow proteolytic enzyme activity ex vivo. Matrix mettaloproteinase 2, (MMP2), matrix metalloproteinase 9 (MMP9), cathepsin B and cathepsin K have been evidenced with these techniques in early aortic valve disease (236) and atherosclerosis (237, 238). Future improvements might lead to the direct visualisation of proteolytic enzyme activity in the replaced valves, as was shown for cancer models in mice (239).

Finally, evaluation of endothelial function in situ is not yet foreseeable. However, expression of vascular cell adhesion molecule-1 (VCAM1) can be seen using VCAM1-targeted magnetofluorescent particles (Fig. 16) and MRI (240, 241). VCAM1 is present in inflammatory processes (242) and not in normal endothelium (236). As such, it could be used to detect pathological behavior of the implanted valves.

figure 16Figure 16. Insight of a potential non-invasive technique for studying endothelial cell function. VCAM1-targeted magnetofluorescent particles are uptaken by endothelial cells in a hypercholesterolemic mouse model as visualized by MRI of the aortic arch and root (left). The short axis view of the root confirms the uptake (negative signal enhancement, middle) while color coded signal intensity shows a focused uptake (in red) in the aortic valve commissures (arrows, right). Adapted from (236).

Conclusions

Aortic valve replacement has shown tremendous scientific and clinical progress in the last century. However, prosthetic heart valve replacement for the treatment of aortic valve disease has drawbacks and limitations. Tissue engineering of aortic valves has the potential to overcome these limitations by creating living substitutes with growth ability. If proof of concept has been demonstrated in animal models, the challenge lies in translating tissue engineered aortic valves to clinical practice. Aside from finding the optimal components for the tissue engineering approach, the elucidation of key biological processes in tissue formation and the definition of biomarkers linked to valve functionality must carefully be assessed prior implantation, to ultimately provide clinically valid engineered aortic valves.

The definition of effective translational platforms is an important step in this direction (Fig. 17). Such structure must regroup competences and expertise as multiple and complex as the aortic valve itself. To summarize, this translational effort should be based on a strong technological platform to design and evaluate biomaterials and their interactions with cells. Doing so, at a fundamental level, allows us to uncover and define key biological processes ruling tissue formation and regeneration. The corollary of such fundamental studies leads to the establishment of key biomarkers in animal models that provide the tools, through quality indicators and selection criteria, for monitoring and extrapolating the functionality of the replaced aortic valves. The role of clinical surgeons is critical to assess relevance of the results and provide enhancement feedback. Ultimately, such feedback must be the fertile soil on which functional predictive models can help establish the mandatory clinical safety and relevance prior human studies.

figure 17Figure 17. Overview of translational platform for aortic valve replacement.

References

  1. El-Hamamsy I, Eryigit Z, Stevens L-M, Sarang Z, George R, Clark L, Melina G, Takkenberg J, Yacoub M. Long-term outcomes after autograft versus homograft aortic root replacement in adults with aortic valve disease: a randomised controlled trial. Lancet 2010; 376:524-31.
  2. Yacoub M. The Ross operation–an evolutionary tale. Asian cardiovascular & thoracic annals 2006; 14:1-2.
  3. Higashidate M, Tamiya K, Beppu T, Imai Y. Regulation of the aortic valve opening. In vivo dynamic measurement of aortic valve orifice area. The Journal of thoracic and cardiovascular surgery 1995; 110:496-503.
  4. Chester A, El-Hamamsy I, Butcher J, Latif N, Bertazzo S, Yacoub M. The Living Aortic Valve – From Molecules to Function. Global Cardiology Science & Practice in press.
  5. Butcher J, Mahler G, Hockaday L. Aortic valve disease and treatment: the need for naturally engineered solutions. Advanced drug delivery reviews 2011; 63:242-68.
  6. Vesely I. The role of elastin in aortic valve mechanics. Journal of biomechanics 1998; 31:115-23.
  7. Balguid A, Rubbens M, Mol A, Bank R, Bogers AJ, van Kats J, de Mol B, Baaijens F, Bouten C. The role of collagen cross-links in biomechanical behavior of human aortic heart valve leaflets–relevance for tissue engineering. Tissue engineering 2007; 13:1501-11.
  8. Schoen F. Cardiac valves and valvular pathology: update on function, disease, repair, and replacement. Cardiovascular pathology : the official journal of the Society for Cardiovascular Pathology 2005; 14:189-94.
  9. Cimini M, Rogers K, Boughner D. Smoothelin-positive cells in human and porcine semilunar valves. Histochemistry and cell biology 2003; 120:307-17.
  10. Chester A, Kershaw J, Sarathchandra P, Yacoub M. Localisation and function of nerves in the aortic root. Journal of molecular and cellular cardiology 2008; 44:1045-52.
  11. Marron K, Yacoub M, Polak J, Sheppard M, Fagan D, Whitehead B, de Leval M, Anderson R, Wharton J. Innervation of human atrioventricular and arterial valves. Circulation 1996; 94:368-75.
  12. Taylor P, Batten P, Brand N, Thomas P, Yacoub M. The cardiac valve interstitial cell. The international journal of biochemistry & cell biology 2003; 35:113-8.
  13. Aikawa E, Whittaker P, Farber M, Mendelson K, Padera R, Aikawa M, Schoen F. Human semilunar cardiac valve remodeling by activated cells from fetus to adult: implications for postnatal adaptation, pathology, and tissue engineering. Circulation 2006; 113:1344-52.
  14. Rabkin-Aikawa E, Farber M, Aikawa M, Schoen F. Dynamic and reversible changes of interstitial cell phenotype during remodeling of cardiac valves. The Journal of heart valve disease 2004; 13:841-7.
  15. Merryman W, Youn I, Lukoff H, Krueger P, Guilak F, Hopkins R, Sacks M. Correlation between heart valve interstitial cell stiffness and transvalvular pressure: implications for collagen biosynthesis. American journal of physiology Heart and circulatory physiology 2006; 290:31.
  16. Butcher J, Simmons C, Warnock J. Mechanobiology of the aortic heart valve. The Journal of heart valve disease 2008; 17:62-73.
  17. Stephens E, Durst C, West J, Grande-Allen K. Mitral valvular interstitial cell responses to substrate stiffness depend on age and anatomic region. Acta biomaterialia 2011; 7:75-82.
  18. Schneider P, Deck J. Tissue and cell renewal in the natural aortic valve of rats: an autoradiographic study. Cardiovascular research 1981.
  19. Butcher J, Penrod A, García As, Nerem R. Unique morphology and focal adhesion development of valvular endothelial cells in static and fluid flow environments. Arteriosclerosis, thrombosis, and vascular biology 2004; 24:1429-34.
  20. Risberg B. Pathophysiological mechanisms of thromboembolism. Acta Chir Scand Suppl 1989; 550:104-14.
  21. Simmons C, Grant G, Manduchi E, Davies P. Spatial heterogeneity of endothelial phenotypes correlates with side-specific vulnerability to calcification in normal porcine aortic valves. Circulation research 2005; 96:792-9.
  22. Weston M, LaBorde D, Yoganathan A. Estimation of the shear stress on the surface of an aortic valve leaflet. Annals of biomedical engineering 1999; 27:572-9.
  23. Kilner P, Yang G, Wilkes A, Mohiaddin R, Firmin D, Yacoub M. Asymmetric redirection of flow through the heart. Nature 2000; 404:759-61.
  24. Butcher J, Nerem R. Valvular endothelial cells regulate the phenotype of interstitial cells in co-culture: effects of steady shear stress. Tissue engineering 2006; 12:905-15.
  25. Dagum P, Green G, Nistal F, Daughters G, Timek T, Foppiano L, Bolger A, Ingels N, Miller D. Deformational dynamics of the aortic root: modes and physiologic determinants. Circulation 1999; 100:62.
  26. Bellhouse BJ, Bellhouse FH, Reid KG. Fluid Mechanics of the Aortic Root with Application to Coronary Flow. Nature 1968; 219.
  27. Underwood M, El Khoury G, Deronck D, Glineur D, Dion R. The aortic root: structure, function, and surgical reconstruction. Heart (British Cardiac Society) 2000; 83:376-80.
  28. Billiar K, Sacks M. Biaxial mechanical properties of the natural and glutaraldehyde treated aortic valve cusp–Part I: Experimental results. Journal of biomechanical engineering 2000; 122:23-30.
  29. Schoen F. Aortic valve structure-function correlations: role of elastic fibers no longer a stretch of the imagination. The Journal of heart valve disease 1997; 6:1-6.
  30. Vesely I, Noseworthy R. Micromechanics of the fibrosa and the ventricularis in aortic valve leaflets. Journal of biomechanics 1992; 25:101-13.
  31. Stella J, Sacks M. On the biaxial mechanical properties of the layers of the aortic valve leaflet. Journal of biomechanical engineering 2007; 129:757-66.
  32. Liao J, Yang L, Grashow J, Sacks M. The relation between collagen fibril kinematics and mechanical properties in the mitral valve anterior leaflet. Journal of biomechanical engineering 2007; 129:78-87.
  33. Messika-Zeitoun D, Bielak L, Peyser P, Sheedy P, Turner S, Nkomo V, Breen J, Maalouf J, Scott C, Tajik A, Enriquez-Sarano M. Aortic valve calcification: determinants and progression in the population. Arteriosclerosis, thrombosis, and vascular biology 2007; 27:642-8.
  34. Otto C, Lind B, Kitzman D, Gersh B, Siscovick D. Association of aortic-valve sclerosis with cardiovascular mortality and morbidity in the elderly. The New England journal of medicine 1999; 341:142-7.
  35. Lloyd-Jones D, Adams R, Carnethon M, De Simone G, Ferguson T, Flegal K, Ford E, Furie K, Go A, Greenlund K, Haase N, Hailpern S, Ho M, Howard V, Kissela B, Kittner S, Lackland D, Lisabeth L, Marelli A, McDermott M, Meigs J, Mozaffarian D, Nichol G, O’Donnell C, Roger V, Rosamond W, Sacco R, Sorlie P, Stafford R, Steinberger J, Thom T, Wasserthiel-Smoller S, Wong N, Wylie-Rosett J, Hong Y, American Heart Association Statistics C, Stroke Statistics S. Heart disease and stroke statistics–2009 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2009; 119:181.
  36. Sani M, Karaye K, Borodo M. Prevalence and pattern of rheumatic heart disease in the Nigerian savannah: an echocardiographic study. Cardiovascular journal of Africa 2007; 18:295-9.
  37. Mohler E. Mechanisms of aortic valve calcification. The American journal of cardiology 2004; 94:1396.
  38. Bertazzo S, Gentleman E, Cloyd K, Chester A, Yacoub M, Stevens M. Nano-analytical electron microscopy reveals fundamental insights into human cardiovascular tissue calcification. Nature materials 2013; 12:576-83.
  39. Osman L, Yacoub M, Latif N, Amrani M, Chester A. Role of human valve interstitial cells in valve calcification and their response to atorvastatin. Circulation 2006; 114:52.
  40. Carabello B. Introduction to aortic stenosis. Circulation research 2013; 113:179-85.
  41. Claiborne T, Slepian M, Hossainy S, Bluestein D. Polymeric trileaflet prosthetic heart valves: evolution and path to clinical reality. Expert review of medical devices 2012; 9:577-94.
  42. Nakamura T, Hultgren H, Shettigar U, Fowles R. Noninvasive evaluation of the severity of aortic stenosis in adult patients. American heart journal 1984; 107:959-66.
  43. Braun H, Comeau W. The importance of high-pitched squeaking systolic murmur in the diagnosis of aortic stenosis and calcification of the aortic valve. The New England journal of medicine 1951; 244:507-9.
  44. Bonow R, Carabello B, Chatterjee K, de Leon A, Faxon D, Freed M, Gaasch W, Lytle B, Nishimura R, O’Gara P, O’Rourke R, Otto C, Shah P, Shanewise J, Writing Committee M, American College of Cardiology/American Heart Association Task F. 2008 Focused update incorporated into the ACC/AHA 2006 guidelines 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 (Writing Committee to Revise the 1998 Guidelines for the Management of Patients With Valvular Heart Disease): endorsed by the Society of Cardiovascular Anesthesiologists, Society for Cardiovascular Angiography and Interventions, and Society of Thoracic Surgeons. Circulation 2008; 118:661.
  45. Carabello B. How does the heart respond to aortic stenosis: let me count the ways. Circulation Cardiovascular imaging 2013; 6:858-60.
  46. Cheng A, Dagum P, Miller DC. Aortic root dynamics and surgery: from craft to science. Philosophical Transactions of the Royal Society B: Biological Sciences 2007; 362.
  47. Hufnagel C. Plastic cardiac valvular prostheses. American Journal of Medicine 1951.
  48. Matthews A. The development of the Starr-Edwards heart valve. Texas Heart Institute journal / from the Texas Heart Institute of St Luke’s Episcopal Hospital, Texas Children’s Hospital 1998; 25:282-93.
  49. Wieting D. The Bjork-Shiley Delrin tilting disc heart valve: historical perspective, design and need for scientific analyses after 25 years. The Journal of heart valve disease 1996; 5 Suppl 2:68.
  50. Emery R, Mettler E, Nicoloff D. A new cardiac prosthesis: the St. Jude Medical cardiac valve: in vivo results. Circulation 1979; 60:48-54.
  51. Gott V, Alejo D, Cameron D. Mechanical heart valves: 50 years of evolution. The Annals of thoracic surgery 2003; 76:9.
  52. Zilla P, Brink J, Human P, Bezuidenhout D. Prosthetic heart valves: catering for the few. Biomaterials 2008; 29:385-406.
  53. Hammermeister K, Sethi G, Henderson W, Grover F, Oprian C, Rahimtoola S. Outcomes 15 years after valve replacement with a mechanical versus a bioprosthetic valve: final report of the Veterans Affairs randomized trial. Journal of the American College of Cardiology 2000; 36:1152-8.
  54. Grunkemeier G, Li H, Naftel D, Starr A, Rahimtoola S. Long-term performance of heart valve prostheses. Current problems in cardiology 2000; 25:73-154.
  55. Ross D. Homograft replacement of the aortic valve. Lancet 1962; 2:487.
  56. Neumann A, Cebotari S, Tudorache I, Haverich A, Sarikouch S. Heart valve engineering: decellularized allograft matrices in clinical practice. Biomedizinische Technik Biomedical engineering 2013; 58:453-6.
  57. Kaiser G, Hancock W, Lukban S, Litwak R. Clinical use of a new design stented xenograft heart valve prosthesis. Surgical forum 1969; 20:137-8.
  58. Carpentier A. From valvular xenograft to valvular bioprosthesis (1965-1977). Medical instrumentation 1977; 11:98-101.
  59. Carpentier A. From valvular xenograft to valvular bioprosthesis: 1965-1970. The Annals of thoracic surgery 1989; 48:4.
  60. Oxenham H, Bloomfield P, Wheatley D, Lee R, Cunningham J, Prescott R, Miller H. Twenty year comparison of a Bjork-Shiley mechanical heart valve with porcine bioprostheses. Heart (British Cardiac Society) 2003; 89:715-21.
  61. Rahimtoola S. Choice of prosthetic heart valve in adults an update. Journal of the American College of Cardiology 2010; 55:2413-26.
  62. Butany J, Ahluwalia M, Munroe C, Fayet C, Ahn C, Blit P, Kepron C, Cusimano R, Leask R. Mechanical heart valve prostheses: identification and evaluation. Cardiovascular pathology : the official journal of the Society for Cardiovascular Pathology 2003; 12:1-22.
  63. Lupinetti F, Warner J, Jones T, Herndon S. Comparison of human tissues and mechanical prostheses for aortic valve replacement in children. Circulation 1997; 96:321-5.
  64. Ross D. Replacement of aortic and mitral valves with a pulmonary autograft. Lancet 1967; 2:956-8.
  65. Yacoub M, Klieverik L, Melina G, Edwards S, Sarathchandra P, Bogers AJ, Squarcia U, Sani G, van Herwerden L, Takkenberg J. An evaluation of the Ross operation in adults. The Journal of heart valve disease 2006; 15:531-9.
  66. Stewart R, Backer C, Hillman N, Lundt C, Mavroudis C. The Ross operation in children: effects of aortic annuloplasty. The Annals of thoracic surgery 2007; 84:1326-30.
  67. Takkenberg J, van Herwerden L, Galema T, Bekkers J, Kleyburg-Linkers V, Eijkemans M, Bogers AJ. Serial echocardiographic assessment of neo-aortic regurgitation and root dimensions after the modified Ross procedure. The Journal of heart valve disease 2006; 15:100.
  68. Hörer J, Hanke T, Stierle U, Takkenberg J, Bogers AJ, Hemmer W, Rein J, Hetzer R, Hübler M, Robinson D, Sievers H, Lange Rd. Neoaortic root diameters and aortic regurgitation in children after the Ross operation. The Annals of thoracic surgery 2009; 88:594.
  69. Cameron D, Vricella L. What is the proper place of the Ross procedure in our modern armamentarium? Current cardiology reports 2007; 9:93-8.
  70. Heineken FG, Skalak R. Tissue Engineering: A Brief Overview. Journal of biomechanical engineering 1991; 113:111-2.
  71. Yacoub M, Takkenberg J. Will heart valve tissue engineering change the world? Nature clinical practice Cardiovascular medicine 2005; 2:60-1.
  72. Mendelson K, Schoen F. Heart valve tissue engineering: concepts, approaches, progress, and challenges. Annals of biomedical engineering 2006; 34:1799-819.
  73. Mol A, Smits A, Bouten C, Baaijens F. Tissue engineering of heart valves: advances and current challenges. Expert review of medical devices 2009; 6:259-75.
  74. Vesely I. Heart valve tissue engineering. Circulation research 2005; 97:743-55.
  75. Hopkins R. Tissue engineering of heart valves: decellularized valve scaffolds. Circulation 2005; 111:2712-4.
  76. Rabkin E, Schoen F. Cardiovascular tissue engineering. Cardiovascular pathology : the official journal of the Society for Cardiovascular Pathology 2002; 11:305-17.
  77. Shinoka T, Breuer C, Tanel R, Zund G, Miura T, Ma P, Langer R, Vacanti J, Mayer J. Tissue engineering heart valves: valve leaflet replacement study in a lamb model. The Annals of thoracic surgery 1995; 60:6.
  78. Shinoka T, Ma PX, Shum-Tim D, Breuer CK, Cusick RA, Zund G, Langer R, Vacanti JP, Mayer JE, Jr. Tissue-engineered heart valves. Autologous valve leaflet replacement study in a lamb model. Circulation 1996; 94:II164-8.
  79. Shinoka T. Tissue engineered heart valves: autologous cell seeding on biodegradable polymer scaffold. Artificial organs 2002; 26:402-6.
  80. Mol A, Rutten M, Driessen N, Bouten C, Zünd G, Baaijens F, Hoerstrup S. Autologous human tissue-engineered heart valves: prospects for systemic application. Circulation 2006; 114:8.
  81. Sacks M, Schoen F, Mayer J. Bioengineering challenges for heart valve tissue engineering. Annual review of biomedical engineering 2009; 11:289-313.
  82. Rieder E, Kasimir M-T, Silberhumer G, Seebacher G, Wolner E, Simon P, Weigel G. Decellularization protocols of porcine heart valves differ importantly in efficiency of cell removal and susceptibility of the matrix to recellularization with human vascular cells. Journal of Thoracic and Cardiovascular Surgery 2004.
  83. Schmidt Dr, Stock U, Hoerstrup S. Tissue engineering of heart valves using decellularized xenogeneic or polymeric starter matrices. Philosophical Transactions of the Royal Society B: Biological Sciences 2007.
  84. Naso F, Gandaglia A, Formato M, Cigliano A, Lepedda A, Gerosa G, Spina M. Differential distribution of structural components and hydration in aortic and pulmonary heart valve conduits: Impact of detergent-based cell removal. Acta biomaterialia 2010.
  85. Liao J, Joyce E, Sacks M. Effects of decellularization on the mechanical and structural properties of the porcine aortic valve leaflet. Biomaterials 2008.
  86. Bottio T, Tarzia V, Lin CD, Buratto E, Rizzoli G, Spina M, Gandaglia A, Naso F, Gerosa G. The changing hydrodynamic performance of the decellularized intact porcine aortic root: considerations on in-vitro testing. Journal of Heart Valve Disease 2010; 19:485.
  87. Simon P, Kasimir M, Seebacher G, Weigel G, Ullrich R, Salzer-Muhar U, Rieder E, Wolner E. Early failure of the tissue engineered porcine heart valve SYNERGRAFT in pediatric patients. European journal of cardio-thoracic surgery : official journal of the European Association for Cardio-thoracic Surgery 2003; 23:1002.
  88. Friedhelm S, Inge B, Ute S, Thilo W, Hans HS. Histopathologic Findings in a Novel Decellularized Pulmonary Homograft: An Autopsy Study. The Annals of thoracic surgery 2005; 79.
  89. Konuma T, Devaney E, Bove E, Gelehrter S, Hirsch J, Tavakkol Z, Ohye R. Performance of CryoValve SG decellularized pulmonary allografts compared with standard cryopreserved allografts. The Annals of thoracic surgery 2009; 88:849.
  90. Zehr K, Yagubyan M, Connolly H, Nelson S, Schaff H. Aortic root replacement with a novel decellularized cryopreserved aortic homograft: postoperative immunoreactivity and early results. The Journal of thoracic and cardiovascular surgery 2005; 130:1010-5.
  91. Tedder M, Liao J, Weed B, Stabler C, Zhang H, Simionescu A, Simionescu D. Stabilized collagen scaffolds for heart valve tissue engineering. Tissue engineering Part A 2009; 15:1257-68.
  92. Tedder M, Simionescu A, Chen J, Liao J, Simionescu D. Assembly and testing of stem cell-seeded layered collagen constructs for heart valve tissue engineering. Tissue engineering Part A 2011; 17:25-36.
  93. Ye X, Zhao Q, Sun X, Li H. Enhancement of mesenchymal stem cell attachment to decellularized porcine aortic valve scaffold by in vitro coating with antibody against CD90: a preliminary study on antibody-modified tissue-engineered heart valve. Tissue engineering Part A 2009; 15:1-11.
  94. Sodian R, Hoerstrup S, Sperling J, Martin D, Daebritz S, Mayer J, Vacanti J. Evaluation of biodegradable, three-dimensional matrices for tissue engineering of heart valves. ASAIO journal (American Society for Artificial Internal Organs : 1992) 2000; 46:107-10.
  95. Hoerstrup SP, Sodian R, Daebritz S, Wang J, Bacha EA, Martin DP, Moran AM, Guleserian KJ, Sperling JS, Kaushal S, Vacanti JP, Schoen FJ, Mayer JE. Functional Living Trileaflet Heart Valves Grown In Vitro. Circulation 2000; 102.
  96. Sodian R, Loebe M, Hein A, Martin D, Hoerstrup S, Potapov E, Hausmann H, Lueth T, Hetzer R. Application of stereolithography for scaffold fabrication for tissue engineered heart valves. ASAIO journal (American Society for Artificial Internal Organs : 1992) 2002; 48:12-6.
  97. van Lieshout M, Vaz C, Rutten M, Peters G, Baaijens F. Electrospinning versus knitting: two scaffolds for tissue engineering of the aortic valve. Journal of biomaterials science Polymer edition 2006; 17:77-89.
  98. Sohier Jrm, Corre P, Perret C, Pilet P, Weiss P. Novel and Simple Alternative to Create Nanofibrillar Matrices of Interest for Tissue Engineering. Tissue engineering Part C, Methods 2013.
  99. Sohier Jrm, Carubelli I, Sarathchandra P, Latif N, Chester A, Yacoub M. The potential of anisotropic matrices as substrate for heart valve engineering. Biomaterials 2014; 35:1833-44.
  100. Gunatillake P, Adhikari R. Biodegradable synthetic polymers for tissue engineering. European cells & materials 2003; 5:1.
  101. Sodian R, Sperling J, Martin D, Egozy A, Stock U, Mayer J, Vacanti J. Fabrication of a trileaflet heart valve scaffold from a polyhydroxyalkanoate biopolyester for use in tissue engineering. Tissue engineering 2000; 6:183-8.
  102. Shinoka T, Ma P, Shum-Tim D, Breuer C, Cusick R, Zund G, Langer R, Vacanti J, Mayer J. Tissue-engineered heart valves. Autologous valve leaflet replacement study in a lamb model. Circulation 1996; 94:8.
  103. Sharan R, Danielle G, George CE, Elena A, David ES, Diana MG-L, Virna LS, John EM, Michael SS. The role of organ level conditioning on the promotion of engineered heart valve tissue development in-vitro using mesenchymal stem cells. Biomaterials 2010; 31.
  104. Zund G, Breuer C, Shinoka T, Ma P, Langer R, Mayer J, Vacanti J. The in vitro construction of a tissue engineered bioprosthetic heart valve. European journal of cardio-thoracic surgery : official journal of the European Association for Cardio-thoracic Surgery 1997; 11:493-7.
  105. Stock U, Nagashima M, Khalil P, Nollert G, Herden T, Sperling J, Moran A, Lien J, Martin D, Schoen F, Vacanti J, Mayer J. Tissue-engineered valved conduits in the pulmonary circulation. The Journal of thoracic and cardiovascular surgery 2000; 119:732-40.
  106. Rabkin E, Hoerstrup S, Aikawa M, Mayer J, Schoen F. Evolution of cell phenotype and extracellular matrix in tissue-engineered heart valves during in-vitro maturation and in-vivo remodeling. The Journal of heart valve disease 2002; 11:308.
  107. Brugmans M, Driessen-Mol A, Rubbens M, Cox M, Baaijens F. Poly-ε-caprolactone scaffold and reduced in vitro cell culture: beneficial effect on compaction and improved valvular tissue formation. Journal of tissue engineering and regenerative medicine 2013.
  108. Balguid A, Mol A, van Marion M, Bank R, Bouten C, Baaijens F. Tailoring fiber diameter in electrospun poly(epsilon-caprolactone) scaffolds for optimal cellular infiltration in cardiovascular tissue engineering. Tissue engineering Part A 2009; 15:437-44.
  109. Weber B, Scherman J, Emmert M, Gruenenfelder J, Verbeek R, Bracher M, Black M, Kortsmit J, Franz T, Schoenauer R, Baumgartner L, Brokopp C, Agarkova I, Wolint P, Zund G, Falk V, Zilla P, Hoerstrup S. Injectable living marrow stromal cell-based autologous tissue engineered heart valves: first experiences with a one-step intervention in primates. Eur Heart J 2011; 32:2830-40.
  110. Butcher J, Nerem R. Porcine aortic valve interstitial cells in three-dimensional culture: comparison of phenotype with aortic smooth muscle cells. The Journal of heart valve disease 2004; 13:478.
  111. Stefan J, Thomas CF. Cardiovascular Tissue Engineering Based on Fibrin-Gel-Scaffolds. 2011.
  112. Ye Q, Zünd G, Benedikt P, Jockenhoevel S, Hoerstrup S, Sakyama S, Hubbell J, Turina M. Fibrin gel as a three dimensional matrix in cardiovascular tissue engineering. European Journal of Cardio-thoracic Surgery 2000.
  113. Jockenhoevel S, Zund G, Hoerstrup S, Chalabi K, Sachweh J, Demircan L, Messmer B, Turina M. Fibrin gel – advantages of a new scaffold in cardiovascular tissue engineering. European Journal of Cardio-thoracic Surgery 2001.
  114. Jockenhoevel S, Chalabi K, Sachweh J, Groesdonk H, Demircan L, Grossmann M, Zund G, Messmer B. Tissue Engineering: Complete Autologous Valve Conduit – A New Moulding Technique. Thoracic and Cardiovascular Surgeon 2001.
  115. Flanagan T, Cornelissen C, Koch S, Tschoeke B, Sachweh J, Schmitz-Rode T, Jockenhoevel S. The in vitro development of autologous fibrin-based tissue-engineered heart valves through optimised dynamic conditioning. Biomaterials 2007.
  116. Flanagan T, Sachweh Jr, Frese J, Schnöring H, Gronloh N, Koch S, Tolba R, Schmitz-Rode T, Jockenhoevel S. In Vivo Remodeling and Structural Characterization of Fibrin-Based Tissue-Engineered Heart Valves in the Adult Sheep Model. Tissue Engineering Part A 2009.
  117. Mol A, Lieshout M, Veen C, Neuenschwander S, Hoerstrup S, Baaijens F, Bouten C. Fibrin as a cell carrier in cardiovascular tissue engineering applications. Biomaterials 2005.
  118. Albanna M, Bou-Akl T, Walters H, Matthew H. Improving the mechanical properties of chitosan-based heart valve scaffolds using chitosan fibers. Journal of the mechanical behavior of biomedical materials 2012; 5:171-80.
  119. Syedain Z, Bradee A, Kren S, Taylor D, Tranquillo R. Decellularized tissue-engineered heart valve leaflets with recellularization potential. Tissue engineering Part A 2013; 19:759-69.
  120. Aoki J, Serruys PW, van Beusekom H, Ong ATL, McFadden EP, Sianos G, van der Giessen WJ, Regar E, de Feyter PJ, Davis HR, Rowland S, Kutryk MJB. Endothelial Progenitor Cell Capture by Stents Coated With Antibody Against CD34: The HEALING-FIM (Healthy Endothelial Accelerated Lining Inhibits Neointimal Growth-First In Man) Registry. Journal of the American College of Cardiology 2005; 45:1574-9.
  121. Lee JM, Choe W, Kim B-K, Seo W-W, Lim W-H, Kang C-K, Kyeong S, Eom KD, Cho H-J, Kim Y-C, Hur J, Yang H-M, Cho H-J, Lee Y-S, Kim H-S. Comparison of endothelialization and neointimal formation with stents coated with antibodies against CD34 and vascular endothelial-cadherin. Biomaterials 2012; 33:8917-27.
  122. Avci-Adali M, Paul A, Ziemer G, Wendel H. New strategies for in vivo tissue engineering by mimicry of homing factors for self-endothelialisation of blood contacting materials. Biomaterials 2008.
  123. Ota T, Sawa Y, Iwai S, Kitajima T, Ueda Y, Coppin C, Matsuda H, Okita Y. Fibronectin-Hepatocyte Growth Factor Enhances Reendothelialization in Tissue-Engineered Heart Valve. Annals of Thoracic Surgery 2005.
  124. Kim T, Park T. Biomimicking Extracellular Matrix: Cell Adhesive RGD Peptide Modified Electrospun Poly(D,L-lactic-co-glycolic acid) Nanofiber Mesh. Tissue engineering 2006.
  125. Meyers SR, Khoo X, Huang X, Walsh EB, Grinstaff MW, Kenan DJ. The development of peptide-based interfacial biomaterials for generating biological functionality on the surface of bioinert materials. Biomaterials 2009; 30:277-86.
  126. Sales VL, Engelmayr GC, Johnson JA, Gao J, Wang Y, Sacks MS, Mayer JE. Protein Precoating of Elastomeric Tissue-Engineering Scaffolds Increased Cellularity, Enhanced Extracellular Matrix Protein Production, and Differentially Regulated the Phenotypes of Circulating Endothelial Progenitor Cells. Circulation 2007; 116:I-55-I-63.
  127. Shinoka T, Shum-Tim D, Ma P, Tanel R, Langer R, Vacanti J, Mayer J. Tissue-engineered heart valve leaflets: does cell origin affect outcome? Circulation 1997; 96.
  128. Brugmans M, Driessen-Mol A, Rubbens M, Cox M, Baaijens F. Polycaprolactone scaffold and reduced in vitro cell culture: beneficial effect on compaction and improved valvular tissue formation. J Tissue Eng Regen Med 2013.
  129. Schmidt Dr, Dijkman P, Driessen-Mol A, Stenger R, Mariani C, Puolakka A, Rissanen M, Deichmann T, Odermatt B, Weber B, Emmert M, Zund G, Baaijens F, Hoerstrup S. Minimally-invasive implantation of living tissue engineered heart valves: a comprehensive approach from autologous vascular cells to stem cells. Journal of the American College of Cardiology 2010; 56:510-20.
  130. Engelmayr G, Sales V, Mayer J, Sacks M. Cyclic flexure and laminar flow synergistically accelerate mesenchymal stem cell-mediated engineered tissue formation: Implications for engineered heart valve tissues. Biomaterials 2006; 27:6083-95.
  131. Batten P, Sarathchandra P, Antoniw J, Tay S, Lowdell M, Taylor P, Yacoub M. Human mesenchymal stem cells induce T cell anergy and downregulate T cell allo-responses via the TH2 pathway: relevance to tissue engineering human heart valves. Tissue engineering 2006; 12:2263-73.
  132. Colazzo F, Sarathchandra P, Smolenski R, Chester A, Tseng Y-T, Czernuszka J, Yacoub M, Taylor P. Extracellular matrix production by adipose-derived stem cells: implications for heart valve tissue engineering. Biomaterials 2011; 32:119-27.
  133. Hoerstrup S, Kadner A, Melnitchouk S, Trojan A, Eid K, Tracy J, Sodian R, Visjager J, Kolb S, Grunenfelder J, Zund G, Turina M. Tissue engineering of functional trileaflet heart valves from human marrow stromal cells. Circulation 2002; 106:50.
  134. Schmidt Dr, Mol A, Odermatt B, Neuenschwander S, Breymann C, Gössi M, Genoni M, Zund G, Hoerstrup S. Engineering of biologically active living heart valve leaflets using human umbilical cord-derived progenitor cells. Tissue engineering 2006; 12:3223-32.
  135. Cebotari S, Lichtenberg A, Tudorache I, Hilfiker A, Mertsching H, Leyh R, Breymann T, Kallenbach K, Maniuc L, Batrinac A, Repin O, Maliga O, Ciubotaru A, Haverich A. Clinical application of tissue engineered human heart valves using autologous progenitor cells. Circulation 2006; 114:7.
  136. Schmidt Dr, Mol A, Breymann C, Achermann J, Odermatt B, Gössi M, Neuenschwander S, Prêtre R, Genoni M, Zund G, Hoerstrup S. Living autologous heart valves engineered from human prenatally harvested progenitors. Circulation 2006; 114:31.
  137. Weber B, Zeisberger S, Hoerstrup S. Prenatally harvested cells for cardiovascular tissue engineering: fabrication of autologous implants prior to birth. Placenta 2011; 32 Suppl 4:9.
  138. Schmidt Dr, Achermann J, Odermatt B, Genoni M, Zund G, Hoerstrup S. Cryopreserved amniotic fluid-derived cells: a lifelong autologous fetal stem cell source for heart valve tissue engineering. The Journal of heart valve disease 2008; 17:446.
  139. Kim S-S, Lim S-H, Hong Y-S, Cho S-W, Ryu J, Chang B-C, Choi C, Kim B-S. Tissue engineering of heart valves in vivo using bone marrow-derived cells. Artificial organs 2006; 30:554-7.
  140. Colazzo F, Chester A, Taylor P, Yacoub M. Induction of mesenchymal to endothelial transformation of adipose-derived stem cells. The Journal of heart valve disease 2010; 19:736-44.
  141. Lichtenberg A, Cebotari S, Tudorache I, Sturz G, Winterhalter M, Hilfiker A, Haverich A. Flow-dependent re-endothelialization of tissue-engineered heart valves. The Journal of heart valve disease 2006; 15:287.
  142. Lichtenberg A, Tudorache I, Cebotari S, Suprunov M, Tudorache G, Goerler H, Park J-K, Hilfiker-Kleiner D, Ringes-Lichtenberg S, Karck M, Brandes G, Hilfiker A, Haverich A. Preclinical testing of tissue-engineered heart valves re-endothelialized under simulated physiological conditions. Circulation 2006; 114:65.
  143. Dohmen P, Lembcke A, Holinski S, Pruss A, Konertz W. Ten years of clinical results with a tissue-engineered pulmonary valve. The Annals of thoracic surgery 2011; 92:1308-14.
  144. Dohmen P, Lembcke A, Hotz H, Kivelitz D, Konertz W. Ross operation with a tissue-engineered heart valve. The Annals of thoracic surgery 2002; 74:1438-42.
  145. Sutherland F, Perry T, Yu Y, Sherwood M, Rabkin E, Masuda Y, Garcia G, McLellan D, Engelmayr G, Sacks M, Schoen F, Mayer J. From stem cells to viable autologous semilunar heart valve. Circulation 2005; 111:2783-91.
  146. Ramaswamy S, Gottlieb D, Engelmayr G, Aikawa E, Schmidt D, Gaitan-Leon D, Sales V, Mayer J, Sacks M. The role of organ level conditioning on the promotion of engineered heart valve tissue development in-vitro using mesenchymal stem cells. Biomaterials 2010; 31:1114-25.
  147. Perry Tr, Kaushal S, Sutherland F, Guleserian K, Bischoff J, Sacks M, Mayer J. Thoracic Surgery Directors Association Award. Bone marrow as a cell source for tissue engineering heart valves. The Annals of thoracic surgery 2003; 75:761.
  148. Sales V, Mettler B, Engelmayr G, Aikawa E, Bischoff J, Martin D, Exarhopoulos A, Moses M, Schoen F, Sacks M, Mayer J. Endothelial progenitor cells as a sole source for ex vivo seeding of tissue-engineered heart valves. Tissue engineering Part A 2010; 16:257-67.
  149. Rippel R, Ghanbari H, Seifalian A. Tissue-engineered heart valve: future of cardiac surgery. World journal of surgery 2012; 36:1581-91.
  150. Steinhoff G, Stock U, Karim N, Mertsching H, Timke A, Meliss R, Pethig K, Haverich A, Bader A. Tissue engineering of pulmonary heart valves on allogenic acellular matrix conduits: in vivo restoration of valve tissue. Circulation 2000; 102:5.
  151. Dweck M, Boon N, Newby D. Calcific aortic stenosis: a disease of the valve and the myocardium. Journal of the American College of Cardiology 2012; 60:1854-63.
  152. Thayer P, Balachandran K, Rathan S, Yap C, Arjunon S, Jo H, Yoganathan A. The effects of combined cyclic stretch and pressure on the aortic valve interstitial cell phenotype. Annals of biomedical engineering 2011; 39:1654-67.
  153. Walker G, Masters K, Shah D, Anseth K, Leinwand L. Valvular myofibroblast activation by transforming growth factor-beta: implications for pathological extracellular matrix remodeling in heart valve disease. Circulation research 2004; 95:253-60.
  154. Xing Y, Warnock J, He Z, Hilbert S, Yoganathan A. Cyclic pressure affects the biological properties of porcine aortic valve leaflets in a magnitude and frequency dependent manner. Annals of biomedical engineering 2004; 32:1461-70.
  155. Balachandran K, Konduri S, Sucosky P, Jo H, Yoganathan A. An ex vivo study of the biological properties of porcine aortic valves in response to circumferential cyclic stretch. Annals of biomedical engineering 2006; 34:1655-65.
  156. Sodian R, Lemke T, Fritsche C, Hoerstrup S, Fu P, Potapov E, Hausmann H, Hetzer R. Tissue-engineering bioreactors: a new combined cell-seeding and perfusion system for vascular tissue engineering. Tissue engineering 2002; 8:863-70.
  157. Narita Y, Hata K-I, Kagami H, Usui A, Ueda M, Ueda Y. Novel pulse duplicating bioreactor system for tissue-engineered vascular construct. Tissue engineering 2004; 10:1224-33.
  158. Hildebrand D, Wu Z, Mayer J, Sacks M. Design and hydrodynamic evaluation of a novel pulsatile bioreactor for biologically active heart valves. Annals of biomedical engineering 2004; 32:1039-49.
  159. Bowles C, New S, Loon R, Dreger S, Biglino G, Chan C, Parker K, Chester A, Yacoub M, Taylor P. Hydrodynamic Evaluation of a Bioreactor for Tissue Engineering Heart Valves. Cardiovascular Engineering and Technology 2010.
  160. Mol A, Driessen N, Rutten M, Hoerstrup S, Bouten C, Baaijens F. Tissue engineering of human heart valve leaflets: a novel bioreactor for a strain-based conditioning approach. Annals of biomedical engineering 2005; 33:1778-88.
  161. Schmidt Dr, Mol A, Kelm J, Hoerstrup S. In vitro heart valve tissue engineering. Methods in molecular medicine 2007; 140:319-30.
  162. Sucosky P, Padala M, Elhammali A, Balachandran K, Jo H, Yoganathan A. Design of an ex vivo culture system to investigate the effects of shear stress on cardiovascular tissue. Journal of biomechanical engineering 2008; 130:35001.
  163. Mol A, Bouten C, Zünd G, Günter C, Visjager J, Turina M, Baaijens F, Hoerstrup S. The relevance of large strains in functional tissue engineering of heart valves. The Thoracic and cardiovascular surgeon 2003; 51:78-83.
  164. Isenberg B, Tranquillo R. Long-term cyclic distention enhances the mechanical properties of collagen-based media-equivalents. Annals of biomedical engineering 2003; 31:937-49.
  165. Ku C-H, Johnson P, Batten P, Sarathchandra P, Chambers R, Taylor P, Yacoub M, Chester A. Collagen synthesis by mesenchymal stem cells and aortic valve interstitial cells in response to mechanical stretch. Cardiovascular research 2006; 71:548-56.
  166. Rubbens M, Mol A, van Marion M, Hanemaaijer R, Bank R, Baaijens F, Bouten C. Straining mode-dependent collagen remodeling in engineered cardiovascular tissue. Tissue engineering Part A 2009; 15:841-9.
  167. Boerboom R, Rubbens M, Driessen N, Bouten C, Baaijens F. Effect of strain magnitude on the tissue properties of engineered cardiovascular constructs. Annals of biomedical engineering 2008; 36:244-53.
  168. Rubbens M, Mol A, Boerboom R, Bank R, Baaijens F, Bouten C. Intermittent straining accelerates the development of tissue properties in engineered heart valve tissue. Tissue engineering Part A 2009; 15:999-1008.
  169. Karim N, Golz K, Bader A. The cardiovascular tissue-reactor: a novel device for the engineering of heart valves. Artificial organs 2006; 30:809-14.
  170. Dohmen P. Clinical results of implanted tissue engineered heart valves. HSR proceedings in intensive care & cardiovascular anesthesia 2012; 4:225-31.
  171. Gottlieb D, Kunal T, Emani S, Aikawa E, Brown D, Powell A, Nedder A, Engelmayr G, Melero-Martin J, Sacks M, Mayer J. In vivo monitoring of function of autologous engineered pulmonary valve. J Thorac Cardiovasc Surg 2010; 139:723-31.
  172. Puvimanasinghe J, Steyerberg E, Takkenberg J, Eijkemans M, van Herwerden L, Bogers A, Habbema J. Prognosis after aortic valve replacement with a bioprosthesis: predictions based on meta-analysis and microsimulation. Circulation 2001; 103:1535-41.
  173. Masoumi N, Larson B, Annabi N, Kharaziha M, Zamanian B, Shapero K, Cubberley A, Camci-Unal G, Manning K, Mayer J, Khademhosseini A. Electrospun PGS:PCL Microfibers Align Human Valvular Interstitial Cells and Provide Tunable Scaffold Anisotropy. Advanced healthcare materials 2014.
  174. Pham Q, Sharma U, Mikos A. Electrospun poly(epsilon-caprolactone) microfiber and multilayer nanofiber/microfiber scaffolds: characterization of scaffolds and measurement of cellular infiltration. Biomacromolecules 2006; 7:2796-805.
  175. Zhong S, Zhang Y, Lim C. Fabrication of large pores in electrospun nanofibrous scaffolds for cellular infiltration: a review. Tissue engineering Part B, Reviews 2012; 18:77-87.
  176. Hinderer S, Seifert J, Votteler M, Shen N, Rheinlaender J, Schäffer T, Schenke-Layland K. Engineering of a bio-functionalized hybrid off-the-shelf heart valve. Biomaterials 2014; 35:2130-9.
  177. Zhou M, Qiao W, Liu Z, Shang T, Qiao T, Mao C, Liu C. Development and in vivo evaluation of small-diameter vascular grafts engineered by outgrowth endothelial cells and electrospun chitosan/poly(ε-caprolactone) nanofibrous scaffolds. Tissue engineering Part A 2014; 20:79-91.
  178. Sant S, Iyer D, Gaharwar A, Patel A, Khademhosseini A. Effect of biodegradation and de novo matrix synthesis on the mechanical properties of valvular interstitial cell-seeded polyglycerol sebacate-polycaprolactone scaffolds. Acta biomaterialia 2013; 9:5963-73.
  179. O’Cearbhaill E, Murphy M, Barry F, McHugh P, Barron V. Behavior of Human Mesenchymal Stem Cells in Fibrin-Based Vascular Tissue Engineering Constructs. Annals of biomedical engineering 2010.
  180. Wang Q, McGoron A, Pinchuk L, Schoephoerster R. A novel small animal model for biocompatibility assessment of polymeric materials for use in prosthetic heart valves. Journal of biomedical materials research Part A 2010; 93:442-53.M
  181. Burugapalli K, Pandit A. Characterization of tissue response and in vivo degradation of cholecyst-derived extracellular matrix. Biomacromolecules 2007; 8:3439-51.
  182. Borghi A, New S, Chester A, Taylor P, Yacoub M. Time-dependent mechanical properties of aortic valve cusps: effect of glycosaminoglycan depletion. Acta biomaterialia 2013; 9:4645-52.
  183. Mauri A, Zeisberger S, Hoerstrup S, Mazza E. Analysis of the uniaxial and multiaxial mechanical response of a tissue-engineered vascular graft. Tissue engineering Part A 2013; 19:583-92.
  184. Robinson P, Tranquillo R. Planar biaxial behavior of fibrin-based tissue-engineered heart valve leaflets. Tissue engineering Part A 2009; 15:2763-72.
  185. Tower T, Neidert M, Tranquillo R. Fiber alignment imaging during mechanical testing of soft tissues. Annals of biomedical engineering 2002; 30:1221-33.
  186. Loerakker S, Argento G, Oomens C, Baaijens F. Effects of valve geometry and tissue anisotropy on the radial stretch and coaptation area of tissue-engineered heart valves. Journal of biomechanics 2013; 46:1792-800.
  187. Engelmayr G, Rabkin E, Sutherland F, Schoen F, Mayer J, Sacks M. The independent role of cyclic flexure in the early in vitro development of an engineered heart valve tissue. Biomaterials 2005; 26:175-87.
  188. Engelmayr G, Sacks M. Prediction of extracellular matrix stiffness in engineered heart valve tissues based on nonwoven scaffolds. Biomechanics and modeling in mechanobiology 2008; 7:309-21.
  189. Neidlinger-Wilke C, Grood E, Claes L, Brand R. Fibroblast orientation to stretch begins within three hours. Journal of orthopaedic research : official publication of the Orthopaedic Research Society 2002; 20:953-6.
  190. Dweck M, Boon N, Newby D. Calcific aortic stenosis: a disease of the valve and the myocardium. J Am Coll Cardiol 2012; 60:1854-63.
  191. Pedron S, Kasko A, Peinado C, Anseth K. Effect of heparin oligomer chain length on the activation of valvular interstitial cells. Biomacromolecules 2010; 11:1692-5.
  192. Evans N, Oreffo R, Healy E, Thurner P, Man Y. Epithelial mechanobiology, skin wound healing, and the stem cell niche. Journal of the mechanical behavior of biomedical materials 2013.
  193. Kloxin A, Benton J, Anseth K. In situ elasticity modulation with dynamic substrates to direct cell phenotype. Biomaterials 2010; 31:1-8.
  194. Olsson M, Rosenqvist Mr, Nilsson J. Expression of HLA-DR antigen and smooth muscle cell differentiation markers by valvular fibroblasts in degenerative aortic stenosis. Journal of the American College of Cardiology 1994.
  195. Quinlan A, Billiar K. Investigating the role of substrate stiffness in the persistence of valvular interstitial cell activation. J Biomed Mater Res A 2012; 100:2474-82.
  196. Rabkin E, Aikawa M, Stone J, Fukumoto Y, Libby P, Schoen F. Activated interstitial myofibroblasts express catabolic enzymes and mediate matrix remodeling in myxomatous heart valves. Circulation 2001; 104:2525-32.
  197. Dreger S, Thomas P, Sachlos E, Chester A, Czernuszka J, Taylor P, Yacoub M. Potential for synthesis and degradation of extracellular matrix proteins by valve interstitial cells seeded onto collagen scaffolds. Tissue engineering 2006; 12:2533-40.
  198. Dreger S, Taylor P, Allen S, Yacoub M. Profile and localization of matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) in human heart valves. The Journal of heart valve disease 2002; 11:875.
  199. Konig K, Schenke-Layland K, Riemann I, Stock U. Multiphoton autofluorescence imaging of intratissue elastic fibers. Biomaterials 2005; 26:495-500.
  200. Schenke-Layland K, Riemann I, Damour O, Stock U, König K. Two-photon microscopes and in vivo multiphoton tomographs–powerful diagnostic tools for tissue engineering and drug delivery. Advanced drug delivery reviews 2006; 58:878-96.
  201. Zipfel W, Williams R, Webb W. Nonlinear magic: multiphoton microscopy in the biosciences. Nature biotechnology 2003; 21:1369-77.
  202. Boerboom R, Krahn K, Megens R, van Zandvoort M, Merkx M, Bouten C. High resolution imaging of collagen organisation and synthesis using a versatile collagen specific probe. Journal of structural biology 2007; 159:392-9.
  203. Krahn K, Bouten C, van Tuijl S, van Zandvoort M, Merkx M. Fluorescently labeled collagen binding proteins allow specific visualization of collagen in tissues and live cell culture. Analytical biochemistry 2006; 350:177-85.
  204. Schoof P, Takkenberg J, van Suylen R-J, Zondervan P, Hazekamp M, Dion R, Bogers AJ. Degeneration of the pulmonary autograft: an explant study. The Journal of thoracic and cardiovascular surgery 2006; 132:1426-32.
  205. Lee T, Midura R, Hascall V, Vesely I. The effect of elastin damage on the mechanics of the aortic valve. Journal of biomechanics 2001; 34:203-10.
  206. Driessen N, Mol A, Bouten C, Baaijens F. Modeling the mechanics of tissue-engineered human heart valve leaflets. Journal of biomechanics 2007; 40:325-34.
  207. Marquez S, Hon R, Yoganathan A. Comparative hydrodynamic evaluation of bioprosthetic heart valves. The Journal of heart valve disease 2001; 10:802-11.
  208. Warnock J, Konduri S, He Z, Yoganathan A. Design of a sterile organ culture system for the ex vivo study of aortic heart valves. Journal of biomechanical engineering 2005; 127:857-61.
  209. Sumikura H, Nakayama Y, Ohnuma K, Takewa Y, Tatsumi E. In Vitro Evaluation of a Novel Autologous Aortic Valve (Biovalve) With a Pulsatile Circulation Circuit. Artificial organs 2013.
  210. Gallegos R, Nockel P, Rivard A, Bianco R. The current state of in-vivo pre-clinical animal models for heart valve evaluation. The Journal of heart valve disease 2005; 14:423-32.
  211. Rashid S, Salacinski H, Hamilton G, Seifalian A. The use of animal models in developing the discipline of cardiovascular tissue engineering: a review. Biomaterials 2004.
  212. Ali M, Kumar S, Bjornstad K, Duran C. The sheep as an animal model for heart valve research. Cardiovascular surgery (London, England) 1996; 4:543-9.
  213. Chanda J, Kuribayashi R, Abe T. Valved conduit in the descending thoracic aorta in juvenile sheep: a useful, cost-effective model for accelerated calcification study in systemic circulation. Biomaterials 1997; 18:1317-21.
  214. Meuris B, Ozaki S, Herijgers P, Verbeken E, Flameng W. Bioprosthetic tissue calcification: influence of blood contact and arterial pressure. an experimental study in rats and sheep. Journal of Heart Valve Disease 2003; 12:392-9.
  215. Ouyang D, Salerno C, Pederson T, Bolman R, Bianco R. Long-term evaluation of orthotopically implanted stentless bioprosthetic aortic valves in juvenile sheep. Journal of investigative surgery : the official journal of the Academy of Surgical Research 1998; 11:175-83.
  216. Wolfensohn S, Lloyd M. Handbook of laboratory animal management and welfare. John Wiley & Sons 2013.
  217. Hjortnaes J, Bouten C, Van Herwerden L, Gründeman P, Kluin J. Translating autologous heart valve tissue engineering from bench to bed. Tissue engineering Part B, Reviews 2009; 15:307-17.
  218. Sodian R, Hoerstrup S, Sperling J, Daebritz S, Martin D, Moran A, Kim B, Schoen F, Vacanti J, Mayer J. Early in vivo experience with tissue-engineered trileaflet heart valves. Circulation 2000; 102:9.
  219. Weber B, Scherman J, Emmert M, Gruenenfelder J, Verbeek R, Bracher M, Black M, Kortsmit J, Franz T, Schoenauer R, Baumgartner L, Brokopp C, Agarkova I, Wolint P, Zund G, Falk V, Zilla P, Hoerstrup S. Injectable living marrow stromal cell-based autologous tissue engineered heart valves: first experiences with a one-step intervention in primates. European heart journal 2011; 32:2830-40.
  220. Brugmans M, Driessen-Mol A, Rubbens M, Cox M, Baaijens F. Polycaprolactone scaffold and reduced in vitro cell culture: beneficial effect on compaction and improved valvular tissue formation. Journal of tissue engineering and regenerative medicine 2013.
  221. Gottlieb D, Kunal T, Emani S, Aikawa E, Brown D, Powell A, Nedder A, Engelmayr G, Melero-Martin J, Sacks M, Mayer J. In vivo monitoring of function of autologous engineered pulmonary valve. The Journal of thoracic and cardiovascular surgery 2010; 139:723-31.
  222. Cloyd K, El-Hamamsy I, Boonrungsiman S, Hedegaard M, Gentleman E, Sarathchandra P, Colazzo F, Gentleman M, Yacoub M, Chester A, Stevens M. Characterization of porcine aortic valvular interstitial cell ‘calcified’ nodules. PloS one 2012; 7.
  223. American College of C, American Heart Association Task Force on Practice G, Society of Cardiovascular A, Bonow R, Carabello B, Chatterjee K, de Leon A, Faxon D, Freed M, Gaasch W, Lytle B, Nishimura R, O’Gara P, O’Rourke R, Otto C, Shah P, Shanewise J, Smith S, Jacobs A, Adams C, Anderson J, Antman E, Fuster V, Halperin J, Hiratzka L, Hunt S, Page R, Riegel B. ACC/AHA 2006 guidelines 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 (writing Committee to Revise the 1998 guidelines for the management of patients with valvular heart disease) developed in collaboration with the Society of Cardiovascular Anesthesiologists endorsed by the Society for Cardiovascular Angiography and Interventions and the Society of Thoracic Surgeons. Journal of the American College of Cardiology 2006; 48:148.
  224. Van den Brink R. Evaluation of prosthetic heart valves by transesophageal echocardiography: problems, pitfalls, and timing of echocardiography. Seminars in cardiothoracic and vascular anesthesia 2006; 10:89-100.
  225. Willems T, Takkenberg J, Steyerberg E, Kleyburg-Linkers V, Roelandt J, Bos E, van Herwerden L. Human tissue valves in aortic position: determinants of reoperation and valve regurgitation. Circulation 2001; 103:1515-21.
  226. Aslam A, Vasavada B, Khan I. Prosthetic heart valves: Types and echocardiographic evaluation. International Journal of Cardiology 2007.
  227. Vogel-Claussen J, Pannu H, Spevak P, Fishman E, Bluemke D. Cardiac valve assessment with MR imaging and 64-section multi-detector row CT. Radiographics : a review publication of the Radiological Society of North America, Inc 2006; 26:1769-84.
  228. Boxt L, Lipton M, Kwong R, Rybicki F, Clouse M. Computed tomography for assessment of cardiac chambers, valves, myocardium and pericardium. Cardiology clinics 2003; 21:561-85.
  229. Braun J, Oldendorf M, Moshage W, Heidler R, Zeitler E, Luft F, C. Electron beam computed tomography in the evaluation of cardiac calcifications in chronic dialysis patients. American Journal of Kidney Diseases 1996; 27.
  230. Grigioni M, Daniele C, D’Avenio G, Morbiducci U, Del Gaudio C, Abbate M, Di Meo D. Innovative technologies for the assessment of cardiovascular medical devices: state-of-the-art techniques for artificial heart valve testing. Expert review of medical devices 2004; 1:81-93.
  231. Rabkin S, Jue J, Tsang M. Aortic valve sclerosis is associated with an echocardiographically determined thinner aortic wall. The Journal of heart valve disease 2006; 15:158-64.
  232. Markl M, Draney M, Hope M, Levin J, Chan F, Alley M, Pelc N, Herfkens R. Time-resolved 3-dimensional velocity mapping in the thoracic aorta: visualization of 3-directional blood flow patterns in healthy volunteers and patients. Journal of computer assisted tomography 2004; 28:459-68.
  233. Markl M, Draney M, Miller D, Levin J, Williamson E, Pelc N, Liang D, Herfkens R. Time-resolved three-dimensional magnetic resonance velocity mapping of aortic flow in healthy volunteers and patients after valve-sparing aortic root replacement. The Journal of thoracic and cardiovascular surgery 2005; 130:456-63.
  234. Konig K, Ehlers A, Riemann I, Schenkl S, Bückle R, Kaatz M. Clinical two-photon microendoscopy. Microscopy research and technique 2007; 70:398-402.
  235. Jung J, Schnitzer M. Multiphoton endoscopy. Optics letters 2003; 28:902-4.
  236. Aikawa E, Nahrendorf M, Sosnovik D, Lok VM, Jaffer FA, Aikawa M, Weissleder R. Multimodality Molecular Imaging Identifies Proteolytic and Osteogenic Activities in Early Aortic Valve Disease. Circulation 2007; 115.
  237. Deguchi J-o, Aikawa M, Tung C-H, Aikawa E, Kim D-E, Ntziachristos V, Weissleder R, Libby P. Inflammation in atherosclerosis: visualizing matrix metalloproteinase action in macrophages in vivo. Circulation 2006; 114:55-62.
  238. Jaffer F, Kim D-E, Quinti L, Tung C-H, Aikawa E, Pande A, Kohler R, Shi G-P, Libby P, Weissleder R. Optical visualization of cathepsin K activity in atherosclerosis with a novel, protease-activatable fluorescence sensor. Circulation 2007; 115:2292-8.
  239. Bogdanov A, Mazzanti M. Fluorescent macromolecular sensors of enzymatic activity for in vivo imaging. Progress in molecular biology and translational science 2013; 113:349-87.
  240. Kelly KA, Allport JR, Tsourkas A, Shinde-Patil VR, Josephson L, Weissleder R. Detection of vascular adhesion molecule-1 expression using a novel multimodal nanoparticle. Circulation research 2005; 96:327-36.
  241. Nahrendorf M, Jaffer F, Kelly K, Sosnovik D, Aikawa E, Libby P, Weissleder R. Noninvasive vascular cell adhesion molecule-1 imaging identifies inflammatory activation of cells in atherosclerosis. Circulation 2006; 114:1504-11.
  242. Cybulsky M, Gimbrone M. Endothelial expression of a mononuclear leukocyte adhesion molecule during atherogenesis. Science (New York, NY) 1991; 251:788-91.

 

Hide picture