Next Article in Journal
Dimensional Optimization of TiO2 Nanodisk Photonic Crystals on Lead Iodide (MAPbI3) Perovskite Solar Cells by Using FDTD Simulations
Next Article in Special Issue
Characterizing the Mechanical Performance of a Bare-Metal Stent with an Auxetic Cell Geometry
Previous Article in Journal
Experimental Study on Microstructure of Unsaturated Expansive Soil Improved by MICP Method
Previous Article in Special Issue
Feasibility of a Drug-Releasing Radiofrequency Ablation System in a Porcine Liver Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Interwoven versus Knitted Self-Expandable Metallic Stents: A Comparison Study of Stent-Induced Tissue Hyperplasia in the Rat Esophagus

1
Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea
2
Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Appl. Sci. 2022, 12(1), 346; https://doi.org/10.3390/app12010346
Submission received: 23 November 2021 / Revised: 22 December 2021 / Accepted: 28 December 2021 / Published: 30 December 2021

Abstract

:
This study investigated whether interwoven self-expandable metallic stent (I-SEMS) placement suppresses stent-induced tissue hyperplasia compared with conventional knitted self-expandable metallic stent (K-SEMS) placement in a rat esophageal model. Twelve rats were randomly assigned to the I-SEMS (n = 6) and K-SEMS (n = 6) groups. All rats were sacrificed 4 weeks after the stent placement. The degree of stent-induced tissue hyperplasia on esophagography and histologic examination was compared between the groups. Stent placement was technically successful in all rats. Esophagography revealed significantly greater mean luminal diameter of the stented esophagus in the I-SEMS group than in the K-SEMS group (p < 0.001). Histologic examination revealed a significantly lower percentage of tissue hyperplasia area and a significantly thinner submucosal fibrosis in the I-SEMS group than in the K-SEMS group (all p < 0.001). There were no significant differences in the thickness of the epithelial layers (p = 0.290) and degree of inflammatory cell infiltration (p = 0.506). Formation of stent-induced tissue hyperplasia was evident with both I-SEMS and K-SEMS. Placement of I-SEMSs with a small cell size and high flexibility seems to be effective in suppressing stent-induced tissue hyperplasia compared with placement of K-SEMSs in rat esophageal models.

1. Introduction

Placement of hand-crafted and knitted self-expandable metallic stents (SEMSs; K-SEMSs) is the most common therapeutic strategy for malignant or benign esophageal strictures [1,2,3]. However, excessive proliferative response to mechanical injury is a consequential obstacle for successful treatment [3,4,5]. Fully or partially covered SEMSs have been developed for esophageal strictures to overcome this problem [6,7,8]. Although the covering membrane could prevent tissue hyperplasia through the wire meshes, granulation tissue formation could still occur at the uncovered ends of the stent [8,9]. Furthermore, covered SEMS placement significantly increases the stent migration rate, and tissue ingrowth through the wire mesh can occur through the disrupted covering membrane [8,10]. Stent-induced tissue hyperplasia can lead to recurrent symptoms and technical difficulties in stent removal [2,3].
An interwoven SEMS (I-SEMS) is a weave nitinol alloy stent designed to resist the unique stressor in vascular stenosis [11,12]. Its helical interwoven structure and small-sized cells help to minimize the stress on the entire strut when it is bent, making the stent highly flexible [12,13]. Additionally, this design makes the I-SEMS resistant to torsion caused by friction between the nitinol wires [14]. Compared with the design of conventional K-SEMSs, that of I-SEMSs increases the resistance for kinking owing to the structure with a smaller size of cells, thereby preventing stent-related complications, such as stent fracture and stent-in-restenosis. The K-SEMS is a closed-cell design, and it is a knitted design by joining the intersection of the stent wires. This closed-cell design gives high radial force and uniformity but limited flexibility [13]. On the other hand, open-cell stents have the feature of increasing flexibility and low radial force by sacrificing strut uniformity by eliminating the intersection of the stent wire. Although the I-SEMS structure has a lower radial force than the K-SEMS designed with the same number of wires, it combines the advantages of a closed-cell stent and an open-cell stent to provide high flexibility while maintaining a uniform cell size [15]. Armstrong et al. reported that I-SEMS placement improved stent patency and decreased the stent fracture rates compared with conventional K-SEMS placement in the femoropopliteal segment [16]. Despite their better performance in the vascular lumen, no study has identified the value of I-SEMSs with small-sized cells in suppressing stent-induced tissue hyperplasia in non-vascular luminal organs. We hypothesized that I-SEMS placement would reduce stent-induced tissue hyperplasia owing to the small size of the stent cell. Therefore, the purpose of this study was to investigate whether I-SEMS placement suppresses stent-induced tissue hyperplasia formation compared with conventional K-SEMS placement in a rat esophageal model.

2. Materials and Methods

2.1. Stent Preparation

The I-SEMSs used were designed and manufactured by S&G Biotech Co., Ltd. (Yongin, Korea). A total of 32 nitinol wires with a thickness of 0.09 mm were interwoven using a braiding machine. When fully expanded, the I-SEMSs used for the rat esophagus were 5 mm in diameter and 10 mm in length. One gold wire was wrapped around the stent in a spiral shape to facilitate precise placement under fluoroscopic guidance. Having the same size at full expansion, the K-SEMSs were knitted from a single thread of 0.127 mm-thick nitinol wire filament into a tubular configuration with six bent points. The stent delivery system consisted of a 6 Fr Teflon sheath (Cook, Bloomington, IN, USA) and a pusher catheter. To match the radial force of the two types of SEMSs, different wire thickness was used.

2.2. Stent Cell Size Measurement and Bending Test

The I-SEMSs and knitted SEMSs used for measurement of the stent cell size and the bending test were 5 mm in diameter and 50 mm in length with 10 samples each. Images of the two types of SEMSs were obtained and analyzed using the ImageJ 1.53c software (National Institutes of Health, Bethesda, MD, USA) to measure the cell size of the stents. The areas of the cell size were calculated, and the average value at 10 points from the proximal to the distal ends of the stents was compared between the two groups.
The modeling of the bending test phantom was designed and manufactured by a local manufacturer (ANYMEDI, Seoul, Korea). The phantom was made of a photocurable resin material (Xiamen Zhisen Electro, Xiamen, China) consisting of a body part and two fixation parts. It was 150 mm in length, 60 mm in width, and 50 mm in height. The fixation parts had a central cylindrical bar to fix the stent. The bending test was performed to evaluate and compare the degree of stent deformation, such as bending and kinking, of the I-SEMSs and K-SEMSs. The proximal and distal ends of the stent samples were placed in each cylindrical bar of the phantom 20 mm apart.

2.3. Animal Study Design

This study was approved by the Institutional Animal Care and Use Committee of our center and conformed to US National Institutes of Health guidelines for humane handling of laboratory animals. A total of 12 Sprague–Dawley rats weighing 330–362 g (median weight = 345.5 g) were used in this study. The rats were randomly assigned to two groups: an I-SEMS group (n = 6) and a K-SEMS group (n = 6). All rats were provided with food and water ad libitum and maintained at 24 ± 2 °C with a 12 h day–night cycle. They were sacrificed 4 weeks after stent placement via administration of inhalable pure carbon dioxide.

2.4. Stent Placement

Anesthesia was induced via intramuscular injection of 50 mg/kg zolazepam and tiletamine (Zoletil 50l, Virbac, Carros, France) and 10 mg/kg xylazine (Rompun, Bayer Healthcare, Leverkusen, Germany). Under fluoroscopic guidance, a 30 cm micro guidewire (Boston Scientific/Medi-Tech, Watertown, MA, USA) was inserted through a custom-made mouthpiece and advanced into the stomach (Figure 1a). The stent delivery system loaded with the I-SEMS or K-SEMS was advanced over the guidewire into the middle portion of the esophagus (Figure 1b). The stent was released in the cervical esophagus below the clavicle level by withdrawing the sheath while the pusher catheter stayed in place. The sheath and the pusher catheter were removed after the stent was completely released (Figure 1c). Esophagography was performed immediately after the procedure to confirm stent patency (Figure 1d).

2.5. Esophagography

Follow-up esophagography was performed using a contrast medium (Telebrix 30 meglumine, Guerbet, Aulnay-sous-Bois, France) immediately before all rats were sacrificed. The stented esophagus was divided into proximal and distal segments. The luminal diameter of each segment was measured on the esophagographic images using RadiAnt DICOM viewer (version 1.1.20, Medixant Company, Poznan, Poland).

2.6. Histologic Examination

The esophagus and stomach were extracted surgically. The esophageal tissue samples were fixed in 10% neutral buffered formalin for 24 h. The samples were dehydrated sequentially with alcohols of different concentrations and embedded in a resin block via infiltration with glycol methacrylate (Technovit 7200® VLC; Heraus Kulzer GMBH, Wertheim, Germany). The embedded samples were axially sectioned at the stented esophagus. Thereafter, the blocks obtained were mounted on an acrylic slide. Microgrinding and polishing with silicon carbide papers using a grinding system (Apparatebau GMBH, Hamburg, Germany) were performed for all slides until a thickness of 30 µm was reached. The slides were stained with hematoxylin and eosin (H&E). Histologic evaluation via H&E staining was based on the percentage of the tissue hyperplasia area, degree of inflammatory cell infiltration, thickness of the submucosal fibrosis, and thickness of the epithelial layers. The tissue hyperplasia formation-related percentage of the esophageal cross-sectional area stenosis was calculated using the following equation:
The   percentage   of   tissue   hyperplasia   area = 100 × ( Stenotic   stented   area   ( mm 2 ) original   stented   area   ( mm 2 ) )
The degree of inflammatory cell infiltration was subjectively determined according to the distribution and density of the inflammatory cells (graded as 1, mild; 2, mild to moderate; 3, moderate; 4, moderate to severe; and 5, severe) [16,17]. The average values of the submucosal fibrosis and epithelial layer thickness and the degree of inflammatory cell infiltration were calculated from eight points around the circumference. Histologic analysis of the esophagus was performed using a digital slide scanner (Pannoramic 250 FLASH III, 3D HISTECH Ltd., Budapest, Hungary). Measurements were obtained using a digital microscope viewer (CaseViewer, 3D HISTECH Ltd.).

2.7. Statistical Analysis

The Mann–Whitney U test was used to analyze the differences between the groups as appropriate. A p-value of <0.05 was considered statistically significant. A Bonferroni-corrected Mann–Whitney U test was further performed for p-values of <0.05 to detect group differences (p < 0.008 indicated statistical significance). Statistical analyses were performed using the SPSS software (version 27.0; SPSS, IBM, Chicago, IL, USA).

3. Results

3.1. Cell Size and Bending Test Findings

The mean ± standard deviation value of the cell size of the I-SEMS was 0.251 ± 0.02 mm2, whereas that of the K-SEMS was 3.712 ± 0.37 mm2 (Figure 2a,c). The cell size of the I-SEMS was 14.7 times smaller than that of the K-SEMS. In the bending test, the I-SEMS showed highly flexible bending without kinking and maintained an inner lumen of the stent (Figure 2b). However, the K-SEMS was not able to withstand the bending force and showed severe kinking in the middle portion, which means an inner lumen of the stent could not be maintained (Figure 2d).

3.2. Procedural Outcomes

Stent placement was technically successful in all rats, and all of them survived until the end of the study. A small amount of hematemesis occurred immediately after the procedure in three rats (one in the I-SEMS group and two in the K-SEMS group), but subsided spontaneously. Stent-related complications, such as stent migration and in-stent stenosis, were not observed during follow-up.

3.3. Esophagographic Findings

The esophagographic findings are shown in Figure 3. The mean overall luminal diameter of the stented esophagus was significantly higher in the I-SEMS group (3.81 ± 0.63 mm) than in the K-SEMS group (2.08 ± 0.86 mm, p < 0.001). This significant difference was also observed in the proximal portion (K-SEMS group: 2.40 ± 0.92 mm vs. I-SEMS group: 4.29 ± 0.51 mm, p < 0.001) and distal portion (K-SEMS group: 1.76 ± 0.77 mm vs. I-SEMS group: 3.35 ± 0.35 mm, p < 0.001). Moreover, severe narrowing was seen around the stent in the K-SEMS group, whereas a straight-line filling defect was observed in the stented esophagus without narrowing in the I-SEMS group (Figure 3a,b).

3.4. Histological Findings

The histologic findings are shown in Figure 4. The mean percentage of the tissue hyperplasia area was significantly higher in the K-SEMS group (48.07% ± 7.30%) than in the I-SEMS group (26.68% ± 8.24%, p < 0.001). The submucosal fibrosis was significantly thicker in the K-SEMS group (0.65 ± 0.13 mm) than in the I-SEMS group (0.25 ± 0.06 mm, p < 0.001). However, the mean thickness of the epithelial layer (0.18 ± 0.07 mm in the K-SEMS group vs. 0.22 ± 0.08 mm in the I-SEMS group, p = 0.290) and the degree of inflammatory cell infiltration (3.00 ± 0.76 in the K-SEMS group vs. 2.75 ± 0.71 in the I-SEMS group, p = 0.506) were not significantly different between the two groups.

4. Discussion

I-SEMSs were developed and designed to increase flexibility and fracture resistance [12,13,14]. They have a uniform and small cell size resulting from fabrication by braiding machines using multiple nitinol wires [13]. I-SEMSs have a significantly smaller cell size than conventional K-SEMSs. Stent-induced tissue hyperplasia commonly and definitely grows through the wire mesh within 4 weeks after stent placement. According to the results of previous studies, stent-induced tissue hyperplasia occurred after stent placement in the rat within 4 weeks, so this study also processed a follow-up for 4 weeks [17,18,19,20]. Our results demonstrated that I-SEMS placement successfully inhibited stent-induced tissue hyperplasia caused by mechanical injury after stent placement in a rat esophageal model. Tissue penetration through the wire meshes may have been relatively preventable due to the small cell size. The high flexibility also relatively reduced the mechanical damages to the esophageal wall. After stent placement, the luminal diameter of the stented esophagus was significantly larger in the I-SEMS group than in the K-SEMS group. Consistently, the histologic results also demonstrated that the tissue hyperplasia-related variables in the K-SEMS group were significantly higher than those in the I-SEMS group. These findings support that the smaller cell size and high flexibility of I-SEMSs are key factors for inhibiting stent-related tissue hyperplasia formation.
The physical properties of I-SEMSs yielded high flexibility to maintain a round shape when they are bent. The flexibility of these stents is associated with stent-related complications, such as stent collapse, fracture, abutment, and kinking [20,21,22,23,24]. In our study, the small cell size of the I-SEMSs increased the stent flexibility. In the bending test, the K-SEMSs showed severe kinking at the middle portion owing to their low flexibility. Conversely, the I-SEMSs successfully maintained an inner lumen without any stent deposition. With their superior physical advantages, I-SEMSs may prevent various stent-related complications in non-vascular luminal organs, including the esophagus, biliopancreatic duct, colon, and airway. Furthermore, they seem to be particularly useful in tortuous anatomical structures of non-vascular luminal organs.
I-SEMSs have been extensively used in managing vascular stenosis [11,12,25]. Clinical investigators reported excellent outcomes of I-SEMS placement for primary superficial femoral artery lesions to prolong stent patency and reduce stent fracture [26,27,28,29,30]. However, I-SEMSs have high variations of stent length caused by shortening and elongation during stent placement [12]. In our study, wherein I-SEMSs were applied for the first time in the rat esophagus, stent shortening and elongation during stent placement did not occur because the esophageal stent delivery system has a better profile than the vascular stent delivery system.
Our study has several limitations. First, tissue responses were observed in the normal rat esophagus after stent placement. Stent-induced hyperplasia formation after stent placement may differ with esophageal strictures. Second, only a few tests of the stent samples were conducted in this study. Although the bending test was successfully performed to compare the degree of stent deformation, the bending stiffness could not be investigated because of the lack of a measuring device. Additional mechanical properties of the newly developed I-SEMS for non-vascular luminal organs, including radial force, axial tension, and torsion test findings, should be evaluated. Although the tortuous anatomical structures of non-vascular luminal organs have not been investigated to prove the benefits of the high flexibility of I-SEMSs, our results demonstrated the basic concept of the effectiveness and safety of these stents in the rat esophagus.

5. Conclusions

Formation of stent-induced tissue hyperplasia was evident in both the I-SEMS and K-SEMS groups. Placement of I-SEMSs with a small cell size and high flexibility relatively suppressed stent-induced tissue hyperplasia compared with placement of K-SEMSs in the rat esophageal model. Placement of I-SEMSs with excellent flexibility seems to be applicable to highly tortuous non-vascular luminal organs for palliative treatment of malignant obstructions. Although further preclinical studies are required to investigate its efficacy and safety, I-SEMS placement can be a palliative therapeutic option for malignant stricture diseases of non-vascular organs.

Author Contributions

Conceptualization, J.-H.P. and S.S.L.; methodology, J.M.K. and D.O.; software, S.H.K.; validation, J.M.K., D.O. and D.S.R.; formal analysis, Y.P.; investigation, D.-S.W. and J.W.K.; resources, C.H.Z.; data curation, J.-H.P. and S.S.L.; writing—original draft preparation, J.M.K. and D.O.; writing—review and editing, C.H.Z., J.-H.P. and S.S.L.; visualization, J.M.K.; supervision, J.-H.P.; project administration, S.S.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a grant (2021IE0017) from the Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea.

Institutional Review Board Statement

This study was approved by the Institutional Animal Care and Use Committee of the Asan Institute for Life Sciences (2021-14-231) and conformed to the US National Institutes of Health guidelines for humane handling of laboratory animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to ethical issues.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Knyrim, K.; Wagner, H.J.; Bethge, N.; Keymling, M.; Vakil, N. A controlled trial of an expansile metal stent for palliation of esophageal obstruction due to inoperable cancer. N. Engl. J. Med. 1993, 329, 1302–1307. [Google Scholar] [CrossRef] [PubMed]
  2. Ross, W.A.; Alkassab, F.; Lynch, P.M.; Ayers, G.D.; Ajani, J.; Lee, J.H.; Bismar, M. Evolving role of self-expanding metal stents in the treatment of malignant dysphagia and fistulas. Gastrointest. Endosc. 2007, 65, 70–76. [Google Scholar] [CrossRef]
  3. Sharma, P.; Kozarek, R. Role of esophageal stents in benign and malignant diseases. Am. J. Gastroenterol. 2010, 105, 258–273; quiz 274. [Google Scholar] [CrossRef]
  4. Park, J.H.; Song, H.Y.; Park, J.Y.; Kim, J.H.; Kim, Y.H.; Kim, J.H.; Kim, S.B. Temporary stent placement with concurrent chemoradiation therapy in patients with unresectable oesophageal carcinoma: Is there an optimal time for stent removal? Eur. Radiol. 2013, 23, 1940–1945. [Google Scholar] [CrossRef]
  5. Kim, J.H.; Song, H.Y.; Choi, E.K.; Kim, K.R.; Shin, J.H.; Lim, J.O. Temporary metallic stent placement in the treatment of refractory benign esophageal strictures: Results and factors associated with outcome in 55 patients. Eur. Radiol. 2009, 19, 384–390. [Google Scholar] [CrossRef]
  6. Park, J.H.; Song, H.Y.; Kim, J.H.; Jung, H.Y.; Kim, J.H.; Kim, S.B.; Lee, H. Polytetrafluoroethylene-covered retrievable expandable nitinol stents for malignant esophageal obstructions: Factors influencing the outcome of 270 patients. AJR Am. J. Roentgenol. 2012, 199, 1380–1386. [Google Scholar] [CrossRef]
  7. Schaer, J.; Katon, R.M.; Ivancev, K.; Uchida, B.; Rösch, J.; Binmoeller, K. Treatment of malignant esophageal obstruction with silicone-coated metallic self-expanding stents. Gastrointest. Endosc. 1992, 38, 7–11. [Google Scholar] [CrossRef]
  8. Shin, J.H.; Song, H.Y.; Ko, G.Y.; Lim, J.O.; Yoon, H.K.; Sung, K.B. Esophagorespiratory fistula: Long-term results of palliative treatment with covered expandable metallic stents in 61 patients. Radiology 2004, 232, 252–259. [Google Scholar] [CrossRef]
  9. Song, H.Y.; Lee, D.H.; Seo, T.S.; Kim, S.B.; Jung, H.Y.; Kim, J.H.; Park, S.I. Retrievable covered nitinol stents: Experiences in 108 patients with malignant esophageal strictures. J. Vasc. Interv. Radiol. 2002, 13, 285–293. [Google Scholar] [CrossRef]
  10. Kim, K.R.; Shin, J.H.; Song, H.Y.; Ko, G.Y.; Kim, J.H.; Yoon, H.K.; Sung, K.B. Palliative treatment of malignant esophagopulmonary fistulas with covered expandable metallic stents. AJR Am. J. Roentgenol. 2009, 193, W278–W282. [Google Scholar] [CrossRef]
  11. Garcia, L.A.; Rosenfield, K.R.; Metzger, C.D.; Zidar, F.; Pershad, A.; Popma, J.J.; Zaugg, M.; Jaff, M.R. SUPERB final 3-year outcomes using interwoven nitinol biomimetic supera stent. Catheter. Cardiovasc. Interv. 2017, 89, 1259–1267. [Google Scholar] [CrossRef]
  12. Garcia, L.; Jaff, M.R.; Metzger, C.; Sedillo, G.; Pershad, A.; Zidar, F.; Patlola, R.; Wilkins, R.G.; Espinoza, A.; Iskander, A.; et al. Wire-Interwoven Nitinol Stent Outcome in the Superficial Femoral and Proximal Popliteal Arteries: Twelve-Month Results of the SUPERB Trial. Circ. Cardiovasc. Interv. 2015, 8, e000937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wholey, M.H.; Finol, E.A. Designing the ideal stent. Endovasc. Today 2007, 6, 25–34. [Google Scholar]
  14. Maleckis, K.; Anttila, E.; Aylward, P.; Poulson, W.; Desyatova, A.; MacTaggart, J.; Kamenskiy, A. Nitinol Stents in the Femoropopliteal Artery: A Mechanical Perspective on Material, Design, and Performance. Ann. Biomed. Eng. 2018, 46, 684–704. [Google Scholar] [CrossRef] [PubMed]
  15. Armstrong, E.J.; Jeon-Slaughter, H.; Kahlon, R.S.; Niazi, K.A.; Shammas, N.W.; Banerjee, S. Comparative Outcomes of Supera Interwoven Nitinol vs Bare Nitinol Stents for the Treatment of Femoropopliteal Disease: Insights From the XLPAD Registry. J. Endovasc. Ther. 2020, 27, 60–65. [Google Scholar] [CrossRef]
  16. Park, J.H.; Kim, J.H.; Kim, E.Y.; Kim, J.; Song, H.Y.; Kim, W.J.; Lee, D.; Park, J.; Kim, S. Bioreducible polymer-delivered siRNA targeting MMP-9: Suppression of granulation tissue formation after bare metallic stent placement in a rat urethral model. Radiology 2014, 271, 87–95. [Google Scholar] [CrossRef] [Green Version]
  17. Kim, J.H.; Song, H.Y.; Park, J.H.; Yoon, H.J.; Park, H.G.; Kim, D.K. IN-1233, an ALK-5 inhibitor: Prevention of granulation tissue formation after bare metallic stent placement in a rat urethral model. Radiology 2010, 255, 75–82. [Google Scholar] [CrossRef]
  18. Park, J.H.; Park, W.; Cho, S.; Kim, K.Y.; Tsauo, J.; Yoon, S.H.; Son, W.C.; Kim, D.H.; Song, H.Y. Nanofunctionalized Stent-Mediated Local Heat Treatment for the Suppression of Stent-Induced Tissue Hyperplasia. ACS Appl. Mater. Interfaces 2018, 10, 29357–29366. [Google Scholar] [CrossRef] [PubMed]
  19. Park, J.H.; Kim, T.H.; Cho, Y.C.; Bakheet, N.; Lee, S.O.; Kim, S.H.; Kim, K.Y. Balloon-Expandable Biodegradable Stents Versus Self-Expandable Metallic Stents: A Comparison Study of Stent-Induced Tissue Hyperplasia in the Rat Urethra. Cardiovasc. Interv. Radiol. 2019, 42, 1343–1351. [Google Scholar] [CrossRef]
  20. Li, H.; Rha, S.W.; Choi, B.G.; Choi, S.Y.; Moon, S.K.; Jang, W.Y.; Kim, W.; Ahn, J.H.; Park, S.H.; Choi, W.G.; et al. Impact of chronic outward force on arterial responses of proximal and distal of long superficial femoral artery stent. BMC Cardiovasc. Disord. 2021, 21, 323. [Google Scholar] [CrossRef]
  21. Berry, J.L.; Manoach, E.; Mekkaoui, C.; Rolland, P.H.; Moore, J.E., Jr.; Rachev, A. Hemodynamics and wall mechanics of a compliance matching stent: In vitro and in vivo analysis. J. Vasc. Interv. Radiol. 2002, 13, 97–105. [Google Scholar] [CrossRef]
  22. Rachev, A.; Manoach, E.; Berry, J.; Moore, J.E., Jr. A model of stress-induced geometrical remodeling of vessel segments adjacent to stents and artery/graft anastomoses. J. Theor. Biol. 2000, 206, 429–443. [Google Scholar] [CrossRef] [PubMed]
  23. Krankenberg, H.; Schlüter, M.; Steinkamp, H.J.; Bürgelin, K.; Scheinert, D.; Schulte, K.L.; Minar, E.; Peeters, P.; Bosiers, M.; Tepe, G.; et al. Nitinol stent implantation versus percutaneous transluminal angioplasty in superficial femoral artery lesions up to 10 cm in length: The femoral artery stenting trial (FAST). Circulation 2007, 116, 285–292. [Google Scholar] [CrossRef] [Green Version]
  24. Ormiston, J.A.; Webber, B.; Ubod, B.; White, J.; Webster, M.W. Coronary stent durability and fracture: An independent bench comparison of six contemporary designs using a repetitive bend test. EuroIntervention 2015, 10, 1449–1455. [Google Scholar] [CrossRef] [PubMed]
  25. Rocha-Singh, K.J.; Jaff, M.R.; Crabtree, T.R.; Bloch, D.A.; Ansel, G. Performance goals and endpoint assessments for clinical trials of femoropopliteal bare nitinol stents in patients with symptomatic peripheral arterial disease. Catheter. Cardiovasc. Interv. 2007, 69, 910–919. [Google Scholar] [CrossRef]
  26. Nakama, T.; Obunai, K.; Muraishi, M.; Kojima, S.; Watanabe, H. Invagination of an Interwoven Nitinol Stent During Femoropopliteal Placement. JACC Cardiovasc. Interv. 2020, 13, e71–e72. [Google Scholar] [CrossRef]
  27. Werner, M.; Paetzold, A.; Banning-Eichenseer, U.; Scheinert, S.; Piorkowski, M.; Ulrich, M.; Bausback, Y.; Bräunlich, S.; Schmidt, A.; Scheinert, D. Treatment of complex atherosclerotic femoropopliteal artery disease with a self-expanding interwoven nitinol stent: Midterm results from the Leipzig SUPERA 500 registry. EuroIntervention 2014, 10, 861–868. [Google Scholar] [CrossRef]
  28. Nakama, T.; Obunai, K.; Muraishi, M.; Kojima, S.; Watanabe, H. Angioscopic Findings on 15-Month Follow-Up for Interwoven Nitinol Stent Invagination in the Femoropopliteal Artery. JACC Cardiovasc. Interv. 2020, 13, 1958–1959. [Google Scholar] [CrossRef]
  29. Dumantepe, M.; Seren, M.; Fazlıogullari, O.; Ayoglu, U.; Teymen, B. Treatment of complex atherosclerotic femoropopliteal artery disease with a self-expanding interwoven nitinol stent: Midterm results. Vascular 2021, 29, 711–719. [Google Scholar] [CrossRef] [PubMed]
  30. George, J.C.; Rosen, E.S.; Nachtigall, J.; VanHise, A.; Kovach, R. SUPERA interwoven nitinol Stent Outcomes in Above-Knee IntErventions (SAKE) study. J. Vasc. Interv. Radiol. 2014, 25, 954–961. [Google Scholar] [CrossRef]
Figure 1. Radiographic images obtained during I-SEMS placement showing the technical steps of stent placement. (a) Radiographic image showing a guidewire with a catheter. (b) A stent delivery system (arrows) inserted into the rat esophagus. (c) Radiographic image showing the placed stent (arrows) in the esophagus. (d) Esophagographic image showing the patency of the I-SEMS immediately after placement. I-SEMS, interwoven self-expandable metallic stent.
Figure 1. Radiographic images obtained during I-SEMS placement showing the technical steps of stent placement. (a) Radiographic image showing a guidewire with a catheter. (b) A stent delivery system (arrows) inserted into the rat esophagus. (c) Radiographic image showing the placed stent (arrows) in the esophagus. (d) Esophagographic image showing the patency of the I-SEMS immediately after placement. I-SEMS, interwoven self-expandable metallic stent.
Applsci 12 00346 g001
Figure 2. Interwoven and knitted SEMSs. Images showing (a) the interwoven SEMS with a mean cell size of 0.251 mm2 and (b) its bending test finding using the phantom. (c) Images showing the knitted SEMS with a mean cell size of 3.712 mm2. (d) the bending test was performed to evaluate the degrees of stent deformation, such as bending and kinking, of the two SEMSs. SEMS, self-expandable metallic stent.
Figure 2. Interwoven and knitted SEMSs. Images showing (a) the interwoven SEMS with a mean cell size of 0.251 mm2 and (b) its bending test finding using the phantom. (c) Images showing the knitted SEMS with a mean cell size of 3.712 mm2. (d) the bending test was performed to evaluate the degrees of stent deformation, such as bending and kinking, of the two SEMSs. SEMS, self-expandable metallic stent.
Applsci 12 00346 g002
Figure 3. Esophagographic findings at 4 weeks after stent placement. Esophagographic images showing (a) severe luminal narrowing (arrows) in the K-SEMS group and (b) straight-line filling defect (arrowheads) in the I-SEMS group. (c) Graph showing the mean luminal diameter of the stented esophagus at 4 weeks after stent placement in both groups. K-SEMS, knitted self-expandable metallic stent; I-SEMS, interwoven self-expandable metallic stent; CI, confidence interval. * p < 0.001.
Figure 3. Esophagographic findings at 4 weeks after stent placement. Esophagographic images showing (a) severe luminal narrowing (arrows) in the K-SEMS group and (b) straight-line filling defect (arrowheads) in the I-SEMS group. (c) Graph showing the mean luminal diameter of the stented esophagus at 4 weeks after stent placement in both groups. K-SEMS, knitted self-expandable metallic stent; I-SEMS, interwoven self-expandable metallic stent; CI, confidence interval. * p < 0.001.
Applsci 12 00346 g003
Figure 4. Representative microscopic images of the histologic sections. Formation of stent-induced tissue hyperplasia through the wire filaments (arrowheads) was evident in both the K-SEMS and I-SEMS groups. The degree of stent-induced tissue hyperplasia in (a) the K-SEMS group was significantly higher than that in (b) the I-SEMS group. (c,d) The submucosal fibrosis (arrow) in the K-SEMS group was significantly thicker than that in the I-SEMS group. The thickness of the epithelial layer (black arrows) was not significantly different between the two groups. (e) Histologic results of the stented rat esophagus at 4 weeks after stent placement in the K-SEMS and I-SEMS groups. K-SEMS, knitted self-expandable metallic stent; I-SEMS, interwoven self-expandable metallic stent; CI, confidence interval.
Figure 4. Representative microscopic images of the histologic sections. Formation of stent-induced tissue hyperplasia through the wire filaments (arrowheads) was evident in both the K-SEMS and I-SEMS groups. The degree of stent-induced tissue hyperplasia in (a) the K-SEMS group was significantly higher than that in (b) the I-SEMS group. (c,d) The submucosal fibrosis (arrow) in the K-SEMS group was significantly thicker than that in the I-SEMS group. The thickness of the epithelial layer (black arrows) was not significantly different between the two groups. (e) Histologic results of the stented rat esophagus at 4 weeks after stent placement in the K-SEMS and I-SEMS groups. K-SEMS, knitted self-expandable metallic stent; I-SEMS, interwoven self-expandable metallic stent; CI, confidence interval.
Applsci 12 00346 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kang, J.M.; Oh, D.; Kim, S.H.; Ryu, D.S.; Park, Y.; Won, D.-S.; Kim, J.W.; Zeng, C.H.; Park, J.-H.; Lee, S.S. Interwoven versus Knitted Self-Expandable Metallic Stents: A Comparison Study of Stent-Induced Tissue Hyperplasia in the Rat Esophagus. Appl. Sci. 2022, 12, 346. https://doi.org/10.3390/app12010346

AMA Style

Kang JM, Oh D, Kim SH, Ryu DS, Park Y, Won D-S, Kim JW, Zeng CH, Park J-H, Lee SS. Interwoven versus Knitted Self-Expandable Metallic Stents: A Comparison Study of Stent-Induced Tissue Hyperplasia in the Rat Esophagus. Applied Sciences. 2022; 12(1):346. https://doi.org/10.3390/app12010346

Chicago/Turabian Style

Kang, Jeon Min, Dongwook Oh, Song Hee Kim, Dae Sung Ryu, Yubeen Park, Dong-Sung Won, Ji Won Kim, Chu Hui Zeng, Jung-Hoon Park, and Sang Soo Lee. 2022. "Interwoven versus Knitted Self-Expandable Metallic Stents: A Comparison Study of Stent-Induced Tissue Hyperplasia in the Rat Esophagus" Applied Sciences 12, no. 1: 346. https://doi.org/10.3390/app12010346

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop