Small Diameter Cell-Free Tissue-Engineered Vascular Grafts: Comparison
Please note this is a comparison between Version 2 by Jason Zhu and Version 1 by Maria Alejandra Rodriguez Soto.

Vascular grafts (VGs) are medical devices intended to replace the function of a blood vessel. Available VGs in the market present low patency rates for small diameter applications setting the VG failure. This event arises from the inadequate response of the cells interacting with the biomaterial in the context of operative conditions generating chronic inflammation and a lack of regenerative signals where stenosis or aneurysms can occur. Tissue Engineered Vascular grafts (TEVGs) aim to induce the regeneration of the native vessel to overcome these limitations. Besides the biochemical stimuli, the biomaterial and the particular micro and macrostructure of the graft will determine the specific behavior under pulsatile pressure. The TEVG must support blood flow withstanding the exerted pressure, allowing the proper compliance required for the biomechanical stimulation needed for regeneration. Although the international standards outline the specific requirements to evaluate vascular grafts, the challenge remains in choosing the proper biomaterial and manufacturing TEVGs with good quality features to perform satisfactorily.

  • tissue engineered vascular grafts
  • biomaterials
  • biodegradable
  • mechanical properties
  • Polymers
  • biomechanical stimulation
  • Cells
  • Failure
  • Success
  • Small diameter

1. Introduction

Cardiovascular diseases (CVDs) are a group of conditions affecting the heart and blood vessels. According to the World Health Organization, CVDs are currently the leading cause of death worldwide, representing 32% of all global deaths [1]. Accordingly, almost 18 million people died from a CVD in 2019. Among CVDs, vascular diseases compromising the blood vessel structure take hold of more than 8.5 million people worldwide. For instance, coronary artery disease (CAD) is the most common type of CVD, affecting 6.7% worldwide. CAD alters the mechanical properties of the coronary artery supplying the heart muscle, resulting in a decrease in the blood flow [2]. Moreover, other conditions also affect the mechanical properties of the blood vessels, such as the Peripheral Artery Disease (PAD), which compromises the blood flow in the limbs, and aortic aneurysms, identified by a balloon-like bulge in the aorta [3].
Currently, surgical approaches for vascular reconstruction focus on using autologous or synthetic vascular grafts (VGs) to recover the blood flow. Autologous grafts such as the great saphenous vein (GSV) and Internal Mamary Artery (IMA) can maintain long-term patency due to their regenerative properties (86% up to five years). However, GSV has limited availability and represents additional risks due to possible post-surgical complications with morbidity rates up to 50% [4]. Furthermore, synthetic VGs in the market are made from Polytetrafluoroethylene (PTFE—known as Teflon®) and Polyethylene terephthalate (PET—known as Dacron®). Although their performance is suitable in straight blood vessels and large diameters, they are inefficient in replacing small diameter vessels (>6 mm diameter) and under non-unidirectional or non-fully developed flows, as seen in their decreased patency rates below 32% after two years [5]. These low patency rates have been correlated to a foreign body response promoting thrombogenesis and stenosis, compromising the blood flow. With 3 out of 100,000 adults requiring peripheral vascular segment repairs through small diameter VGs [6], new approaches to overcome the current challenges include the use of bio-based VGs, such as allogeneic cryopreserved blood vessels (Cryograft®—CryoLife) [7], xenografts from bovine blood vessels (Artegraft®—LeMaitre) [8], bovine pericardium [9], or xenografts from sheep extracellular matrix (ECM) (Omniflow® II—LeMaitre) [10]. Nevertheless, they still do not show a clear advantage compared to commercial synthetic non-biodegradable VGs [11].
These bio-based VGs are part of Tissue-engineered vascular grafts (TEVGs); these scaffolds are intended to guide the tissue regeneration of the vascular wall and are considered as a strategy to get closer to the biological response of native blood vessels. Under physiological conditions, native arteries have a complex, multilayered structure with different ECM compositions and microstructures to contribute to the required compliance precisely responding to pulsatile pressure. Components in each layer will determine the total vessel elastic, viscous, and inertial properties that provide the critical biomechanical signals required to regulate cell adhesion, growth, and differentiation [12].

2. The Potential of TEVGs on Clinical Success

2.1. Failure Causes of TEVG’s Related to the Mechanical Properties

The leading cause of failure of VG and TEVGs is patency loss where blood flow becomes compromised and can occur through different mechanisms. The most common mechanism is related to thrombogenesis when the surface of the biomaterial lacks the required hemocompatibility [15][13]. Furthermore, this is the most common cause of failure on VGs and TEVGs used as arteriovenous fistulas, given that clots are formed after repeated puncture, and the thrombus might spread if there is not an anticoagulant surface or treatment. In these cases, secondary patency in a VG is achieved by removing the clot with a catheter, and the lumen of the graft can be restored, allowing blood perfusion. However, this procedure is not always successful if the main thrombogenic conditions are still present, in which case the condition may evolve towards a fibrotic thrombus that will make it impossible to perform a new procedure to recover patency [14]. However, the most common cause of failure related to the mechanical properties of the VG or TEVG includes intimal hyperplasia, in which the overproliferation of smooth muscle cells thickens the tunica intima in the blood vessel, causing the contraction of the construct and the loss of patency [16][15]. On the other hand, the formation of fibrotic tissue surrounding the graft–in this case, the microstructure of the VG or TEVG–does not allow cell infiltration, and a dense layer of collagen is formed surrounding the graft. In this case, stem cells differentiate toward myofibroblasts, causing the contraction of the VG or TEVG, and stenosis is developed [43][16]. However, another critical failure cause is the formation of aneurysms in which the chemical or biological degradation of the graft and/or structural defects may lead to the dilatation or rupture of the graft [44][17].

Unstable Flow Conditions and Intimal Hyperplasia Development in TEVGs

Intimal hyperplasia has been reported to present in 10–30% of failure causes of VGs. This high failure rate has been correlated with the compliance mismatch between the VG and the native vessel. In this sense, it has also been reported that the patency loss is directly proportional to the compliance mismatch [45][18]. The low compliance not only depends on the biomaterial origin but also depends on the microstructure. For instance, it has been reported that there is a strong correlation between low porosity and increasing TEVG wall thickness with a low compliance [46][19]. The flow stability is key to maintaining the graft patency. The flow regime, velocity profile, and cyclical deformation caused by the pulsatile flow create determinant wall shear stresses (WSS), which are mechanical signals in the cells interacting and repopulating the TEVG [46][19]. From this perspective, low compliances have also been strongly correlated with low WSS due to the low-compliant grafts presenting a significant difference in diameter under the pulsatile pressure [45][18]. Furthermore, while the VG maintains a constant diameter, the artery will dilate and contract. When the artery dilates, the VG maintains its low diameter, and the blood flow profile produces a sizeable corresponding effect. This profile is characterized by zones of blood recirculation negatively affecting the velocity profile, inducing turbulent flows, and altering the pressure wave, decreasing the WSS [45][18]. Regarding the compliance mismatch effect on WSS, normal WSS reported on small diameter vessels—such as the coronary artery—is 0.68 N/m2 ranging between 0.3 to 1.24 N/m2 [47][20]. It has been reported that non-compliant VGs present low shear stress near 0.03 N/m2, medium compliance VGs display similar shear stresses with an average of 1.04 N/m2, and high compliance grafts present higher WSS with an average of 3.6 N/m2 [45][18]. Based on this correlation, it could be expected that low-compliant TEVGs will induce intimal hyperplasia. Accordingly, TEVGs for different applications must consider the compliance values and the physiological shear stress in which the TEVG will be anastomosed. For instance, GSV has an average compliance value of 4.4%/100 mmHg, and it has been reported that implanted GSV grafts for coronary artery bypass present a range of 1.22 N/m2 to 1.73 N/m2, whereas GSVs grafts below 0.71 N/m2 are predictors for the VG failure [48][21]. Recent reports have also shown that the left internal mammary artery (IMA), with compliance of 5.22%/100 mmHg, used as a VG for a coronary bypass, maintained its patency for one year and presented high WSS values near 4.43 N/m2, contrasted with those occluded with a WSS 2.56 N/m2 [49][22]. Although the differences between the compliance of the GSV and IMA are not significantly different, the WSS produced as VGs for CABG are very different due to differences between the layer’s composition in the media and adventitia layers. Although GSV has a different structure from arteries, the vein adapts to the arterial environment due to the different hemodynamic conditions and increased oxygen tension [50][23]. The most significant change is the increase in the vein diameter between 20% and 85% for arteriovenous fistulas or lower extremity grafts, respectively. The vein wall also augments the wall thickness due to the increased pressure from the proliferation of smooth muscle cells and adventitial fibroblasts derived from bone marrow progenitor cells. These changes have been associated with the normalization of shear stress, occurring during the first months where the initial shear stress will reach up to 9.6 N/m2, leading to a patency rate between 75–90% at one year and 50% at 15 years [51][24].

2.2. Biomechanical Stimulation for Physiological Regenerative Responses; Physiological Wall Shear Stresses

When developing TEVGs, it is essential to induce the regeneration of the vascular wall within the anastomosis; not only will the bioactive chemical properties of the graft induce this process, but biomechanical signals are required. Aside from the microstructure allowing the cell infiltration and proliferation, the wall shear stress will generate different responses in the cells interacting with the biomaterial in the hemodynamic context. Therefore, physiological WSS will lead to the regeneration of the vascular wall, whereas the low WSS induced by the lack of compliance will have a detrimental effect on the gene expression of immune cells, smooth muscle cells, and endothelial cells. For instance, for regeneration to occur, the inflammatory response must be modulated towards the differentiation of Macrophages to the M2 type, recognized by the cytokine secretion to induce extracellular matrix deposition. Low WSS has been correlated to the maintenance of the M1 phenotype leading to chronic inflammatory responses. On the other hand, smooth muscle cells must begin their differentiation towards a contractile phenotype rather than a synthetic phenotype. The synthetic phenotype related to low WSS has been associated with lower blood pressure responses affecting the construct’s overall compliance. Finally, endothelial cells should differentiate towards a functional phenotype able to release nitric oxide (NO) as a regulator of the vascular wall tone. Low WSS has been shown to induce senescence on endothelial cells without proliferation capacity and limited release of NO. On the other hand, low WSS on endothelial cells also increases the expression of MCP-1 (Monocyte Chemoattractant Protein-1), which is a chemoattractant for proinflammatory monocytes. At the same time, the expression of VCAM-1 (Vascular cell adhesion protein-1), ICAM-1 (Intercellular Adhesion Molecule-1), and EDN-1 (Endothelin-1) increases, which are adhesive molecules for monocytes, and PDGF (platelet-derived growth factor) also increases, promoting thrombus formation. Regarding smooth muscle cells, a low WSS decreases the expression of a-SMA (smooth muscle actin), SM22 (Transgelin), SMTN (Smothelin), and CNN (calponin); all are genes related to the contractile function of smooth muscle cells required to respond to contractile and dilating signals from endothelial cells [16][15]. Furthermore, low WSS induces the up-regulation of proliferative genes, responsible for intimal hyperplasia, TGF-β1 (transforming growth factor-beta) inducing inflammation, and the MMP2 (matrix metalloproteinase-2) that not only degrades the vascular wall but also increase the migration and proliferation of the smooth muscle cells with the synthetic phenotype [51,55][24][25]. Finally, macrophage modulation due to low WSS has been shown to reduce the expression of M2 phenotype-related genes such as CD206 (macrophage mannose receptor 1), IL-10 (Interleukin 10), which blocks the NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) proinflammatory pathway, and TGF-B1. Furthermore, genes related to increased inflammation are reported to be activated, such as the signaling pathway related to NF-κB activation, MCP-1, and Selectin as chemotactic for new inflammatory cells, as well as present an increase in MMP9, related to the degradation of the scaffold [56,57][26][27].

3. Biomechanical Design Requirement for Vascular Grafts

Due to the significant differences between vascular grafts and native arteries in their mechanical properties and behavior under implantation conditions, several requirements have been studied to develop mechanical characterizations of these new grafts. The requirements to evaluate vascular grafts are outlined in International Standards ANSI/AAMI VP20: 1994 (American National Standard for cardiovascular implants and vascular prostheses) [58][28], ISO 7198:2016 (Cardiovascular implants and extracorporeal systems—Vascular prostheses—Tubular vascular grafts and vascular patches 94, and ASTM F3225-17 (Standard Guide for Characterization and Assessment of Vascular Graft Tissue Engineered Medical Products (TEMPs) [59][29].

ANSI/AAMI VP20: 1994, ISO 7198:2016 and ASTM F3225-17

Test guidelines for international standards are defined for all vascular grafts: synthetic, biological, and coated [59][29]. According to ASTM F3225-17, mechanical testing must be performed in an environment that emulates the vascular grafting conditions of use, most commonly in buffered saline solutions at 37 °C. Otherwise, non-physiological test conditions must be justified. In addition, parameters such as the time between tissue collection and testing and sample storage may modify the mechanical properties [59][29]. A minimum of three samples from at least three manufactured lots is necessary to have an appropriate variability of the characteristics in the samples studied. Due to the anisotropic properties associated with vascular grafts, strength tests should be performed, including more than one axis. Therefore, longitudinal, and circumferential tensile strength tests are performed to determine whether the axial and radial yield and/or breakpoint are reached, respectively. Stress vs. strain plots must be made to include and analyze the data. In addition, suture retention strength tests should be developed to determine the force required to pull a suture from the vascular graft to simulate clinical techniques (straight-across, oblique, and longitudinal procedures). The suture is usually placed 2 mm from the edge and tested in various directions to determine the strength that the prosthesis withstands before mechanical failure. Moreover, a burst strength test is performed to determine the pressure rate change until sample bursting occurs. This method allows to report the diameter of the sample when it is pressurized directly with fluid or gas. A repeated puncture test with a dialysis needle (16G) should be performed to measure the strength that supports the prosthesis through a force test such as pressurized burst strength or circumferential tensile strength [59][29]. It is necessary to perform tests to determine the thickness of the wall and the relaxed and pressurized internal diameter to observe changes in diameter under different hemodynamic conditions. Furthermore, the vascular graft discontinuity should be reported when the lumen diameter decreases during kinking and the radius of curvature that impedes normal flow through the graft. Another way to evaluate conditions that approach the preclinical environment is by measuring diameter change simulated under cardiac cycle conditions to determine the radial dynamic compliance. In addition, the rate of water leakage through the prosthesis wall should be characterized to avoid leakage at the time of implantation [59][29].

4. Trends on TEVGs Design, Biomaterials, and Manufacture Techniques in the Context of Desired Mechanical Properties

4.1. TEVGs That Have Reached Clinical Trials

The most recent TEVG that has entered to clinical trial phase is a Polyhedral oligomeric silsesquioxane poly (carbonate-urea) urethane (POSS-PCU) small-diameter vascular graft, fabricated through extrusion and phase inversion method using sodium bicarbonate as a porogen. This TEVG is currently being tested as arteriovenous fistulas for hemodialysis; the study began in 2021 and is expected to reach completion in April of 2025. This TEVG has a small diameter (<5 mm) with a mean wall thickness of 0.94 mm that is placed in adult female patients undergoing hemodialysis and in need of new arteriovenous access but without any viable access for dialysis. This study has an inclusion criteria of patients on oral contraceptives or with an intrauterine device, aiming to demonstrate patency in patients with increased risk of thrombosis. It is expected to measure patency rates at 18 months by Doppler ultrasonography (d-US) and compare it to the patency of the PTFE grafts. They also will measure the occurrence of any Serious Adverse Event (SAE) related to the implantation of the POSS-PCU TEVG. For the initial characterization of the POSS-PCU vascular graft, they used Delfino’s strain energy potential to capture the viscoelastic properties of the materials. Later, they conducted different experimental and computational experiments to show shear stress within the graft wall in the context of increased uniaxial strength. They concluded that besides the overall resistance of the graft, the stiffness and long-term viscoelastic properties could be improved with better manufacturing techniques. The POSS-PCU TEVG offers similar longitudinal tensile strength compared to GSV and IMA grafts (3210 vs. 2405 and 4300 KPa), however, its suture resistance strength is markedly higher (4460 vs. 3200 and 1350 KPa, respectively). Furthermore, it presents reduced dynamic compliance compared to the allografts (1.59 vs. 4.4 and 5.22%/100 mmHg respectively) may explain its increased stiffness and poor vascular physiology resemblance. A Human Acellular Vessel (HAV) from Humacyte is a graft also developed for patients with need of hemodialysis access but who are not candidates for a fistula. This TEVG is created in vitro through the culture of human smooth muscle cells and fibroblast cultured over a biodegradable polymer. Further decellularization process creates a structure that retains the extracellular matrix protein and provides corresponding mechanical properties. Animal models were performed in baboons as arteriovenous grafts with 80% patency at six months. This vascular graft has been included in different clinical trials. One of the most relevant, made in 2016 as a phase 2 trial in 40 elected patients, demonstrated primary patency rates of 63% (95% CI 47–72) at six months and 28% (95% CI 17–40) at 12 months. Conversely, secondary assisted patency rates at six and 12 months were 97% (95% CI 85–98) and 89% (95% CI 74–93), respectively. Secondary patency rates in extensive multicenter cohort studies with PTFE were 55–66% at one year, making HAV a suitable option. In the group of patients, they also measured serious adverse events (SAEs) that occurred 155 times in 33 patients, leading to increased reintervention rates. SAEs mainly included patency loss, infection, and one case of steal syndrome. HAV provides an exciting conclusion, as they have better secondary patency rates than PTFE-based VGs, leading to a well-tolerated TEVG with no signs of aneurysm formation or degradation with low immunogenicity. For which it has been included in different clinical trials from 2015 to 2020. Research-ers found that Artegraft® has primary, primary-assisted, and secondary patency rates of immunogenicity. Likewise, the TRUE vascular graft is also a decellularized arteriovenous TEVG designed by neonatal human dermal fibroblasts seeded in a bovine fibrin gel. A baboon pre-clinical model was performed on 10 subjects to determine its safety in terms of occurrence of adverse events and to find patency rates. They found patency rates at 3 and 6 months of 83% (5 of 6) and 60% (3 of 5), respectively, with evidence of recellularization of smooth muscle cells and endothelium formation. However, circumferential tensile strength and suture resistance strength were like the IMA (3800 and 1950 vs. 4100 and 1350 KPa, respectively), making it a newly suitable option for further study. Another kind of TEVG for hemodialysis accesses is Artegraft®, a decellularized bovine carotid artery used for arteriovenous fistula generation. These TEVG have been included in different clinical trials from 2015 to 2020. Researchers found that Artegraft® has primary, primary-assisted, and secondary patency rates of 73.3%, 67%, and 89%, respectively. SAE was present in one immunocompromised patient that presented a resistant infection, leading to the early removal of Artegaft® at two months. A relevant finding is that anastomotic venous stenosis occurred in some grafts and was the most common indication of graft removal. Artegraft® demonstrated similar tensile strength properties compared to PTFE VGs. However, its low dynamic compliance [1,5][1][5] can explain the occurrence of stenotic venous anastomosis in most grafts. BioIntegral Surgical No-React ® are bovine pericardial xenografts used as a strategy for vascular graft infections due to their regenerative properties; these series of clinical trials began in 2019. A prospective study of six patients with infected aortoiliac segments treated with BioIntegral Surgical No-React ® demonstrated that four out of the six patients are still alive with complete patency demonstrated by d-US. Unfortunately, two died from acute myocardial infarction, and the other due to sepsis secondary to the vascular infection. Although this graft has a remarkably increased longitudinal tensile strength compared to GSVs and IMA (10,000 vs. 2405 and 4300 KPa), it conferred to most patients (4 out of 6) a proper control of the infection with reasonable long-term patency rates.

4.2. TEVGs That Have Reached Pre-Clinical Animal Models

After the analysis of the recruited data, a total of 43 studies fulfilled inclusion criteria for TEVGs that have reached pre-clinical animal models [66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107][30][31][32][33][34][35][36][37][38][39][40][41][42][43][44][45][46][47][48][49][50][51][52][53][54][55][56][57][58][59][60][61][62][63][64][65][66][67][68][69][70][71]. It was found that 46.43% (n = 26; 46.46%) of the studies reported the use of Poly ε-caprolactone (PCL), mostly manufactured through electrospinning (n = 23, 88.46%) and more frequently tested on murine animal models (rats). PCL has been shown to be biocompatible, and the constructs exhibit slow degradation rates of about 1–2 years [67,68,70,72,74,76,78,79,80,81,82,89,90,92,95,96,97,99,102,103,104,105,107][31][32][34][36][38][40][42][43][44][45][46][53][54][56][59][60][61][63][66][67][68][69][71]. This has been correlated with the ability to maintain the TEVGs stability under hemodynamic operative conditions and less acidic breakdown products compared to other polyesters because it can be easily degraded by lipases and macrophages, presenting a low inflammatory profile and the potential for loadbearing [29][72]. However, although it has been proven that PCL is a relatively easy material to work with, PCL-Based TEVGs usually present low compliance compared to native vessels (GSV). From this perspective, the raw PCL has a compliance value close to 2%/mmHg, and PCL-Based TEVGs present a value close to 3%/mmHg; both values are lower than those of the GSV or IMA, closer to 5%/mmHg. To overcome these limitations, various reseauthorchers have chosen to use a variation of PCL such as poly (L-lactide-co-ε-caprolactone) or PLCL with a softer and more elastic nature. Compared to PCL, PLCL-based TEVGs present higher compliance values close to 8%/mmHg [108][73]. It has also been reported that PLCL has excellent mechanical properties such as high plasticity and higher degradation rates than PCL. Nevertheless, the lactide group inclusion generates low biocompatibility, poor hydrophilicity, and presents acidic degradation. Polyurethane-Based TEVGs (PUs) are the second most reported TEVGs (n = 6; 10.71%). PUs has been considered as a good base biomaterial due to its relatively high tensile and flexural strength [37,77,81,91,93,100,104][41][45][55][57][64][68][74]. Furthermore, it has been shown that PU has relatively high compliance compared to other materials. PU-Based TEVGs present an average compliance value of 6.5%/mmHg [37][74], being closer to the value found in native arteries. However, although none of the consulted manuscripts had a specific value for the circumferential tensile strength of the material, it has been reported that repeated puncture of PU-Based TEVGs might lead to aneurism generation, meaning that the material’s circumferential strength might be compromised, and it could thus not be comparable to the native arteries. Therefore, for its use in TEVGs, different alternatives should be included to improve the circumferential strength of the material while maintaining the required elasticity for compliance. The third most common biomaterial for TEVGs fabrication corresponds to decellularized arteries (n = 4; 7.14%), which can be correlated to the advantages of maintaining the structure and mechanical properties of native tissues. Moreover, these biomaterials have a relatively low immune response from the patient, cause minor damage to other bioactive components, and usually do not use chemical reagents to reduce adverse reactions in the body [109][75]. Regarding the compliance of the material, it has been reported that the compliance of this type of graft is close to 9.7%/mmHg, which is a high value compared to previously described materials. However, it has also been reported that most decellularized grafts involve high costs, and they can also require two or more surgeries.

4.3. Current Strategies of TEVGs on In Vitro Testing

To succeed with international standards, various reseauthorchers have overseen the innovation of new techniques, materials, and practices for developing vascular grafts. A total of 46 articlesresearch fulfilled the inclusion criteria regarding TEVGs undergoing in vitro testing. Among the most used materials in in vitro processes, the PCL, TPU (Thermoplastic Polyurethane), and PLGA (poly(lactic-co-glycolic acid)) are the most used, together with natural biomaterials such as collagen and gelatin [109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156][75][76][77][78][79][80][81][82][83][84][85][86][87][88][89][90][91][92][93][94][95][96][97][98][99][100][101][102][103][104][105][106][107][108][109][110][111][112][113][114][115][116][117][118][119][120][121][122]. Based on these biomaterials, there has been a significant trend of various manufacturing methods, in which the electrospinning method is one of the most used. This methodology is based on the formation of nanofibers through the application of electricity to the material. This process is usually used due to its versatility in adjusting different parameters such as porosity, size, thickness, and density of the graft. About 37% of the researticlesch mentioned used the electrospinning method in manufacturing [114,116,118,120,121,122,123,125,127,128,132,133,134,135,137,139,141,142,143,144,146,147,150,151,154,155][80][82][84][86][87][88][89][91][93][94][98][99][100][101][103][105][107][108][109][110][112][113][116][117][120][121]. On the other hand, the second most used process is the Solvent Casting method, with 13.04% of researticlesch using this process [117,136,148,149,156][83][102][114][115][122]. However, different reseauthorchers carry out mechanical and cell viability tests to prove the possible behavior of the grafts in a patient. Due to the mechanical tests, a trend has been found regarding the tests carried out, of which the longitudinal tensile strength tests are most often reported. This measurement helps to determine the vascular graft resistance against longitudinal tensile forces. Along with the revised manuscripts, an average of 94,300 KPa (n = 37) was evidenced, thus indicating that most of the grafts resist high longitudinal forces [113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156][79][80][81][82][83][84][85][86][87][88][89][90][91][92][93][94][95][96][97][98][99][100][101][102][103][104][105][106][107][108][109][110][111][112][113][114][115][116][117][118][119][120][121][122]. Nonetheless, circumferential tensile strength is not often reported, same as compliance, both being highly important data required to establish suitable mechanical properties for TEVGs applications. On the other hand, the second most reported measurement is the Burst Strength since it is essential to determine the pressure that can resist substantial flows. The reseauthorchers recorded a mean value of 2161.14 mmHg (n = 17) [118,120,122,123,126,128,142,143,146,147,151,152,154,155,156,157][84][86][88][89][92][94][108][109][112][113][117][118][120][121][122][123]. Likewise, some researticlesch have also included other relevant tests to comply with international standards. Another of the most important tests is circumferential traction resistance, with an average result of 9210 KPa (n = 13), suture retention resistance with an average result of 7.34 N (n = 11), and finally dynamic compliance with a mean result of 327.28 mmHg (n = 6). These materials show different factors such as longitudinal tensile strength, circumferential tensile strength, suture retention strength, and dynamic compliance, thus making it a material with the necessary characteristics of durability and hardness for a device that is going to be subjected to such a changing environment. It was previously mentioned that the ideal graft should have mechanical characteristics similar to a vein. In this way, it can be argued that the PCL meets these characteristics, even surpassing the behavior of the superior saphenous vein in said properties. For these reasons, there has been a trend among reseauthorchers to use this material by combining it with one that can help improve properties such as burst pressure, and thus generate an ideal replacement material [120,123,124,125,127,132,133,135,139,140,142,145,147,149,154,155,156,157][86][89][90][91][93][98][99][101][105][106][108][111][113][115][120][121][122][123]. Another critical aspect of in vitro tests is cell analysis. Cell viability, cytotoxicity, and cell adhesion, among others, are necessary tests to comply with standards. Thus, the researchers have developed several tests to reach a physiological environment and understand what the possible behavior of the graft will be in an authentic context. This review has found interesting data regarding the biomaterials used for TEVGs that have been reported in in vitro testing. The three most common materials in manufacturing TEGVs and tested in vitro were PCL (37.5%), TPU (8.93%), and PLGA (7.14%). PCL-based TEGVs tend to be more like the internal mammary artery in circumferential (3594 ± 2486 vs. 4100 KPa), longitudinal (4107 ± 2376 vs. 4300 KPa) tensile strengths, and dynamic compliance (5.42 ± 2.65 vs. 5.22%/100 mmHg), respectively. However, its suture resistance strength (4520 ± 4130 KPa) is much higher than that of saphenous vein grafts and the internal mammary artery (3200 and 1350 KPa), respectively, leading to decreased adaptability of the TEVG to suture tension. In addition, 12 out of 22 TEGVs in vitro studies reported the presence of endothelization of the vascular graft lumen. The principal manufacturing technique was electrospinning (16 out of 22) and most products were monolayered (11 out of 22) [118,127,132,137,139,140,141,145,147,154,155,156,157][84][93][98][103][105][106][107][111][113][120][121][122][123]. On the other hand, TPU-based TEGVs showed the highest mechanical properties regarding longitudinal tensile strength (15,750 ± 4546 KPa), suture resistance strength (7860 ± 1600 KPa) compared to saphenous vein grafts (2405 and 3200 KPa), and internal mammary artery (4300 and 1300 KPa), respectively. These results show that TPU is a highly rigid material that does not resemble vascular physiology. However, when used as part of a mixture of other materials, TPU can provide the rigidity necessary for more flexible materials rather than being used alone. Regardless of there being no information regarding dynamic compliance, three out of five TPU-based TEGVs had endothelization. All TEGVs were manufactured by electrospinning [118,137,141,154][84][103][107][120]. Finally, PLGA-based TEGVs had intermediate mechanical properties. Its circumferential tensile strength (8350 KPa) and suture resistance strength (1950 ± 1670 KPa) was higher than the internal mammary artery properties (4100 and 1350 KPa), but lower than the saphenous vein graft (9760 and 3200 KPa), respectively. Its longitudinal tensile strength (5008 ± 3468 KPa) was higher than the saphenous vein graft (2450 KPa) and the internal mammary artery (4300 KPa), but its dynamic compliance (3.41%/100 mmHg) was lower than both vessels (4.4 and 5.22%/100 mmHg). In addition, endothelization was found in two out of four PLGA-based TEGVs and the principal manufacturing technique was electrospinning. These results conclude that PLGA is a rigid material that, as with TPU, can give rigidity to a TEVG, but more flexible materials need to be used to offer compliance to the vascular graft [118,119,137,141,154][84][85][103][107][120].

4.4. Promising Biomaterials and Clinical Practice Correlation on TEVGs Applications

Despite the advances made in TEVGs development, the pursuit of the optimal mechanical properties that approximate those of native arteries is still ongoing. Multiple factors are essential to achieve suitable surgical and mechanical properties, including the proper selection of biomaterials, manufacturing techniques, and surface functionalizations. For instance, new biomaterials have been developed to increase patency rates and avoid complications such as pulmonary thromboembolism [158][124]. In t These sections, we are only talking about newer approaches that have been proposed for use on TEVGs development. The article puresearch published by Bai et al. showed structured scaffolds with biodegradable polyester-polydepsipeptid and silk fibroin (PCL-PIBMD/SF). The manufacturing process was on a sandwich-like composite delivering plasmid complexes aiming to induce endothelialization and was manufactured through layer-by-layer electrospinning and electrospraying techniques. The scaffold was tested with human umbilical vein endothelial cell cultures (HUVECs). The average diameter of nanofibers decreased from 573.8 nm to 285.1 nm, with SF content increasing from 0 to 90%. The mechanical properties found were suitable for vascular scaffolds when the weight ratio of SF was 10%, as the found tensile strength and elongation were 7050.34 kPa and 210 ± 21%. A porosity of 27.5 ± 7.4% with a weight ratio of 90/10 was reported. The HUVECs coverage ratio on day three was 66 ± 3%. This scaffold showed mechanical properties that suggest promising application for TEVGs and also demonstrated the promotion of cell proliferation, adhesion, spreading, and migration [159][125]. Adding to the research, the study published by Xie et al. designed a tissue-engineered vascular scaffold with portulaca flavonoid (PTF) via electrospinning and was integrated with PCL. It was tested on a culture of human vascular smooth muscle cells (HVSMCs). The scaffold exhibited biomimetic net-like fiber structures, an elastic modulus of 2–20 MPa, the ultimate tensile stress of 2000 kPa, and a fracture strain of 60% in the transverse direction. In addition, it showed that compared to the PCL scaffold, the integration of bioactive PTF had better hydrophilicity and degradability. In addition, inhibition of abnormal intimal hyperplasia was observed [160][126]. Moreover, one of the most critical factors in understanding the clinical use of each native artery/vein is the clinical practice guidelines that continue to support using native arteries or veins. However, wresearchers have observed that native arteries/veins are used for the clinical condition and the patient’s requirements, whereas even for the same pathology, different vessels with very different mechanical properties are used. In pathologies involving small vessels, the evidence supports the individualized and specific use of different types of native vessels. The clearest example is the variability between myocardial revascularization per se and infrainguinal revascularization. The ACC/AHA/SCAI Coronary Revascularization Guidelines published in 2021 recommend using the radial artery in isolated Coronary Artery Bypass Grafting (CABG) and the left mammary artery in multiple bypasses over the saphenous vein. The evidence shows that using the left mammary artery prolongs the survival of the patient who only requires isolated CABG in a significantly stenosed non–Left Anterior Descending (LAD) vessel. In addition, the case of the radial artery has shown that in the medium and long term, it has higher patency rates and better clinical results at ten years of undergoing CABG to bypass the LAD [161][127]. On the other hand, the Society for Vascular Surgery practice guidelines for the atherosclerotic occlusive disease of the lower extremities recommends infrainguinal bypass using the saphenous vein [162][128]. Depending on the patient’s need and underlying pathology, this variability in mechanical property requirements also applies to vascular grafts. Thus, evaluating the different mechanical properties in each clinical indication is necessary since it is possible to hypothesize that not only a single small vascular graft with specific mechanical properties will be required.

4.5. Approaches/Techniques for Fabrication of Small Diameter TEVGs

Currently, there is a great diversity of manufacturing techniques for small diameter TEVGs. Several biomaterials have the plasticity required for application on different manufacturing techniques. Most studies reported the use of electrospinning as the primary method of fabrication (n = 25; 58.14%); this is because it offers the ability to fine-tune mechanical properties during the fabrication processes while also offering precise control over the composition, dimension, and alignment of the fiber of the material [66,69,70,72,78,80,81,82,83,84,88,93,95,96,97,99,100,101,103,104,107,110][30][33][34][36][42][44][45][46][47][48][52][57][59][60][61][63][64][65][67][68][71][76]. Furthermore, it can combine synthetic and natural materials, meeting specific needs like high mechanical durability in terms of high burst strength and compliance. Finally, this method allows the incorporation of natural polymers that promote the proliferation of different cell types in the matrix of the graft’s wall. Although electrospinning as the primary manufacturing technique is one of the most common for the development of grafts, the fiber diameters should be considered. The reasoning is that if the diameter is too small, the TEVG will have low porosity, meaning that cell infiltration will be limited. On the other hand, if the diameter of the fiber is too big, the biomechanical graft properties could be compromised due to an increase in the porosity, leading to blood leakages. It has been reported that thicker-fiber grafts (5–6 μm) tend to polarize into the immunomodulatory and tissue remodeling (M2) phenotype, while thinner-fiber grafts (2–3 μm) express a pro-inflammatory (M1) phenotype [111][77]. In this case, the average fiber diameter reported for the PCL TEVGs data was 2.4, indicating that this diameter should be improved to enhance regenerative outcomes. Another relatively common technique typically used for the manufacture of TEVGs is Freeze tawing (n = 3; 6.98%) [67,76,86,163,164,165][31][40][50][129][130][131]; some of the advantages of this method are that the structure of the material is maintained, moisture is removed at low temperatures, the stability of the material is increased during storage, and the fast transition of the moisturized material to be dehydrated minimized several degradation reactions [112][78]. Regarding the use of this method in the manufacture of TEVGs, results have reported that the porosity of the graft is high enough to polarize into the immunomodulatory and tissue remodeling (M2) phenotype, meaning that there should be an improvement in regenerative outcome. Nevertheless, this technique has not been widely used now that some mechanical properties seem to be affected. For example, the circumferential tensile strength seems to be lowered compared to other methods such as electrospinning.
 

References

  1. World Health Organization. Cardiovascular Diseases (CVDs). Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 22 May 2022).
  2. Ralapanawa, U.; Sivakanesan, R. Epidemiology and the Magnitude of Coronary Artery Disease and Acute Coronary Syndrome: A Narrative Review. J. Epidemiol. Glob. Health 2021, 11, 169–177.
  3. Shu, J.; Santulli, G. Update on peripheral artery disease: Epidemiology and evidence-based facts. Atherosclerosis 2018, 275, 379–381.
  4. Royse, A.; Pamment, W.; Pawanis, Z.; Clarke-Errey, S.; Eccleston, D.; Ajani, A.; Wilson, W.; Canty, D.; Royse, C. Patency of conduits in patients who received internal mammary artery, radial artery and saphenous vein grafts. BMC Cardiovasc. Disord. 2020, 20, 148.
  5. Skovrind, I.; Harvald, E.B.; Belling, H.J.; Jørgensen, C.D.; Lindholt, J.S.; Andersen, D.C. Concise Review: Patency of Small-Diameter Tissue-Engineered Vascular Grafts: A Meta-Analysis of Preclinical Trials. STEM CELLS Transl. Med. 2019, 8, 671–680.
  6. MedSuite. Peripheral Vascular Devices Market Analysis, Size, Trends. iData Research: Intellenge behind the Data. Available online: https://idataresearch.com/product/peripheral-vascular-devices-market-united-states/ (accessed on 24 May 2022).
  7. CryoGraft®. Descending Thoracic Aorta. Cryolife: Life Restoring Technology. Available online: https://www.cryolife.com/products/cardiac-allografts/cryograft/ (accessed on 22 May 2022).
  8. Artegraft®. Collagen Vascular Graft*. LeMaitre. Available online: https://www.lemaitre.com/products/artegraft-collagen-vascular-graft (accessed on 22 May 2022).
  9. Aroz, S.G.; Spaggiari, M.; Jeon, H.; Oberholzer, J.; Benedetti, E.; Tzvetanov, I. The use of bovine pericardial patch for vascular reconstruction in infected fields for transplant recipients. J. Vasc. Surg. Cases, Innov. Tech. 2017, 3, 47–49.
  10. Omniflow® II Vascular Prosthesis*. LeMaitre. Available online: https://www.lemaitre.com/products/omniflow-ii-vascular-prosthesis (accessed on 22 May 2022).
  11. Gupta, P.; Mandal, B.B. Tissue-Engineered Vascular Grafts: Emerging Trends and Technologies. Adv. Funct. Mater. 2021, 31, 2100027.
  12. Camasão, D.B.; Mantovani, D. The mechanical characterization of blood vessels and their substitutes in the continuous quest for physiological-relevant performances. A critical review. Mater. Today Bio 2021, 10, 100106.
  13. Rodriguez-Soto, M.A.; Vargas, N.S.; Riveros, A.; Camargo, C.M.; Cruz, J.C.; Sandoval, N.; Briceño, J.C. Failure Analysis of TEVG’s I: Overcoming the Initial Stages of Blood Material Interaction and Stabilization of the Immune Response. Cells 2021, 10, 3140.
  14. Rodriguez-Soto, M.A.; Riveros, A.; Vargas, N.S.; Garcia-Brand, A.J.; Camargo, C.M.; Cruz, J.C.; Sandoval, N.; Briceño, J.C. Failure Analysis of TEVG’s II: Late Failure and Entering the Regeneration Pathway. Cells 2022, 11, 939.
  15. Jeong, Y.; Yao, Y.; Yim, E.K.F. Current understanding of intimal hyperplasia and effect of compliance in synthetic small diameter vascular grafts. Biomater. Sci. 2020, 8, 4383–4395.
  16. Mehta, R.I.; Mukherjee, A.K.; Patterson, T.D.; Fishbein, M.C. Pathology of explanted polytetrafluoroethylene vascular grafts. Cardiovasc. Pathol. 2011, 20, 213–221.
  17. Hamilton, G.; Megerman, J.; L’Italien, G.J.; Warnock, D.F.; Schmitz-Rixen, T.; Brewster, D.C.; Abbott, W.M. Prediction of aneurysm formation in vascular grafts of biologic origin. J. Vasc. Surg. 1988, 7, 400–408.
  18. Post, A.; Diaz-Rodriguez, P.; Balouch, B.; Paulsen, S.; Wu, S.; Miller, J.; Hahn, M.; Cosgriff-Hernandez, E. Elucidating the role of graft compliance mismatch on intimal hyperplasia using an ex vivo organ culture model. Acta Biomater. 2019, 89, 84–94.
  19. He, F.; Hua, L.; Gao, L.-J. A Computational Model for Biomechanical Effects of Arterial Compliance Mismatch. Appl. Bionics Biomech. 2015, 2015, 213236.
  20. Reneman, R.S.; Hoeks, A.P.G. Wall shear stress as measured in vivo: Consequences for the design of the arterial system. Med Biol. Eng. Comput. 2008, 46, 499–507.
  21. Khan, M.O.; Tran, J.S.; Zhu, H.; Boyd, J.; Packard, R.R.S.; Karlsberg, R.P.; Kahn, A.M.; Marsden, A.L. Low Wall Shear Stress Is Associated with Saphenous Vein Graft Stenosis in Patients with Coronary Artery Bypass Grafting. J. Cardiovasc. Transl. Res. 2021, 14, 770–781.
  22. Zhu, L.; Pan, Z.; Li, Z.; Chang, Y.; Zhu, Y.; Yan, F.; Tu, S.; Yang, W. Can the Wall Shear Stress Values of Left Internal Mammary Artery Grafts during the Perioperative Period Reflect the One-Year Patency? Thorac. Cardiovasc. Surg. 2020, 68, 723–729.
  23. Owens, C.D. Adaptive changes in autogenous vein grafts for arterial reconstruction: Clinical implications. J. Vasc. Surg. 2010, 51, 736–746.
  24. Lu, D.; Kassab, G.S. Role of shear stress and stretch in vascular mechanobiology. J. R. Soc. Interface 2011, 8, 1379–1385.
  25. Seo, K.W.; Lee, S.J.; Kim, Y.H.; Bae, J.U.; Park, S.Y.; Bae, S.S.; Kim, C.D. Mechanical Stretch Increases MMP-2 Production in Vascular Smooth Muscle Cells via Activation of PDGFR-β/Akt Signaling Pathway. PLoS ONE 2013, 8, e70437.
  26. Seneviratne, A.N.; Cole, J.; Goddard, M.E.; Park, I.; Mohri, Z.; Sansom, S.; Udalova, I.; Krams, R.; Monaco, C. Low shear stress induces M1 macrophage polarization in murine thin-cap atherosclerotic plaques. J. Mol. Cell. Cardiol. 2015, 89, 168–172.
  27. Seneviratne, A.N.; Cole, J.E.; Goddard, M.E.; Mohri, Z.; Krams, R.; Monaco, C. Macrophage polarisation in shear stress modulated atherosclerotic plaque vulnerability. Atherosclerosis 2012, 225, e2.
  28. ANSI/AAMI/ISO 7198:1998/2001 (R2010); Cardiovascular Implants—Tubular Vascular Prostheses. American National Standards Institute: Arlington, MA, USA, 2010.
  29. ISO 7198:2016; Cardiovascular Implants and Extracorporeal Systems—Vascular Prostheses—Tubular Vascular Grafts and Vascular Patches. ISO: Geneva, Switzerland, 2016.
  30. Lorentz, K.L.; Gupta, P.; Shehabeldin, M.S.; Cunnane, E.M.; Ramaswamy, A.K.; Verdelis, K.; DiLeo, M.V.; Little, S.R.; Weinbaum, J.S.; Sfeir, C.S.; et al. CCL2 loaded microparticles promote acute patency in silk-based vascular grafts implanted in rat aortae. Acta Biomater. 2021, 135, 126–138.
  31. Zhang, C.; Xie, Q.; Cha, R.; Ding, L.; Jia, L.; Mou, L.; Cheng, S.; Wang, N.; Li, Z.; Sun, Y.; et al. Anticoagulant Hydrogel Tubes with Poly(ε-Caprolactone) Sheaths for Small-Diameter Vascular Grafts. Adv. Health Mater. 2021, 10, 2100839.
  32. Antonova, L.; Krivkina, E.; Sevostianova, V.; Mironov, A.; Rezvova, M.; Shabaev, A.; Tkachenko, V.; Krutitskiy, S.; Khanova, M.; Sergeeva, T.; et al. Tissue-Engineered Carotid Artery Interposition Grafts Demonstrate High Primary Patency and Promote Vascular Tissue Regeneration in the Ovine Model. Polymers 2021, 13, 2637.
  33. Wang, Y.; Wu, T.; Zhang, J.; Feng, Z.; Yin, M.; Mo, X. A bilayer vascular scaffold with spatially controlled release of growth factors to enhance in situ rapid endothelialization and smooth muscle regeneration. Mater. Des. 2021, 204, 109649.
  34. Wu, Y.-L.; Szafron, J.M.; Blum, K.M.; Zbinden, J.C.; Khosravi, M.R.; Best, C.A.; Reinhardt, J.W.; Zheng, Q.; Yi, T.; Shinoka, T.; et al. Electrospun Tissue-Engineered Arterial Graft Thickness Affects Long-Term Composition and Mechanics. Tissue Eng. Part A 2020, 27, 593–603.
  35. Kimicata, M.; Allbritton-King, J.D.; Navarro, J.; Santoro, M.; Inoue, T.; Hibino, N.; Fisher, J.P. Assessment of decellularized pericardial extracellular matrix and poly(propylene fumarate) biohybrid for small-diameter vascular graft applications. Acta Biomater. 2020, 110, 68–81.
  36. Shi, J.; Chen, S.; Wang, L.; Zhang, X.; Gao, J.; Jiang, L.; Tang, D.; Zhang, L.; Midgley, A.; Kong, D.; et al. Rapid endothelialization and controlled smooth muscle regeneration by electrospun heparin-loaded polycaprolactone/gelatin hybrid vascular grafts. J. Biomed. Mater. Res. Part B: Appl. Biomater. 2019, 107, 2040–2049.
  37. Nakayama, Y.; Furukoshi, M.; Terazawa, T.; Iwai, R. Development of long in vivo tissue-engineered “Biotube” vascular grafts. Biomaterials 2018, 185, 232–239.
  38. Szafron, J.M.; Khosravi, R.; Reinhardt, J.; Best, C.A.; Bersi, M.R.; Yi, T.; Breuer, C.K.; Humphrey, J.D. Immuno-driven and Mechano-mediated Neotissue Formation in Tissue Engineered Vascular Grafts. Ann. Biomed. Eng. 2018, 46, 1938–1950.
  39. Zhu, M.; Wu, Y.; Li, W.; Dong, X.; Chang, H.; Wang, K.; Wu, P.; Zhang, J.; Fan, G.; Wang, L.; et al. Biodegradable and elastomeric vascular grafts enable vascular remodeling. Biomaterials 2018, 183, 306–318.
  40. Lee, K.-W.; Gade, P.S.; Dong, L.; Zhang, Z.; Aral, A.M.; Gao, J.; Ding, X.; Stowell, C.E.; Nisar, M.U.; Kim, K.; et al. A biodegradable synthetic graft for small arteries matches the performance of autologous vein in rat carotid arteries. Biomaterials 2018, 181, 67–80.
  41. Wang, W.; Nie, W.; Zhou, X.; Feng, W.; Chen, L.; Zhang, Q.; You, Z.; Shi, Q.; Peng, C.; He, C. Fabrication of heterogeneous porous bilayered nanofibrous vascular grafts by two-step phase separation technique. Acta Biomater. 2018, 79, 168–181.
  42. Wu, T.; Zhang, J.; Wang, Y.; Sun, B.; Yin, M.; Bowlin, G.L.; Mo, X. Design and Fabrication of a Biomimetic Vascular Scaffold Promoting in Situ Endothelialization and Tunica Media Regeneration. ACS Appl. Bio Mater. 2018, 1, 833–844.
  43. Huang, R.; Gao, X.; Wang, J.; Chen, H.; Tong, C.; Tan, Y.; Tan, Z. Triple-Layer Vascular Grafts Fabricated by Combined E-Jet 3D Printing and Electrospinning. Ann. Biomed. Eng. 2018, 46, 1254–1266.
  44. Duijvelshoff, R.; Van Engeland, N.C.A.; Gabriels, K.M.R.; Söntjens, S.H.M.; Smits, A.I.P.M.; Dankers, P.Y.W.; Bouten, C.V.C. Host Response and Neo-Tissue Development during Resorption of a Fast Degrading Supramolecular Electrospun Arterial Scaffold. Bioengineering 2018, 5, 61.
  45. Jirofti, N.; Mohebbi-Kalhori, D.; Samimi, A.; Hadjizadeh, A.; Kazemzadeh, G.H. Small-diameter vascular graft using co-electrospun composite PCL/PU nanofibers. Biomed. Mater. 2018, 13, 055014.
  46. Shafiq, M.; Zhang, Q.; Zhi, D.; Wang, K.; Kong, D.; Kim, D.-H.; Kim, S.H. In Situ Blood Vessel Regeneration Using SP (Substance P) and SDF (Stromal Cell–Derived Factor)-1α Peptide Eluting Vascular Grafts. Arter. Thromb. Vasc. Biol. 2018, 38, e117–e134.
  47. Gao, J.; Jiang, L.; Liang, Q.; Shi, J.; Hou, D.; Tang, D.; Chen, S.; Kong, D.; Wang, S. The grafts modified by heparinization and catalytic nitric oxide generation used for vascular implantation in rats. Regen. Biomater. 2018, 5, 105–114.
  48. Wu, T.; Zhang, J.; Wang, Y.; Li, D.; Sun, B.; El-Hamshary, H.; Yin, M.; Mo, X. Fabrication and preliminary study of a biomimetic tri-layer tubular graft based on fibers and fiber yarns for vascular tissue engineering. Mater. Sci. Eng. C 2017, 82, 121–129.
  49. Jiang, B.; Suen, R.; Wang, J.-J.; Zhang, Z.J.; Wertheim, J.A.; Ameer, G.A. Vascular scaffolds with enhanced antioxidant activity inhibit graft calcification. Biomaterials 2017, 144, 166–175.
  50. Aussel, A.; Thébaud, N.B.; Bérard, X.; Brizzi, V.; Delmond, S.; Bareille, R.; Siadous, R.; James, C.; Ripoche, J.; Durand, M.; et al. Chitosan-based hydrogels for developing a small-diameter vascular graft: In vitro and in vivo evaluation. Biomed. Mater. 2017, 12, 065003.
  51. Kristofik, N.J.; Qin, L.; Calabro, N.E.; Dimitrievska, S.; Li, G.; Tellides, G.; Niklason, L.E.; Kyriakides, T.R. Improving in vivo outcomes of decellularized vascular grafts via incorporation of a novel extracellular matrix. Biomaterials 2017, 141, 63–73.
  52. Wang, Z.; Wu, Y.; Wang, J.; Zhang, C.; Yan, H.; Zhu, M.; Wang, K.; Li, C.; Xu, Q.; Kong, D. Effect of Resveratrol on Modulation of Endothelial Cells and Macrophages for Rapid Vascular Regeneration from Electrospun Poly(ε-caprolactone) Scaffolds. ACS Appl. Mater. Interfaces 2017, 9, 19541–19551.
  53. Emechebe, A.G.; Obiweluozor, O.F.; Jeong, I.S.; Park, J.-K.; Park, C.H.; Kim, C.S. Merging 3D printing with electrospun biodegradable small-caliber vascular grafts immobilized with VEGF. Nanomed. Nanotechnol. Biol. Med. 2020, 30, 102306.
  54. Wang, H.; Xing, M.; Deng, W.; Qian, M.; Wang, F.; Wang, K.; Midgley, A.C.; Zhao, Q. Anti-Sca-1 antibody-functionalized vascular grafts improve vascular regeneration via selective capture of endogenous vascular stem/progenitor cells. Bioact. Mater. 2022, 16, 433–450.
  55. Inoue, T.; Kanda, K.; Yamanami, M.; Kami, D.; Gojo, S.; Yaku, H. Modifications of the mechanical properties of in vivo tissue-engineered vascular grafts by chemical treatments for a short duration. PLoS ONE 2021, 16, e0248346.
  56. Antonova, L.; Kutikhin, A.; Sevostianova, V.; Velikanova, E.; Matveeva, V.; Glushkova, T.; Mironov, A.; Krivkina, E.; Shabaev, A.; Senokosova, E.; et al. bFGF and SDF-1α Improve In Vivo Performance of VEGF-Incorporating Small-Diameter Vascular Grafts. Pharmaceuticals 2021, 14, 302.
  57. Guo, H.-F.; Dai, W.-W.; Qian, D.-H.; Qin, Z.-X.; Lei, Y.; Hou, X.-Y.; Wen, C. A simply prepared small-diameter artificial blood vessel that promotes in situ endothelialization. Acta Biomater. 2017, 54, 107–116.
  58. Negishi, J.; Hashimoto, Y.; Yamashita, A.; Zhang, Y.; Kimura, T.; Kishida, A.; Funamoto, S. Evaluation of small-diameter vascular grafts reconstructed from decellularized aorta sheets: Vascular grafts from decellularized aorta sheets. J. Biomed. Mater. Res. Part A 2017, 105, 1293–1298.
  59. Stowell, C.E.; Li, X.; Matsunaga, M.H.; Cockreham, C.B.; Kelly, K.M.; Cheetham, J.; Tzeng, E.; Wang, Y. Resorbable vascular grafts show rapid cellularization and degradation in the ovine carotid. J. Tissue Eng. Regen. Med. 2020, 14, 1673–1684.
  60. Matsuzaki, Y.; Iwaki, R.; Reinhardt, J.W.; Chang, Y.-C.; Miyamoto, S.; Kelly, J.; Zbinden, J.; Blum, K.; Mirhaidari, G.; Ulziibayar, A.; et al. The effect of pore diameter on neo-tissue formation in electrospun biodegradable tissue-engineered arterial grafts in a large animal model. Acta Biomater. 2020, 115, 176–184.
  61. Wen, M.; Zhi, D.; Wang, L.; Cui, C.; Huang, Z.; Zhao, Y.; Wang, K.; Kong, D.; Yuan, X. Local Delivery of Dual MicroRNAs in Trilayered Electrospun Grafts for Vascular Regeneration. ACS Appl. Mater. Interfaces 2020, 12, 6863–6875.
  62. Fukunishi, T.; Ong, C.S.; Yesantharao, P.; Best, C.A.; Yi, T.; Zhang, H.; Mattson, G.; Boktor, J.; Nelson, K.; Shinoka, T.; et al. Different degradation rates of nanofiber vascular grafts in small and large animal models. J. Tissue Eng. Regen. Med. 2019, 14, 203–214.
  63. Shojaee, M.; Sameti, M.; Vuppuluri, K.; Ziff, M.; Carriero, A.; Bashur, C.A. Design and characterization of a porous pouch to prevent peritoneal adhesions during in vivo vascular graft maturation. J. Mech. Behav. Biomed. Mater. 2020, 102, 103461.
  64. Yang, L.; Li, X.; Wu, Y.; Du, P.; Sun, L.; Yu, Z.; Song, S.; Yin, J.; Ma, X.; Jing, C.; et al. Preparation of PU/Fibrin Vascular Scaffold with Good Biomechanical Properties and Evaluation of Its Performance in vitro and in vivo. Int. J. Nanomed. 2020, 15, 8697–8715.
  65. Gostev, A.A.; Chernonosova, V.S.; Murashov, I.S.; Sergeevichev, D.S.; Korobeinikov, A.A.; Karaskov, A.M.; Karpenko, A.A.; Laktionov, P.P. Electrospun polyurethane-based vascular grafts: Physicochemical properties and functioning in vivo. Biomed. Mater. 2020, 15, 015010.
  66. Li, W.; Wu, P.; Zhang, Y.; Midgley, A.C.; Yuan, X.; Wu, Y.; Wang, L.; Wang, Z.; Zhu, M.F.; Kong, D. Bilayered Polymeric Micro- and Nanofiber Vascular Grafts as Abdominal Aorta Replacements: Long-Term in Vivo Studies in a Rat Model. ACS Appl. Bio Mater. 2019, 2, 4493–4502.
  67. Yang, Y.; Lei, D.; Zou, H.; Huang, S.; Yang, Q.; Li, S.; Qing, F.-L.; Ye, X.; You, Z.; Zhao, Q. Hybrid electrospun rapamycin-loaded small-diameter decellularized vascular grafts effectively inhibit intimal hyperplasia. Acta Biomater. 2019, 97, 321–332.
  68. Johnson, R.; Ding, Y.; Nagiah, N.; Monnet, E.; Tan, W. Coaxially-structured fibres with tailored material properties for vascular graft implant. Mater. Sci. Eng. C 2019, 97, 1–11.
  69. Yang, S.; Zheng, X.; Qian, M.; Wang, H.; Wang, F.; Wei, Y.; Midgley, A.C.; He, J.; Tian, H.; Zhao, Q. Nitrate-Functionalized poly(ε-Caprolactone) Small-Diameter Vascular Grafts Enhance Vascular Regeneration via Sustained Release of Nitric Oxide. Front. Bioeng. Biotechnol. 2021, 9, 770121.
  70. Wang, C.; Li, Z.; Zhang, L.; Sun, W.; Zhou, J. Long-term results of triple-layered small diameter vascular grafts in sheep carotid arteries. Med. Eng. Phys. 2020, 85, 1–6.
  71. Hui, X.; Geng, X.; Jia, L.; Xu, Z.; Ye, L.; Gu, Y.; Zhang, A.-Y.; Feng, Z.-G. Preparation and in vivo evaluation of surface heparinized small diameter tissue engineered vascular scaffolds of poly(ε-caprolactone) embedded with collagen suture. J. Biomater. Appl. 2020, 34, 812–826.
  72. Soccerplots 1.0.0. PyPI. Available online: https://pypi.org/project/soccerplots/ (accessed on 29 May 2022).
  73. Yang, X.; Wang, Y.; Zhou, Y.; Chen, J.; Wan, Q. The Application of Polycaprolactone in Three-Dimensional Printing Scaffolds for Bone Tissue Engineering. Polymers 2021, 13, 2754.
  74. Kim, L.; Scott, R.A.P.; Ashton, H.A.; Thompson, S.G. For the Multicentre Aneurysm Screening Study Group A Sustained Mortality Benefit from Screening for Abdominal Aortic Aneurysm. Ann. Intern. Med. 2007, 146, 699–706.
  75. Liao, J.; Xu, B.; Zhang, R.; Fan, Y.; Xie, H.; Li, X. Applications of decellularized materials in tissue engineering: Advantages, drawbacks and current improvements, and future perspectives. J. Mater. Chem. B 2020, 8, 10023–10049.
  76. Wan, Y.; Yang, S.; Peng, M.; Gama, M.; Yang, Z.; Deng, X.; Zhou, J.; Ouyang, C.; Luo, H. Controllable synthesis of biomimetic nano/submicro-fibrous tubes for potential small-diameter vascular grafts. J. Mater. Chem. B 2020, 8, 5694–5706.
  77. Wang, Z.; Cui, Y.; Wang, J.; Yang, X.; Wu, Y.; Wang, K.; Gao, X.; Li, D.; Li, Y.; Zheng, X.-L.; et al. The effect of thick fibers and large pores of electrospun poly(ε-caprolactone) vascular grafts on macrophage polarization and arterial regeneration. Biomaterials 2014, 35, 5700–5710.
  78. Boss, E.A.; Filho, R.M.; de Toledo, E.C.V. Freeze drying process: Real time model and optimization. Chem. Eng. Process. Process Intensif. 2004, 43, 1475–1485.
  79. Li, C.; Wang, F.; Chen, P.; Zhang, Z.; Guidoin, R.; Wang, L. Preventing collapsing of vascular scaffolds: The mechanical behavior of PLA/PCL composite structure prostheses during in vitro degradation. J. Mech. Behav. Biomed. Mater. 2017, 75, 455–462.
  80. Lee, S.J.; Yoo, J.J.; Lim, G.J.; Atala, A.; Stitzel, J. In vitro evaluation of electrospun nanofiber scaffolds for vascular graft application. J. Biomed. Mater. Res. Part A 2007, 83A, 999–1008.
  81. Li, C.; Wang, F.; Douglas, G.; Zhang, Z.; Guidoin, R.; Wang, L. Comprehensive mechanical characterization of PLA fabric combined with PCL to form a composite structure vascular graft. J. Mech. Behav. Biomed. Mater. 2017, 69, 39–49.
  82. Zhang, Y.; Li, X.S.; Guex, A.G.; Liu, S.S.; Müller, E.; Malini, R.I.; Zhao, H.J.; Rottmar, M.; Maniura, K.; Rossi, R.; et al. A compliant and biomimetic three-layered vascular graft for small blood vessels. Biofabrication 2017, 9, 025010.
  83. Wang, N.; Zheng, W.; Cheng, S.; Zhang, W.; Liu, S.; Jiang, X. In Vitro Evaluation of Essential Mechanical Properties and Cell Behaviors of a Novel Polylactic-co-Glycolic Acid (PLGA)-Based Tubular Scaffold for Small-Diameter Vascular Tissue Engineering. Polymers 2017, 9, 318.
  84. Maleki, S.; Shamloo, A.; Kalantarnia, F. Tubular TPU/SF nanofibers covered with chitosan-based hydrogels as small-diameter vascular grafts with enhanced mechanical properties. Sci. Rep. 2022, 12, 6179.
  85. Du, J.; Hu, X.; Su, Y.; Wei, T.; Jiao, Z.; Liu, T.; Wang, H.; Nie, Y.; Li, X.; Song, K. Gelatin/sodium alginate hydrogel-coated decellularized porcine coronary artery to construct bilayer tissue engineered blood vessels. Int. J. Biol. Macromol. 2022, 209, 2070–2083.
  86. Pimenta, F.A.; Carbonari, R.C.; Malmonge, S.M. Nanofibrous tubular scaffolds for tissue engineering of small-diameter vascular grafts—Development using SBS fabrication technique and mechanical performance. Res. Biomed. Eng. 2022, 1–15.
  87. Zakeri, Z.; Salehi, R.; Mahkam, M.; Rahbarghazi, R.; Abbasi, F.; Rezaei, M. Electrospun POSS integrated poly(carbonate-urea)urethane provides appropriate surface and mechanical properties for the fabrication of small-diameter vascular grafts. J. Biomater. Sci. Polym. Ed. 2022, 1–20.
  88. Zhang, X.; Bai, S.; Zang, L.; Chen, X.; Yuan, X. Preparation of Poly(ε-caprolactone)/Poly(ester amide) Electrospun Membranes for Vascular Repair. Chem. Res. Chin. Univ. 2022, 1–7.
  89. Radakovic, D.; Reboredo, J.; Helm, M.; Weigel, T.; Schürlein, S.; Kupczyk, E.; Leyh, R.G.; Walles, H.; Hansmann, J. A multilayered electrospun graft as vascular access for hemodialysis. PLoS ONE 2017, 12, e0185916.
  90. Cao, Y.; Jiang, J.; Jiang, Y.; Li, Z.; Hou, J.; Li, Q. Biodegradable highly porous interconnected poly(ε-caprolactone)/poly(L-lactide-co-ε-caprolactone) scaffolds by supercritical foaming for small-diameter vascular tissue engineering. Polym. Adv. Technol. 2021, 33, 440–451.
  91. Montoya, Y.; Cardenas, J.; Bustamante, J.; Valencia, R. Effect of sequential electrospinning and co-electrospinning on morphological and fluid mechanical wall properties of polycaprolactone and bovine gelatin scaffolds, for potential use in small diameter vascular grafts. Biomater. Res. 2021, 25, 38.
  92. Helms, F.; Haverich, A.; Böer, U.; Wilhelmi, M. Transluminal compression increases mechanical stability, stiffness and endothelialization capacity of fibrin-based bioartificial blood vessels. J. Mech. Behav. Biomed. Mater. 2021, 124, 104835.
  93. Sun, B.; Hou, L.; Sun, B.; Han, Y.; Zou, Y.; Huang, J.; Zhang, Y.; Feng, C.; Dou, X.; Xu, F. Use of Electrospun Phenylalanine/Poly-ε-Caprolactone Chiral Hybrid Scaffolds to Promote Endothelial Remodeling. Front. Bioeng. Biotechnol. 2021, 9, 773635.
  94. Yang, T.; Xu, H.; Zhao, C.; Tang, D.; Mu, F.; Lu, H.; Rao, Z.; Wang, S. Construction and evaluation of co-electrospun poly (butylene succinate)/gelatin materials as potential vascular grafts. Chin. J. Chem. Eng. 2021, 39, 297–305.
  95. Frazão, L.P.; Fernandes, A.M.; Oliveira, C.; Martins, A.; Silva, T.H.; de Castro, J.V.; Nogueira-Silva, C.; Neves, N.M. New Vascular Graft Using the Decellularized Human Chorion Membrane. ACS Biomater. Sci. Eng. 2021, 7, 3423–3433.
  96. Asadpour, S.; Yeganeh, H.; Ai, J.; Ghanbari, H. A novel polyurethane modified with biomacromolecules for small-diameter vascular graft applications. J. Mater. Sci. 2018, 53, 9913–9927.
  97. Silva, J.M.; Rodrigues, L.C.; Silva, S.S.; Reis, R.L.; Duarte, A.R.C. Engineered tubular structures based on chitosan for tissue engineering applications. J. Biomater. Appl. 2018, 32, 841–852.
  98. Kharazi, A.Z.; Atari, M.; Vatankhah, E.; Javanmard, S.H. A nanofibrous bilayered scaffold for tissue engineering of small-diameter blood vessels. Polym. Adv. Technol. 2018, 29, 3151–3158.
  99. Horakova, J.; Mikes, P.; Saman, A.; Jencova, V.; Klapstova, A.; Svarcova, T.; Ackermann, M.; Novotny, V.; Suchy, T.; Lukas, D. The effect of ethylene oxide sterilization on electrospun vascular grafts made from biodegradable polyesters. Mater. Sci. Eng. C 2018, 92, 132–142.
  100. Braghirolli, D.; Caberlon, B.; Gamba, D.; Petry, J.; Dias, M.; Pranke, P. Poly(trimethylene carbonate-co-L-lactide) electrospun scaffolds for use as vascular grafts. Braz. J. Med. Biol. Res. 2019, 52, e8318.
  101. Wang, Y.; He, C.; Feng, Y.; Yang, Y.; Wei, Z.; Zhao, W.; Zhao, C. A chitosan modified asymmetric small-diameter vascular graft with anti-thrombotic and anti-bacterial functions for vascular tissue engineering. J. Mater. Chem. B 2020, 8, 568–577.
  102. Generali, M.; Casanova, E.A.; Kehl, D.; Wanner, D.; Hoerstrup, S.P.; Cinelli, P.; Weber, B. Autologous endothelialized small-caliber vascular grafts engineered from blood-derived induced pluripotent stem cells. Acta Biomater. 2019, 97, 333–343.
  103. Shokraei, N.; Asadpour, S.; Shokraei, S.; Sabet, M.N.; Faridi-Majidi, R.; Ghanbari, H. Development of electrically conductive hybrid nanofibers based on CNT-polyurethane nanocomposite for cardiac tissue engineering. Microsc. Res. Tech. 2019, 82, 1316–1325.
  104. Nguyen, T.-U.; Shojaee, M.; Bashur, C.; Kishore, V. Electrochemical fabrication of a biomimetic elastin-containing bi-layered scaffold for vascular tissue engineering. Biofabrication 2018, 11, 015007.
  105. Rychter, M.; Milanowski, B.; Grześkowiak, B.F.; Jarek, M.; Kempiński, M.; Coy, E.L.; Borysiak, S.; Baranowska-Korczyc, A.; Lulek, J. Cilostazol-loaded electrospun three-dimensional systems for potential cardiovascular application: Effect of fibers hydrophilization on drug release, and cytocompatibility. J. Colloid Interface Sci. 2019, 536, 310–327.
  106. Zhang, X.; Shi, J.; Chen, S.; Dong, Y.; Zhang, L.; Midgley, A.C.; Kong, D.; Wang, S. Polycaprolactone/gelatin degradable vascular grafts simulating endothelium functions modified by nitric oxide generation. Regen. Med. 2019, 14, 1089–1105.
  107. Yan, S.; Napiwocki, B.; Xu, Y.; Zhang, J.; Zhang, X.; Wang, X.; Crone, W.C.; Li, Q.; Turng, L.-S. Wavy small-diameter vascular graft made of eggshell membrane and thermoplastic polyurethane. Mater. Sci. Eng. C 2019, 107, 110311.
  108. Li, X.; Huang, L.; Li, L.; Tang, Y.; Liu, Q.; Xie, H.; Tian, J.; Zhou, S.; Tang, G. Biomimetic dual-oriented/bilayered electrospun scaffold for vascular tissue engineering. J. Biomater. Sci. Polym. Ed. 2019, 31, 439–455.
  109. Hu, Q.; Wu, C.; Zhang, H. Preparation and Optimization of a Biomimetic Triple-Layered Vascular Scaffold Based on Coaxial Electrospinning. Appl. Biochem. Biotechnol. 2020, 190, 1106–1123.
  110. Wu, P.; Wang, L.; Li, W.; Zhang, Y.; Wu, Y.; Zhi, D.; Wang, H.; Wang, L.; Kong, D.; Zhu, M. Construction of vascular graft with circumferentially oriented microchannels for improving artery regeneration. Biomaterials 2020, 242, 119922.
  111. Oliveira, S.; Felizardo, T.; Amorim, S.; Mithieux, S.M.; Pires, R.; Reis, R.L.; Martins, A.; Weiss, A.; Neves, N.M. Tubular fibrous scaffold functionalized with tropoelastin as a small-diameter vascular graft. Biomacromolecules 2020, 21, 3582–3595.
  112. Hodge, J.; Quint, C. Tissue engineered vessel from a biodegradable electrospun scaffold stimulated with mechanical stretch. Biomed. Mater. 2020, 15, 055006.
  113. Woods, I.; Black, A.; Molloy, E.J.; Jockenhoevel, S.; Flanagan, T.C. Fabrication of blood-derived elastogenic vascular grafts using electrospun fibrinogen and polycaprolactone composite scaffolds for paediatric applications. J. Tissue Eng. Regen. Med. 2020, 14, 1281–1295.
  114. Ryan, A.J.; Ryan, E.J.; Cameron, A.R.; O’Brien, F.J. Hierarchical biofabrication of biomimetic collagen-elastin vascular grafts with controllable properties via lyophilisation. Acta Biomater. 2020, 112, 52–61.
  115. Le, A.N.-M.; Tran, N.M.-P.; Phan, T.B.; Tran, P.A.; Tran, L.D.; Nguyen, T.H. Poloxamer additive as luminal surface modification to modulate wettability and bioactivities of small-diameter polyurethane/polycaprolactone electrospun hollow tube for vascular prosthesis applications. Mater. Today Commun. 2021, 26, 101771.
  116. Zhang, Y.; Liu, Y.; Jiang, Z.; Wang, J.; Xu, Z.; Meng, K.; Zhao, H. Poly(glyceryl sebacate)/silk fibroin small-diameter artificial blood vessels with good elasticity and compliance. Smart Mater. Med. 2021, 2, 74–86.
  117. Hu, Q.; Shen, Z.; Zhang, H.; Liu, S.; Feng, R.; Feng, J.; Ramalingam, M. Designed and fabrication of triple-layered vascular scaffold with microchannels. J. Biomater. Sci. Polym. Ed. 2021, 32, 714–734.
  118. Zhang, F.; Bambharoliya, T.; Xie, Y.; Liu, L.; Celik, H.; Wang, L.; Akkus, O.; King, M.W. A hybrid vascular graft harnessing the superior mechanical properties of synthetic fibers and the biological performance of collagen filaments. Mater. Sci. Eng. C 2021, 118, 111418.
  119. Kuźmińska, A.; Kwarta, D.; Ciach, T.; Butruk-Raszeja, B. Cylindrical Polyurethane Scaffold Fabricated Using the Phase Inversion Method: Influence of Process Parameters on Scaffolds’ Morphology and Mechanical Properties. Materials 2021, 14, 2977.
  120. Chen, X.; Yao, Y.; Liu, S.; Hu, Q. An integrated strategy for designing and fabricating triple-layer vascular graft with oriented microgrooves to promote endothelialization. J. Biomater. Appl. 2021, 36, 297–310.
  121. Malik, S.; Sundarrajan, S.; Hussain, T.; Nazir, A.; Ramakrishna, S. Fabrication of Highly Oriented Cylindrical Polyacrylonitrile, Poly(lactide-co-glycolide), Polycaprolactone and Poly(vinyl acetate) Nanofibers for Vascular Graft Applications. Polymers 2021, 13, 2075.
  122. Zumbardo-Bacelis, G.A.; Meza-Villegas, L.A.; Pérez-Aranda, C.A.; Vargas-Coronado, R.; Castillo-Cruz, O.; Montaño-Machado, V.; Mantovani, D.; Cauich-Rodríguez, J.V. On arginine-based polyurethane-blends specific to vascular prostheses. J. Appl. Polym. Sci. 2021, 138, 51247.
  123. Wang, D.; Xu, Y.; Li, Q.; Turng, L.-S. Artificial small-diameter blood vessels: Materials, fabrication, surface modification, mechanical properties, and bioactive functionalities. J. Mater. Chem. B 2020, 8, 1801–1822.
  124. Stoiber, M.; Grasl, C.; Frieberger, K.; Moscato, F.; Bergmeister, H.; Schima, H. Impact of the testing protocol on the mechanical characterization of small diameter electrospun vascular grafts. J. Mech. Behav. Biomed. Mater. 2020, 104, 103652.
  125. Bai, L.; Li, Q.; Duo, X.; Hao, X.; Zhang, W.; Shi, C.; Guo, J.; Ren, X.; Feng, Y. Electrospun PCL-PIBMD/SF blend scaffolds with plasmid complexes for endothelial cell proliferation. RSC Adv. 2017, 7, 39452–39464.
  126. Xie, C.; Guo, T.; Wang, W.; Li, G.; Cai, Z.; Chen, S.; Wang, X.; Liu, Z.; Wang, Z. Scaffold engineering with flavone-modified biomimetic architecture for vascular tissue engineering applications. Tissue Eng. Regen. Med. 2022.
  127. Lawton, J.S.; Tamis-Holland, J.E.; Bangalore, S.; Bates, E.R.; Beckie, T.M.; Bischoff, J.M.; Bittl, J.A.; Cohen, M.G.; DiMaio, J.M.; Don, C.W.; et al. 2021 ACC/AHA/SCAI Guideline for Coronary Artery Revascularization. J. Am. Coll. Cardiol. 2022, 79, e21–e129.
  128. Conte, M.S.; Pomposelli, F.B.; Clair, D.G.; Geraghty, P.J.; McKinsey, J.F.; Mills, J.L.; Moneta, G.L.; Murad, M.H.; Powell, R.J.; Reed, A.B.; et al. Society for Vascular Surgery practice guidelines for atherosclerotic occlusive disease of the lower extremities: Management of asymptomatic disease and claudication. J. Vasc. Surg. 2015, 61, 2S–41S.
  129. Durán-Rey, D.; Crisóstomo, V.; Sánchez-Margallo, J.A.; Sánchez-Margallo, F.M. Systematic Review of Tissue-Engineered Vascular Grafts. Front. Bioeng. Biotechnol. 2021, 9, 771400.
  130. Weekes, A.; Bartnikowski, N.; Pinto, N.; Jenkins, J.; Meinert, C.; Klein, T.J. Biofabrication of small diameter tissue-engineered vascular grafts. Acta Biomater. 2022, 138, 92–111.
  131. Jafarihaghighi, F.; Ardjmand, M.; Mirzadeh, A.; Hassani, M.S.; Parizi, S.S. Current challenges and future trends in manufacturing small diameter artificial vascular grafts in bioreactors. Cell Tissue Bank. 2020, 21, 377–403.
More
Video Production Service