Iranian Journal of Medical Sciences

Document Type : Review Article

Authors

1 Department of Tissue Engineering, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran

2 Department of Pediatrics Gastroenterology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran

3 Department of Surgery, Shiraz Laparoscopic Research Center, Shiraz University of Medical Sciences, Shiraz, Iran

4 Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran

Abstract

The esophagus is the gastrointestinal tract’s primary organ that transfers bolus into the stomach with peristaltic motion. Therefore, its lesions cause a significant disturbance in the nutrition and digestive system. Esophageal disease treatment sometimes requires surgical procedures that involve removal and circumferential full-thickness replacement. Unlike other organs, the esophagus has a limited regeneration ability and cannot be transplanted from donors. There are various methods of restoring the esophageal continuity; however, they are associated with certain flaws that lead to a non-functional recovery. As an exponentially growing science, tissue engineering has become a leading technique for the development of tissue replacement to repair damaged esophageal segments. Scaffold plays a significant role in the process of tissue engineering, as it acts as a template for the regeneration of growing tissue. A variety of scaffolds have been studied to replace the esophagus. Due to the many tissue quality challenges, the results are still inadequate and need to be improved. The success of esophageal tissue regeneration will finally depend on the scaffold’s capability to mimic natural tissue properties and provide a qualified environment for regeneration. Thereby, scaffold fabrication techniques are fundamental. This article reviews the recent developments in esophageal tissue engineering for the treatment of circumferential lesions based on scaffold biomaterial engineering approaches.

Keywords

What’s Known

Several works have been done on esophageal tissue engineering, but a comprehensive review of recent works that focuses on full-thickness replacement has been missing.

What’s New

This article is a comprehensive review of the work done for natural and synthetic materials used in esophageal tissue engineering in recent years, which has not been presented so far. The present article reviewed the new works done in esophageal tissue engineering with a focus on full-thickness replacements.

Introduction

The esophagus, or gullet, is a muscular tube in the vertebrate body that connects the pharynx to the stomach. Transferring bolus into the stomach is the dominant role of the esophagus in the digestive system. The esophagus in humans can be divided into three regions: cervical, thoracic, and abdominal. The cervical muscle belongs to the category of skeletal muscles, while the thoracic muscle is a smooth muscle. 1 Similar to the other gastrointestinal tract components, the esophagus wall consists of four layers, namely the mucosa, submucosa, muscularis propia, and adventitia layers. Blood vessels, nervous system fibers, and esophageal glands are mostly located on the submucosa layer. The mucosa layer covers the inner wall of the esophagus. The mucosa layer cells in the proximal region are squamous epithelial cells, and in the distal region where the esophagus attaches to the stomach, they are columnar epithelial cells. 2 Accordingly, it can be said that the esophagus is one of the primary organs of the gastrointestinal tract, and its lesions cause a significant disturbance in the nutrition and digestive system. Although the esophageal function is the transfer of food by peristaltic movements, its ailment causes serious gastrointestinal problems and decreases the quality of life.

Esophageal diseases are increasing yearly. Hence, it is necessary to devise a comprehensive plan and find suitable treatments. Accordingly, considering the rapid advancement of science and the integration of various disciplines aiming to advance modern medical treatments and achieve high levels of tissue engineering technology, appropriate therapeutic solutions can be proposed, as they have been for other organ lesions. To create an adequately engineered tissue with the natural human esophagus’ properties, in addition to choosing the right scaffolding method, it is also necessary to use proper polymer structures and materials that have appropriate characteristics, such as mechanical strength, biocompatibility, biodegradability, and elasticity. Besides, the selection of appropriate biological factors and components, such as growth factors, cytokines, differentiated cells, or stem cells, is also required to obtain a suitable engineered tissue that stimulates and promotes regeneration in the human body. 2

This review aims to discuss the advancements in tissue-engineered esophageal fabrications. Furthermore, this study evaluates various techniques utilized to advance proper tissue replacement, and the challenges we face in clinical practice. The esophageal tissue engineering process is briefly illustrated in figure 1.

Figure 1. The diagram shows the esophagus tissue engineering trend

Anatomy and Histology

The esophagus is a muscular tube-shaped organ with a 20-25 cm length and an approximately 2 cm diameter. It is divided into three main parts: cervical, thoracic, and abdominal. 3

The esophageal tube starts within the 6th cervical vertebrae and ends at the junction with the stomach at the 11th thoracic vertebrae level. The cervical segment is about 5 cm, which continues unto a 17-18 cm thoracic part. After crossing the diaphragm, it reaches the abdominal part, which is about 2-3 cm in length. 4

The length of the esophagus depends on age, sex, and physical characteristics. For instance, in newborns, this organ’s proximal and distal ends are typically one or two vertebrae higher than in adults, and it extends to lower vertebrates by age. 5 Mucosa, sub-mucosa, muscular externa and adventitia are the main layers of this organ.

The mucosa is a non-keratinized stratified squamous epithelium, which coats the inner surface of the esophageal wall. Lamina propria and lamina muscularis mucosa are the next subsections of mucosa beneath the epithelium. Loose connective tissue is the major component of lamina propria. Smooth muscle tissue and elastic fibers make the muscular subsection of the mucosa. The “Z line” is the point, where the non-keratinized stratified squamous epithelium of the mucosa attaches to the simple columnar epithelium in the cardia of the stomach. 5

The sub-mucosa is an elastic and collagenous layer formed mainly by dense, irregular connective tissue, containing veins, lymphatics, and Meissner plexus. 5 The muscularis propria layer consists of longitudinal (outer part) and circular (inner) muscle fibers. The longitudinal fibers originate from the posterior portion of the cricoid cartilage and develop a triangle called the “Lamier triangle”, surrounded by the longitudinal muscle fibers laterally and the cricopharyngeal muscle superiorly. The Killian triangle is another anatomic hallmark that is located in this region. Inferior constrictor muscles of the pharynx and the cricopharyngeus muscle form the limiting borders of this triangle. Longitudinal muscle fibers are collected laterally in the upper portion of the esophagus, but these fibers expand and cover all surfaces at the lower sides and make a more potent layer in the lower third part of the esophagus. Circular muscle fibers are a thinner and medial layer relative to longitudinal fibers. These fibers are not circular within different esophagus segments, but they become circular at the lower parts. Despite their irregular formation, their regular pattern makes the entire muscle layer act as a shutter-like apparatus. Sometimes, following a sudden increase in luminal pressure, spontaneous perforation occurs in the lowermost parts, including the entire esophageal wall. Striated muscles are the dominant upper esophageal muscular layer, the lower parts, consisting of mainly smooth muscle fibers. Changing the muscular layers of different segments is associated with a transition zone, including striated and smooth muscle fibers. 5

Adventitia

The adventitia is a loose connective tissue surrounding most of the esophagus and is the outer layer of the organ. This loose layer facilitates the spread of infections and the creation of tumors in the esophagus. 5

Esophageal Arteries

The blood supply of the esophagus is mainly provided by the inferior thyroid artery, aorta, and left gastric artery, which supplies the cervical, thoracic, and abdominal segments, respectively. Since esophageal arterial blood flow is rich enough for anastomosis, any unwanted dissection can cause life-threatening bleeding in the esophagus. 6 , 7

Esophageal Veins

The venous system starts to develop from the submucosa layer, and after passing the outer layers, drains into the inferior thyroid, azygos, and left gastric vein along with cervical, thoracic, and abdominal segments, respectively. The esophageal venous plexus also acts as a venous outflow for the drainage of other veins, including short gastric veins, splenic vein, left gastroepiploic vein, and branches of an inferior phrenic vein. 6 , 8

Sphincters

The upper esophageal sphincter (UES) and the lower esophageal sphincter (LES) are the two main proximal and distal boundaries of the esophagus. 9 The function of these two sphincters is to close the entries of the esophagus. However, they do not have the exact anatomical features of other sphincters. 10 Although UES is a striated muscle, it is not under voluntary control, and the opening is resulted from a swallowing signal. 11

The LES, also called the gastroesophageal sphincter, controls the lower part of the esophagus at the gastroesophageal junction. 12 Some of its functions are presented here.

Swallowing Using Peristaltic Motion

After swallowing, food and fluids pass through the pharynx and esophagus, one of the first parts of the alimentary tract. 10 The swallowing process begins with a backward movement of the epiglottitis and simultaneous UES relaxation, leading to the food and fluids passing the bolus through the esophagus. The downward movement of food results from a rhythmic and regular contraction and relaxation of the different parts of the esophagus. 10

Prohibiting Gastric Acid Reflux

Gastric acid is the main secreting component of the stomach, consisting of hydrochloric acid (HCl) and potassium and sodium salts, which correlate with food ingestion. The normal pressure of LES prevents the backflow of the gastric ingredients, protecting the esophageal mucosa. The lower crura of the diaphragm is another compensatory component that helps decrease the reflux of acid and the acute angle of His. 10

Prevalent Esophageal Diseases and Current Treatments

Esophageal lesions can be divided into two categories: congenital lesions and lesions occurring throughout life due to various factors. In recent years, around 500,000 people globally have gotten malignant esophageal cancer each year. Although the rate of human malignant cancers of other organs, in most cases, has either been kept fixed or decreased, esophageal cancer has increased by about 140% in the last 10 years. 13 , 14

Even though esophageal cancer is more common in adults, other diseases, such as esophageal atresia, are seen in infants and newborns. Depending on the geographic region, in every 2500 to 5000 newborns, one infant gets esophageal atresia. 15 , 16 Because of this congenital defect, the newborn’s esophagus is closed, and he/she will be unable to swallow and digest fluids and milk; therefore, surgery is required. One of the most common types of atresia is the tracheoesophageal fistula (TOF), in which a part of the esophagus is connected to the trachea. 17 In this type of disease, not only the ability to drink milk is disturbed, but also breathing becomes difficult. Therefore, on the first day following birth, an initial surgery is required to split the trachea from the esophagus in order to reduce breathing problems. The life condition will be stabilized, but the problem of esophageal atresia will persist. 18 , 19 Several methods have been suggested and performed to treat esophageal atresia, but a thoroughly efficient method has not been developed yet. The first reports of successful surgeries are from the 1940s. Since then, various surgical techniques have been developed and optimized, 17 leading to an increase in the success of these surgeries by a rate of over 95%. 20 Despite these improvements, neonates have a lower quality of life after surgery. Several factors may be involved, such as anastomosis leakage, stenosis, gastric reflux, dysfunction, and inadequate esophageal motility. Hence, these factors cause frequent referrals to the hospital, and various surgical and non-surgical treatments are needed. Among different esophageal atresia types, long-term esophageal atresia (LGOA) is the most complicated, and consequently, the most difficult to treat. Therefore, esophageal connectivity is not achieved with one surgery alone and requires multiple steps. One of the usual strategies for these patients is to have a gastrostomy performed on their stomach on the first day following birth, helping them stay alive and grow. During the treatment, which can take weeks up to months, patients cannot stay at home and require hospitalization. They should also be admitted to the intensive care unit (ICU), since multiple discharges would be accumulated in the upper esophagus, causing the risk of respiratory complications. Therefore, these substances should be routinely exhaled through a suction tube. When the normal growth of the esophagus is not sufficient, a technique is suggested to stretch the two sides of the esophagus, reducing the distance, and then, connecting them with surgery. This is one of the newest methods currently being implemented. 17

Other methods, such as gastric transposition or colon replacement, have been suggested and are currently performed by surgeons. 21 , 22 Esophagectomy is the primary part of these different procedures, which tries to maintain the uniformity of the esophagus 23 by replacing the missed portion with plastic 24 and synthetic constructs. 25 , 26 One of the approved treatment options is known as an esophageal replacement with gastric tube. 27 - 33 However, utilizing other alternatives, including aortic autografts 34 and prosthetic constructs 26 , 35 , 36 has been a standard method of the last decades. The promotion of surgical techniques and instruments such as stents 37 - 40 has allowed for the progress of valuable methods for restoring esophageal continuity and functionality. These procedures include the omental wrapping of the esophagus, 41 - 43 gastric pull-up, 44 - 48 colonic interpositions, 49 - 51 and deltopectoral, 52 - 54 and pectoralis major 55 - 58 myocutaneous flaps. Nevertheless, all of these various practices are still associated with considerable complications and mortality. The main problem with conventional esophagectomy treatments is the appropriate replacement of esophageal lesions. 59 , 60

It is currently impossible to build a human esophagus based on the existing knowledge. Spontaneous reconstruction and repair of the esophagus following injury do not occur for more than a specified length within the body, and the body cannot regenerate the entirety of esophageal tissues. Moreover, due to the lack of an adequate vascular network in those areas, a repair cannot occur. 61 - 64 Due to the stated therapeutic limitations, there is a strong need for a suitable esophagus substitute. The conduit or construct for the esophagus should have the ability to transfer food and fluids from the mouth to the stomach without leakage, perforation, or rupture, while also having the appropriate mechanical and structural characteristics similar to those of the natural esophagus. The stress and strain of the esophagus wall in the human body undergo up to about 1 MPa and 175% change in pressure and length, respectively, which are significant. 65 , 66 Hence, the esophagus must be able to tolerate this expansion. Despite substantial advances in stem cell therapy and tissue engineering of the skeletal muscle systems and the ability to prepare 3D multi cells culture, there is no significant progress achieved in the field of multilayer tissue engineering of the internal organs of the gastrointestinal tract until now. 67 - 69 Developing an appropriate tissue-engineered product in vitro and implementing the tissue in vivo is the main challenge we face, mostly due to struggles in creating large perfused scaffolds that implement oxygen, nutrients, and waste products’ proper diffusion. These challenges are tough when the organ candidate for regeneration has distinct spatial structural characteristics. 67 A new option for in vitro research is the use of bioreactors to simulate in vivo biological states. Numerous works have been done on acellular matrix materials used in bioreactors to improve esophageal healing. 70 , 71 However, efforts need to be continued to stimulate esophageal regeneration and attachment of the cell population to the scaffold. 72 - 74 The in vivo scope and the use of endogenous signaling stimulation by mesenchymal stem cells (MSCs) have been addressed in further bioreactor works. Pre-clinical trials have been performed on these cells, some of which have reached clinical trials, 75 - 77 and no adverse effect has been reported in the healing stages. Another benefit to using MSCs is their availability; they can be easily obtained from autologous sources. 78 Although clinical data suggest that MSCs are safe to use, their effect on esophageal repair is still unclear. Recent work on esophageal tissue engineering involves the utilization of synthetic materials with a cylindrical structure. 79 - 81 Although these materials provide appropriate mechanical support for the structure, they cannot stimulate regeneration in vivo by themselves. Recent works have focused on combining these materials with biological factors to construct hybrid scaffolds. 82 - 84 Substances of biological origin have a higher ability to simulate natural tissue compositions and properties than synthetic materials, and hence, they have been studied for esophageal repair in several works. 85 , 86 However, their combination with synthetic materials and biological factors has been recently considered due to their benefits. Since about 150 years ago, there have been many ways to replace cancerous esophageal tissues, perhaps the simplest and the most practical one is the use of a rubber tube. 87

Recently, scientists have been using surgical polymers like Dacron and Marlex alone or coupling with silicone to develop synthetic grafts. 80 - 82 The postoperative survival rate in an animal model (canine) was reported to be 44% for one year and 25% for six years. Reconstruction of the mucosa and sub-mucosa layers was observed due to anastomosis of the scaffold with normal esophageal tissue, but no muscle tissue was formed. In another work on pigs, using nitinol and silicone, about 60% of the constriction occurred in the specimens, and it resembled the animal specimen (dog) of the mucosa and sub-mucosa; however, the muscle layer was not formed. 88 These studies are strong evidence that synthetic materials, despite being able to provide the appropriate strength and mechanical properties, are not capable of stimulating and completely reconstructing the organ. In another experience, a poly-glycolic acid-adsorbed polymeric scaffold with an amniotic membrane was used, and a muscle layer was formed during the reconstruction process. 89 It was found that adding specific cells, along with several growth factors, to the polymeric scaffold can greatly enhance the repair and reconstruction. Scaffold design parameters are also important; for example, the effect of different porosities of esophageal scaffolds on cell migration, adhesion, and their proliferation has been studied. 90 Another critical parameter is the scaffold degradation rate in the biological environment of the body. Studies on scaffold degradation rate and tissue regeneration rate have reported that if the scaffold degradation rate exceeds the tissue regeneration rate, the mechanical strength of the structure will be lost, and the structure will collapse. On the other hand, if the degradation rate is below the regeneration rate, blockage builds up, and it is not repaired properly. 91 - 93 As mentioned earlier, recent attempts have been made to utilize biological components with scaffolds; in this area, extracellular matrix (ECM) proteins such as collagen have been used along with scaffold polymers to improve esophageal remodeling. 94 - 96

Collagen and polymeric scaffolds have also been used as composite scaffolds in esophageal epithelial cells. 97 - 99 Another important issue related to the scaffold is the processing method. Various researchers have used electro-spinning to create esophageal scaffolds. Electro-spinning is a new and evolving method, which can create filament structures with nanometer diameters. 100 This is a major advantage to this method, which has thus far produced numerous scaffolds for the esophagus using polymeric materials that can be soluble. However, the major problem is the lack of adequate mechanical strength in the scaffolds produced by electro-spinning. Therefore, other techniques have been proposed to be used or combined with electro-spinning to enhance the scaffold strength. 101 - 103

Clinical Importance of Circumferential Lesions

Various types of esophagus diseases have become known since 200 years ago. 104 This paper tries to focus on the full-thickness breakdown, which engages all layers. Fistulae and leaks are the primary consequence of full-thickness defects. Esophagectomy is a common cause of full-thickness defects, which is performed following malignant and benign conditions. These surgeries require implementing colon or stomach segments to keep the lumen’s uniformity. These interventions can lead to major morbidities, followed by a low quality of life. Due to the limited ability of the esophagus in regenerating its tissues, these injuries can cause refractory strictures, leaks, and fistulae. Attempts at transplanting the cadaveric esophagus have failed. 104 Eventually, regenerating the esophagus would be optimal for these patients.

Moreover, common esophagus diseases, such as adenocarcinoma, squamous cell carcinoma, caustic ingestion, and congenital disorders, can also cause full-thickness defects. Adenocarcinoma and squamous cell carcinoma have engaged about 50,000 and 400,000 people in the world each year, respectively. 105 Esophageal atresia is a common congenital disease, which affects every one in 3000 infants. 106 - 108 About 5000 cases of caustic ingestion have been reported in the US annually. It can cause a severe injury to the esophagus and the stomach and depending on the burn degree, it can affect even all layers of the esophagus. 109

Stenosis

Four points in the esophagus have the potential for stricture. In cases of swallowing a corrosive substance or a solid object, damage to one of the points mentioned in the following is more probable. The compression of the esophagus by surrounding structures causes these constrictions. Acute stricture can also be considered as a full-thickness defect. These constrictions include: 9

  1. ● At the proximal part of the esophagus, where the pharynx connects to the esophagus, behind the cricoid cartilage
  2. ● Where the aortic arch crosses the esophagus in the superior mediastinum
  3. ● Where the left main bronchus compresses the esophagus in the posterior mediastinum
  4. ● The esophageal hiatus where travels across the diaphragm in the posterior mediastinum

Esophageal Cancer

An annual rate of about 400,000 deaths associated with esophageal cancer has been estimated, 110 which makes it the sixth leading cause of death due to cancer worldwide. 111 Esophageal cancer is divided into two main types; I) Squamous cell carcinoma (SCC), which occurs in the esophageal squamous cells (SCC is more commonly seen in China and Iran). 112 - 117 II) Adenocarcinoma, which occurs in the esophageal columnar cells or its glands. Adenocarcinoma is more common in developed countries, especially in patients with Barrett’s esophagus. It also mainly affects the cuboidal cells. 118 No symptoms may manifest in the early stages of the disease. With the progression of the disease, obstruction, difficulty in swallowing, and finally, weight loss may appear. Staging of the cancer is based on the invasion of the tumor into the esophageal wall, the number of affected lymph nodes, and the occurrence of metastases to different parts of the body. Radiotherapy and chemotherapy are often needed for the management of this disease. Moreover, a partial or full-thickness surgical removal of the esophagus may be performed. 118

Esophageal Atresia

Esophageal atresia (EA) is the most common congenital atresia of the GI system. In neonates with EA, the upper and lower esophagi are not connected, which results in the esophagus having two separate parts. Therefore, food can’t pass to the stomach. Besides, affected babies sometimes have difficulty breathing. This condition is often accompanied by a tracheoesophageal fistula, a congenital defect in which a part of the esophagus is connected to the trachea or windpipe. In some children, the missing part of the esophagus is so large that the ends cannot be easily connected with surgery. This condition is known as long-gap esophagus atresia. Without a functioning esophagus, it is impossible to receive enough nutrition through the mouth. Babies with EA are also more prone to infections such as pneumonia and conditions such as acid reflux. 118

Caustic Ingestion

Caustics and corrosives cause tissue injury through a chemical reaction. In contrast with children in whom ingestion is usually accidental, caustic ingestion in adults occurs on purpose and usually after suicidal attempts. The majority (68 %) of cases worldwide involve children due to the unintentional ingestion of caustic chemicals. 118

Discussion

Biomaterials can be acquired from nature or synthesized in the laboratory using a distinctive chemical approach consisting of metal, polymer, or ceramic components. Biomaterials have their own advantages. For instance, one of the advantages of biologically derived biomaterials for esophageal repair over synthetic ones is their ability to incorporate an extracellular matrix, which may improve regeneration. 119 - 121 Concisely, alterations in different regeneration and management stages of inflammation and scar tissue formation are caused by properly-configured biologic scaffolds. The biomaterial-host collaboration helps this process through complex factors. These include both host-related factors, including age, immune system, stem cell populations, and total health state, and biomaterial-related factors, including source and composition, 122 - 125 efficiency of the biomaterial process, 126 , 127 post-processing modifications such as crosslinking and solubilization, 128 - 134 age of the source animal, 135 and surface topography. 136 , 137

To assess the usefulness of biomaterial-mediated esophageal repair, biologic scaffolds have been used in several large animal model studies. In early investigations, porcine-derived acellular small intestinal sub-mucosa (SIS) and urinary bladder matrix (UBM) were utilized for the reconstruction of patchy defects in a dog model. 138 Defects measuring 5 cm in length and encircling either 40% or 50% of the esophageal circumference, or even the entire circumference, were reconstructed using these materials. The stricture was formed within 45 days of implantation by the scaffolds used to repair the full-circumference segmental defects. As for stricture formation during a full-thickness full-circumference defect repair, later studies demonstrated the need for a native (i.e., host) tissue component for adequate esophageal reconstruction without stricture formation. In these investigations, the repair of esophageal defects enclosing different parts of the esophageal circumference was carried out by UBM-ECM.

Full-circumference full-thickness defects, full circumference mucosa resections, and full-thickness defects with 30% intact muscularis externa constituted the treatment groups. In addition, the reinforcement of surgical anastomoses of the esophagus was carried out by biologic scaffolds in a dog model. 138 Afterward, the endoscopic implementation of biologic scaffolds was considered for mucosa repair performed after endoscopic mucosa resection in the dogs. Biologically-made biomaterials have also been investigated in small animal models. The objectives of small animal studies, unlike large animal models, include determining the mechanisms of tissue reconstruction, screening large numbers of potential therapies, and optimizing treatment options through the systematic advancement of design specifications. For instance, a murine model of esophageal reconstruction with chimeric mice has been utilized, which constitutively expressed green fluorescent protein in the bone marrow. 139 , 140

Similar research used gastric acellular matrix in a rat model for repairing the esophageal patch defects formed in the abdominal esophagus. 141 In the mentioned study, no stenosis or dilation was observed in the implant site, when the rats were sacrificed one week to 18 months after the implantation. At the two week mark, regeneration in the entire fabrication with keratinized stratified squamous epithelium was observed. However, the muscle layer or lamina muscularis mucosa was not restored.

Limited success has been achieved using various synthetic biomaterials for esophageal repair in different studies. Combining a synthetic biomechanical properties of materials with the biocompatible features of biologic materials as hybrid fabrications, usually as a coating agent, is becoming increasingly popular in regenerative medicine, 142 - 144 and this has been investigated for esophageal repair. Collagen-coated Vicryl tubes have also been used to replace complete esophageal segments within the thoracic esophagus. 145 Initial results showed prosthetic leakage secondary to acid reflux and digestion of the construct, which resulted in mediastinitis within days following the implantation. Increased material resistance due to the crosslinking of the constructs with glutaraldehyde was the main observed complication; however, stenosis was observed in the animals at an average of 11 days postoperatively, and histologically, substantial granulation tissue and scar formation were observed. Collagen, in addition to being used for coating Vicryl tubes, has also been utilized to coat silicone stents. 93 , 146 In these experiments, they used collagen-coated silicone tubes to replace 5 cm esophageal segmental defects in dogs. At weekly intervals ranging from two to four weeks, the endoscopic removal of the inner silicone stents was performed. When stent removal was carried out at the two to three weeks mark, stricture formation and incapability to swallow were reported. However, at the four weeks mark, a regenerated esophagus with stratified flattened epithelial, striated muscle, and esophageal glands was observed in the dogs. Despite the use of various synthetic materials for the reconstruction of esophageal defects, certain difficulties had emerged, such as having to obtain the proper mechanical strength; on the other hand, stricture formation, inflammation, foreign body reaction, and leakage were observed as complications. Some recent works using synthetic biomaterial for full-thickness esophagus replacement are presented in table 1.

Biomaterial Cell In vivo/in vitro Year Fabrication
PCL-Gelatin 147 - In vitro 2013 Electro-spinning
PLGA-PCL 148 Epithelial and smooth muscle cells In vitro 2015 Electro-spinning
Pluronic F127-PCL 149 Human esophageal fibroblasts In vitro 2018 Electro-hydrodynamic jetting
PCL-SF 150 Epithelial and smooth muscle cells In vivo (rat) 2015 Electro-spinning
PU-PCL 151 MSC In vivo (rat) 2019 3D printed and electro-spinning
Poly(l-lactide-co-ε-caprolactone) (PLC) 152 - In vitro 2016 Melt-drawing method
PU 153 Esophageal mucosa cells In vivo (pig) 2018 Electro-spinning
Polyamide-6 154 AD-MSC and BMD-MSC In vitro 2019 Electro-spinning
PU 155 MSC In vivo (pig) 2018 Electro-spinning-bioreactor
PCL 156 MSC In vivo (rabbit) 2016 3D printing
PCL 157 Mucosa and Muscular cell In vitro 2020 3D Bio-printing
PCL and PU 158 ADSC In vivo (rat) 2020 Electro-spinning-3D printing
PCL: Polycaprolactone; PLGA: Poly Lactic-co-Glycolic Acid; PU: Polyurethane; MSC: Mesenchymal stem cell; ADSC: Adipose-derived stem cell; BMD: Bone marrow-derived
Table 1.Synthetic biomaterials used for a circumferential esophageal replacement since 2013

Conclusion

As a proximal part of the alimentary tract, the esophagus is a complex organ containing multiple tissue layers that cannot regenerate. In recent years, esophageal tissue engineering has become a pioneering specialty for the treatment of esophagus diseases. Despite the different available approaches and the achieved advancements, a gold standard for fully efficient tissue-engineered esophageal fabrication is not yet defined. Various numbers of scaffolds, ranging from non-biodegradable stents to bioactive matrices, have been investigated for esophagus reconstruction, a goal that is now followed by making multi-layered scaffolds that imitate the behavior of different layers of the natural esophagus. The results are still not favorable due to many challenges relating to tissue quality, which requires improvement. The success of esophageal tissue regeneration will finally depend on the capability of the scaffold to resemble natural tissue properties and yield a qualified environment for regeneration. This emphasizes the importance of the scaffold design and fabrication technique.

Authors’ Contribution

M.H, M.H.I, Y.G, and A.A.A: Contributed to initial plan; M.H: Contributed to drafting the manuscript; M.H, H.H, Y.G: Contributed to data acquisition; M.H, H.H, and Y.G: Contributed to drafting the manuscript; M.H.I and A.A.A: contributed to the critical revision. All authors have read and approved the final manuscript and agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Conflict of Interest

None declared.

References

  1. Goyal RK, Chaudhury A. Physiology of normal esophageal motility. J Clin Gastroenterol. 2008; 42:610-9. Publisher Full Text | DOI | PubMed
  2. Goyal RK, Biancani P, Phillips A, Spiro HM. Mechanical properties of the esophageal wall. J Clin Invest. 1971; 50:1456-65. Publisher Full Text | DOI | PubMed
  3. Brasseur JG, Nicosia MA, Pal A, Miller LS. Function of longitudinal vs circular muscle fibers in esophageal peristalsis, deduced with mathematical modeling. World J Gastroenterol. 2007; 13:1335-46. Publisher Full Text | DOI | PubMed
  4. Oezcelik A, DeMeester SR. General anatomy of the esophagus. Thorac Surg Clin. 2011; 21:289-97. DOI | PubMed
  5. Ferhatoglu MF, Kıvılcım T. Esophageal Abnormalities. IntechOpen: London; 2017. DOI
  6. Moore KL, Dalley AF, Agur AM. Clinically oriented anatomy. Philadelphia: Lippincott Williams and Wilkins; 2013.
  7. Swigart LL, Siekert RG, et al. The esophageal arteries; an anatomic study of 150 specimens. Surg Gynecol Obstet. 1950; 90:234-43. PubMed
  8. Patti MG, Gantert W, Way LW. Surgery of the esophagus. Anatomy and physiology. Surg Clin North Am. 1997; 77:959-70. DOI | PubMed
  9. Drake R, Vogl AW, Mitchell A. Gray’s Anatomy for Students-Rental: With Student Consult Online Access. Elsevier Health Sciences: Amsterdam; 2009.
  10. Guyton A, Hall J. Textbook of medical physiology. Elsevier Inc: Amsterdam; 2006.
  11. Mu L, Wang J, Su H, Sanders I. Adult human upper esophageal sphincter contains specialized muscle fibers expressing unusual myosin heavy chain isoforms. J Histochem Cytochem. 2007; 55:199-207. DOI | PubMed
  12. Kahrilas PJ. Gastroesophageal reflux disease. JAMA. 1996; 276:983-8. PubMed
  13. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011; 61:69-90. DOI | PubMed
  14. Lambert R, Hainaut P. The multidisciplinary management of gastrointestinal cancer. Epidemiology of oesophagogastric cancer. Best Pract Res Clin Gastroenterol. 2007; 21:921-45. DOI | PubMed
  15. Haight C. Some observations on esophageal atresias and tracheo-esophageal fistulas of congenital origin. Journal of Thoracic Surgery. 1957; 34:141-72. DOI
  16. Kyyronen P, Hemminki K. Gastro-intestinal atresias in Finland in 1970-79, indicating time-place clustering. J Epidemiol Community Health. 1988; 42:257-65. Publisher Full Text | DOI | PubMed
  17. Spitz L. Oesophageal atresia. Orphanet J Rare Dis. 2007; 2:24. Publisher Full Text | DOI | PubMed
  18. Maghsoudlou P, Eaton S, De Coppi P. Tissue engineering of the esophagus. Semin Pediatr Surg. 2014; 23:127-34. DOI | PubMed
  19. Waisman H. The Surgery of Infancy and Childhood: Its Principles and Techniques. American Journal of Physical Medicine & Rehabilitation. 1953; 32:392.
  20. Malakounides G, Lyon P, Cross K, Pierro A, De Coppi P, Drake D, et al. Esophageal Atresia: Improved Outcome in High-Risk Groups Revisited. Eur J Pediatr Surg. 2016; 26:227-31. DOI | PubMed
  21. Kovesi T, Rubin S. Long-term complications of congenital esophageal atresia and/or tracheoesophageal fistula. Chest. 2004; 126:915-25. DOI | PubMed
  22. Ron O, De Coppi P, Pierro A. The surgical approach to esophageal atresia repair and the management of long-gap atresia: results of a survey. Semin Pediatr Surg. 2009; 18:44-9. DOI | PubMed
  23. Deschamps C. History of esophageal surgery for benign disease. Chest Surg Clin N Am. 2000; 10:135-44. PubMed
  24. Battersby JS, King H. Esophageal replacement with plastic tubes; experimental study. AMA Arch Surg. 1954; 69:400-9. DOI | PubMed
  25. Berman E. Supplemental experiments in synthetic esophageal replacement. Surgical Forum: Hong Kong; 1956.
  26. Lyons AS, Beck AR, Lester LJ. Esophageal replacement with prosthesis. Preliminary report of experimental studies. J Surg Res. 1962; 2:110-3. DOI | PubMed
  27. Bergan F, Bie K. Replacement of the Oesophagus by a Colonic Segment. Acta Chir Scand. 1963; 126:566-72. PubMed
  28. Burrington JD, Stephens CA. Esophageal replacement with a gastric tube in infants and children. J Pediatr Surg. 1968; 3:24-52. DOI | PubMed
  29. Heimlich HJ. Esophageal replacement with a reversed gastric tube. Dis Chest. 1959; 36:478-93. DOI | PubMed
  30. Heimlich HJ. Replacement of the entire esophagus for malignant or benign stenosis. Am J Gastroenterol. 1961; 35:311-25. PubMed
  31. Pellett JR. Colon Interposition for Esophageal Bypass or Replacement. Indications, Limitations, and Complications. Arch Surg. 1964; 89:169-79. DOI | PubMed
  32. Sanders GB. Esophageal replacement with reversed gastric tube. Utilization for bleeding esophageal varices in a four-year-old child. JAMA. 1962; 181:944-7. DOI | PubMed
  33. Yamagishi M, Ikeda N, Yonemoto T. An isoperistaltic gastric tube. New method of esophageal replacement. Arch Surg. 1970; 100:689-92. DOI | PubMed
  34. Young GA, Dagradi AE. Replacement of the cervical segment of the esophagus with an aortic homograft. Am J Surg. 1961; 102:687-90. DOI | PubMed
  35. LaGuerre JN, Schoenfeld H, Calem W, Gould FE, Levowitz BS. Prosthetic replacement of esophageal segments. J Thorac Cardiovasc Surg. 1968; 56:674-82. PubMed
  36. Leininger BJ, Peacock H, Neville WE. Esophageal mucosal regeneration following experimental prosthetic replacement of the esophagus. Surgery. 1970; 67:468-73. PubMed
  37. Rhee K, Kim JH, Jung DH, Han JW, Lee YC, Lee SK, et al. Self-expandable metal stents for malignant esophageal obstruction: a comparative study between extrinsic and intrinsic compression. Dis Esophagus. 2016; 29:224-8. DOI | PubMed
  38. Sioulas AD, Malli C, Dimitriadis GD, Triantafyllou K. Self-expandable metal stents for achalasia: Thinking out of the box!. World J Gastrointest Endosc. 2015; 7:45-52. Publisher Full Text | DOI | PubMed
  39. van Halsema EE, van Hooft JE. Clinical outcomes of self-expandable stent placement for benign esophageal diseases: A pooled analysis of the literature. World J Gastrointest Endosc. 2015; 7:135-53. Publisher Full Text | DOI | PubMed
  40. Zhao H, Wan XJ, Yang CQ. Comparison of endoscopic balloon dilation with metal stent placement in the treatment of achalasia. J Dig Dis. 2015; 16:311-8. DOI | PubMed
  41. Bhat MA, Dar MA, Lone GN, Dar AM. Use of pedicled omentum in esophagogastric anastomosis for prevention of anastomotic leak. Ann Thorac Surg. 2006; 82:1857-62. DOI | PubMed
  42. Dai JG, Zhang ZY, Min JX, Huang XB, Wang JS. Wrapping of the omental pedicle flap around esophagogastric anastomosis after esophagectomy for esophageal cancer. Surgery. 2011; 149:404-10. DOI | PubMed
  43. Dicks JR, Majeed AW, Stoddard CJ. Omental wrapping of perforated esophagus. Dis Esophagus. 1998; 11:276-8. DOI | PubMed
  44. Cense HA, Visser MR, van Sandick JW, de Boer AG, Lamme B, Obertop H, et al. Quality of life after colon interposition by necessity for esophageal cancer replacement. J Surg Oncol. 2004; 88:32-8. DOI | PubMed
  45. Greene CL, DeMeester SR, Worrell SG, Oh DS, Hagen JA, DeMeester TR. Alimentary satisfaction, gastrointestinal symptoms, and quality of life 10 or more years after esophagectomy with gastric pull-up. J Thorac Cardiovasc Surg. 2014; 147:909-14. DOI | PubMed
  46. Reismann M, Granholm T, Ehren H. Partial gastric pull-up in the treatment of patients with long-gap esophageal atresia. World J Pediatr. 2015; 11:267-71. DOI | PubMed
  47. Silver CE. Gastric pull-up operation for replacement of the cervical portion of the esophagus. Surg Gynecol Obstet. 1976; 142:243-5. PubMed
  48. Sreehariprasad AV, Krishnappa R, Chikaraddi BS, Veerendrakumar K. Gastric pull up reconstruction after pharyngo laryngo esophagectomy for advanced hypopharyngeal cancer. Indian J Surg Oncol. 2012; 3:4-7. Publisher Full Text | DOI | PubMed
  49. Davis PA, Law S, Wong J. Colonic interposition after esophagectomy for cancer. Arch Surg. 2003; 138:303-8. DOI | PubMed
  50. DeMeester SR. Colon interposition following esophagectomy. Dis Esophagus. 2001; 14:169-72. DOI | PubMed
  51. Swisher SG, Hofstetter WL, Miller MJ. The supercharged microvascular jejunal interposition. Semin Thorac Cardiovasc Surg. 2007; 19:56-65. DOI | PubMed
  52. Harashina T, Wada M, Imai T, Kakegawa T. A turnover de-epithelialised deltopectoral flap to close fistulae following antethoracic oesophageal reconstruction. Br J Plast Surg. 1979; 32:278-80. DOI | PubMed
  53. Murono S, Ishikawa E, Nakanishi Y, Endo K, Kondo S, Wakisaka N, et al. Closure of tracheoesophageal fistula with prefabricated deltopectoral flap. Asian J Surg. 2016; 39:243-6. DOI | PubMed
  54. Ramadan MF, Stell PM. Reconstruction after pharyngolaryngo-oesophagectomy using delto-pectoral flap. Clin Otolaryngol Allied Sci. 1979; 4:5-11. DOI | PubMed
  55. Fabian RL. Pectoralis major myocutaneous flap reconstruction of the laryngopharynx and cervical esophagus. Laryngoscope. 1988; 98:1227-31.
  56. Hobor B, Borbely L, Halmos L, Horvath OP. The replacement of the esophagus by musculocutaneous flaps. Acta Chir Hung. 1997; 36:132-3. PubMed
  57. Murakami Y, Saito S, Ikari T, Haraguchi S, Okada K, Maruyama T. Esophageal reconstruction with a skin-grafted pectoralis major muscle flap. Arch Otolaryngol. 1982; 108:719-22. DOI | PubMed
  58. Russell RC, Feller AM, Elliott LF, Kucan JO, Zook EG. The extended pectoralis major myocutaneous flap: uses and indications. Plast Reconstr Surg. 1991; 88:814-23. DOI | PubMed
  59. Mecklenburg I, Probst A, Messmann H. Esophagospinal fistula with spondylodiscitis and meningitis after esophagectomy with gastric pull-up. J Gastrointest Surg. 2008; 12:394-5. DOI | PubMed
  60. Vijay K, Godara R, Vijayvergia V. Failed Gastric Pull up after Esophagectomy Managed by Colonic Interposition. Indian J Surg. 2013; 75:347-9. Publisher Full Text | DOI | PubMed
  61. Bailey SH, Bull DA, Harpole DH, Rentz JJ, Neumayer LA, Pappas TN, et al. Outcomes after esophagectomy: a ten-year prospective cohort. Ann Thorac Surg. 2003; 75:217-22. DOI | PubMed
  62. Blencowe NS, Strong S, McNair AG, Brookes ST, Crosby T, Griffin SM, et al. Reporting of short-term clinical outcomes after esophagectomy: a systematic review. Ann Surg. 2012; 255:658-66. DOI | PubMed
  63. Metzger R, Bollschweiler E, Vallbohmer D, Maish M, DeMeester TR, Holscher AH. High volume centers for esophagectomy: what is the number needed to achieve low postoperative mortality?. Dis Esophagus. 2004; 17:310-4. DOI | PubMed
  64. Yoshida N, Watanabe M, Baba Y, Iwagami S, Ishimoto T, Iwatsuki M, et al. Risk factors for pulmonary complications after esophagectomy for esophageal cancer. Surg Today. 2014; 44:526-32. DOI | PubMed
  65. Egorov VI, Schastlivtsev IV, Prut EV, Baranov AO, Turusov RA. Mechanical properties of the human gastrointestinal tract. Journal of Biomechanics. 2002; 35:1417-25. DOI
  66. Yamada H, Evans FG. Strength of biological materials. Agris (FAO): Angouleme; 1970.
  67. Crapo PM, Gilbert TW, Badylak SF. An overview of tissue and whole organ decellularization processes. Biomaterials. 2011; 32:3233-43. Publisher Full Text | DOI | PubMed
  68. Gilbert TW, Sellaro TL, Badylak SF. Decellularization of tissues and organs. Biomaterials. 2006; 27:3675-83. DOI | PubMed
  69. Jank BJ, Xiong L, Moser PT, Guyette JP, Ren X, Cetrulo CL, et al. Engineered composite tissue as a bioartificial limb graft. Biomaterials. 2015; 61:246-56. Publisher Full Text | DOI | PubMed
  70. Dua KS, Hogan WJ, Aadam AA, Gasparri M. In-vivo oesophageal regeneration in a human being by use of a non-biological scaffold and extracellular matrix. The Lancet. 2016; 388:55-61. DOI
  71. Thomas M, Allen MS, Shen KR, Wigle DA. A novel use of human acellular dermis for conduit salvage after esophagectomy. Ann Thorac Surg. 2014; 97:1459-63. DOI | PubMed
  72. Aho JM, Dietz AB, Radel DJ, Butler GW, Thomas M, Nelson TJ, et al. Closure of a Recurrent Bronchopleural Fistula Using a Matrix Seeded With Patient-Derived Mesenchymal Stem Cells. Stem Cells Transl Med. 2016; 5:1375-9. Publisher Full Text | DOI | PubMed
  73. Alvarez PD, Garcia-Arranz M, Georgiev-Hristov T, Garcia-Olmo D. A new bronchoscopic treatment of tracheomediastinal fistula using autologous adipose-derived stem cells. Thorax. 2008; 63:374-6. DOI | PubMed
  74. Petrella F, Spaggiari L, Acocella F, Barberis M, Bellomi M, Brizzola S, et al. Airway fistula closure after stem-cell infusion. N Engl J Med. 2015; 372:96-7. DOI | PubMed
  75. Dave M, Mehta K, Luther J, Baruah A, Dietz AB, Faubion WA. Mesenchymal Stem Cell Therapy for Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. Inflamm Bowel Dis. 2015; 21:2696-707. Publisher Full Text | DOI | PubMed
  76. Liu X, Fang Q, Kim H. Preclinical Studies of Mesenchymal Stem Cell (MSC) Administration in Chronic Obstructive Pulmonary Disease (COPD): A Systematic Review and Meta-Analysis. PLoS One. 2016; 11:e0157099. Publisher Full Text | DOI | PubMed
  77. Petrella F, Toffalorio F, Brizzola S, De Pas TM, Rizzo S, Barberis M, et al. Stem cell transplantation effectively occludes bronchopleural fistula in an animal model. Ann Thorac Surg. 2014; 97:480-3. DOI | PubMed
  78. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006; 8:315-7. DOI | PubMed
  79. Berman EF. The experimental replacement of portions of the esophagus by a plastic tube. Ann Surg. 1952; 135:337-43. Publisher Full Text | DOI | PubMed
  80. Fryfogle JD, Cyrowski GA, Rothwell D, Rheault G, Clark T. Replacement of the middle third of the esophagus with a silicone rubber prosthesis. An experiment and clinical studys. Dis Chest. 1963; 43:464-75. DOI | PubMed
  81. Lister J, Altman RP, Allison WA. Prosthetic Substitution of Thoracic Esophagus in Puppies: Use of Marlex Mesh with Collagen or Anterior Rectus Sheath. Ann Surg. 1965; 162:812-24. Publisher Full Text | DOI | PubMed
  82. Fukushima M, Kako N, Chiba K, Kawaguchi T, Kimura Y, Sato M, et al. Seven-year follow-up study after the replacement of the esophagus with an artificial esophagus in the dog. Surgery. 1983; 93:70-7. PubMed
  83. Takimoto Y, Nakamura T, Teramachi M, Kiyotani T, Shimizu Y. Replacement of long segments of the esophagus with a collagen-silicone composite tube. ASAIO J. 1995; 41:M605-8. DOI | PubMed
  84. Takimoto Y, Okumura N, Nakamura T, Natsume T, Shimizu Y. Long-term follow-up of the experimental replacement of the esophagus with a collagen-silicone composite tube. ASAIO J. 1993; 39:M736-9. PubMed
  85. Spitz L. Esophageal replacement: overcoming the need. J Pediatr Surg. 2014; 49:849-52. DOI | PubMed
  86. Totonelli G, Maghsoudlou P, Fishman JM, Orlando G, Ansari T, Sibbons P, et al. Esophageal tissue engineering: a new approach for esophageal replacement. World J Gastroenterol. 2012; 18:6900-7. Publisher Full Text | DOI | PubMed
  87. Earlam R, Cunha-Melo JR. Malignant oesophageal strictures: a review of techniques for palliative intubation. Br J Surg. 1982; 69:61-8. DOI | PubMed
  88. Liang JH, Zhou X, Zheng ZB, Liang XL. Long-term form and function of neoesophagus after experimental replacement of thoracic esophagus with nitinol composite artificial esophagus. ASAIO J. 2010; 56:232-4. DOI | PubMed
  89. Nakase Y, Nakamura T, Kin S, Nakashima S, Yoshikawa T, Kuriu Y, et al. Intrathoracic esophageal replacement by in situ tissue-engineered esophagus. J Thorac Cardiovasc Surg. 2008; 136:850-9. DOI | PubMed
  90. Beckstead BL, Pan S, Bhrany AD, Bratt-Leal AM, Ratner BD, Giachelli CM. Esophageal epithelial cell interaction with synthetic and natural scaffolds for tissue engineering. Biomaterials. 2005; 26:6217-28. DOI | PubMed
  91. Diemer P, Markoew S, Le DQ, Qvist N. Poly-epsilon-caprolactone mesh as a scaffold for in vivo tissue engineering in rabbit esophagus. Dis Esophagus. 2015; 28:240-5. DOI | PubMed
  92. Lynen Jansen P, Klinge U, Anurov M, Titkova S, Mertens PR, Jansen M. Surgical mesh as a scaffold for tissue regeneration in the esophagus. Eur Surg Res. 2004; 36:104-11. DOI | PubMed
  93. Natsume T, Ike O, Okada T, Takimoto N, Shimizu Y, Ikada Y. Porous collagen sponge for esophageal replacement. J Biomed Mater Res. 1993; 27:867-75. DOI | PubMed
  94. Miki H, Ando N, Ozawa S, Sato M, Hayashi K, Kitajima M. An artificial esophagus constructed of cultured human esophageal epithelial cells, fibroblasts, polyglycolic acid mesh, and collagen. ASAIO J. 1999; 45:502-8. DOI | PubMed
  95. Zhu Y, Chan-Park MB, Sin Chian K. The growth improvement of porcine esophageal smooth muscle cells on collagen-grafted poly(DL-lactide-co-glycolide) membrane. J Biomed Mater Res B Appl Biomater. 2005; 75:193-9. DOI | PubMed
  96. Zhu Y, Chian KS, Chan-Park MB, Mhaisalkar PS, Ratner BD. Protein bonding on biodegradable poly(L-lactide-co-caprolactone) membrane for esophageal tissue engineering. Biomaterials. 2006; 27:68-78. DOI | PubMed
  97. Bitar KN, Zakhem E. Tissue engineering and regenerative medicine as applied to the gastrointestinal tract. Curr Opin Biotechnol. 2013; 24:909-15. Publisher Full Text | DOI | PubMed
  98. Hou L, Gong C, Zhu Y. In vitro construction and in vivo regeneration of esophageal bilamellar muscle tissue. J Biomater Appl. 2016; 30:1373-84. DOI | PubMed
  99. Saxena AK, Ainoedhofer H, Hollwarth ME. Esophagus tissue engineering: in vitro generation of esophageal epithelial cell sheets and viability on scaffold. J Pediatr Surg. 2009; 44:896-901. DOI | PubMed
  100. Homayoni H, Ravandi SAH, Valizadeh M. Electrospinning of chitosan nanofibers: Processing optimization. Carbohydrate polymers. 2009; 77:656-61. DOI
  101. Geng X, Kwon OH, Jang J. Electrospinning of chitosan dissolved in concentrated acetic acid solution. Biomaterials. 2005; 26:5427-32. DOI | PubMed
  102. Pisani S, Dorati R, Conti B, Modena T, Bruni G, Genta I. Design of copolymer PLA-PCL electrospun matrix for biomedical applications. Reactive and Functional Polymers. 2018; 124:77-89. DOI
  103. Reneker DH, Yarin AL, Fong H, Koombhongse S. Bending instability of electrically charged liquid jets of polymer solutions in electrospinning. Journal of Applied physics. 2000; 87:4531-47. DOI
  104. Thota PN, Kistangari G, Esnakula AK, Gonzalo DH, Liu XL. Clinical significance and management of Barrett’s esophagus with epithelial changes indefinite for dysplasia. World J Gastrointest Pharmacol Ther. 2016; 7:406-11. Publisher Full Text | DOI | PubMed
  105. Arnold M, Soerjomataram I, Ferlay J, Forman D. Global incidence of oesophageal cancer by histological subtype in 2012. Gut. 2015; 64:381-7. DOI | PubMed
  106. Depaepe A, Dolk H, Lechat MF. The epidemiology of tracheo-oesophageal fistula and oesophageal atresia in Europe. EUROCAT Working Group. Arch Dis Child. 1993; 68:743-8. Publisher Full Text | DOI | PubMed
  107. Goyal A, Jones MO, Couriel JM, Losty PD. Oesophageal atresia and tracheo-oesophageal fistula. Arch Dis Child Fetal Neonatal Ed. 2006; 91:F381-4. Publisher Full Text | DOI | PubMed
  108. Keckler SJ, St Peter SD, Valusek PA, Tsao K, Snyder CL, Holcomb GW, et al. VACTERL anomalies in patients with esophageal atresia: an updated delineation of the spectrum and review of the literature. Pediatr Surg Int. 2007; 23:309-13. DOI | PubMed
  109. Kikendall JW. Caustic ingestion injuries. Gastroenterol Clin North Am. 1991; 20:847-57. PubMed
  110. Smith M, Zhou M, Whitlock G, Yang G, Offer A, Hui G, et al. Esophageal cancer and body mass index: results from a prospective study of 220,000 men in China and a meta-analysis of published studies. Int J Cancer. 2008; 122:1604-10. DOI | PubMed
  111. Lin CC, Papadopoulos KP. Novel targeted therapies for advanced esophageal cancer. Dis Esophagus. 2007; 20:365-71. DOI | PubMed
  112. Abbaszadegan MR, Keyvani V, Moghbeli M. Genetic and molecular bases of esophageal Cancer among Iranians: an update. Diagn Pathol. 2019; 14:97. Publisher Full Text | DOI | PubMed
  113. Mehdizadeh H, Mahmoudi G, Moslemi D, Bijani A, Jahani MA. A 25-year trend in gastrointestinal cancers in northern Iran (1991-2016). Caspian J Intern Med. 2019; 10:396-401. Publisher Full Text | DOI | PubMed
  114. Moradzadeh R, Golmohammadi P, Ghaitasi B, Nadrian H, Najafi A. Incidence of Esophageal Cancer in Iran, a Population-Based Study: 2001-2015. J Gastrointest Cancer. 2019; 50:507-12. DOI | PubMed
  115. Rafiemanesh H, Maleki F, Mohammadian-Hafshejani A, Salemi M, Salehiniya H. The Trend in Histological Changes and the Incidence of Esophagus Cancer in Iran (2003-2008). Int J Prev Med. 2016; 7:31. Publisher Full Text | DOI | PubMed
  116. Roshandel G, Ghanbari-Motlagh A, Partovipour E, Salavati F, Hasanpour-Heidari S, Mohammadi G, et al. Cancer incidence in Iran in 2014: Results of the Iranian National Population-based Cancer Registry. Cancer Epidemiol. 2019; 61:50-8. DOI | PubMed
  117. Zarean E, Amini P, Yaseri M, Hajihosseini M, Azimi T, Mahmoudi M. Determining Risk Factors for Gastric and Esophageal Cancers between 2009-2015 in East-Azarbayjan, Iran Using Parametric Survival Models. Asian Pac J Cancer Prev. 2019; 20:443-9. Publisher Full Text | DOI | PubMed
  118. Colledge NR, Walker BR, Ralston SH. Davidson’s principles and practice of medicine: Churchill Livingstone. Elsevier Edinburgh: New York; 2010.
  119. Badylak SF. The extracellular matrix as a scaffold for tissue reconstruction. Seminars in Cell & Developmental Biology. 2002; 13:377-83. DOI
  120. Badylak SF, Vorp DA, Spievack AR, Simmons-Byrd A, Hanke J, Freytes DO, et al. Esophageal reconstruction with ECM and muscle tissue in a dog model. J Surg Res. 2005; 128:87-97. DOI | PubMed
  121. Londono R, Badylak SF. Biologic scaffolds for regenerative medicine: mechanisms of in vivo remodeling. Ann Biomed Eng. 2015; 43:577-92. DOI | PubMed
  122. Crapo PM, Medberry CJ, Reing JE, Tottey S, van der Merwe Y, Jones KE, et al. Biologic scaffolds composed of central nervous system extracellular matrix. Biomaterials. 2012; 33:3539-47. Publisher Full Text | DOI | PubMed
  123. Keane TJ, Londono R, Carey RM, Carruthers CA, Reing JE, Dearth CL, et al. Preparation and characterization of a biologic scaffold from esophageal mucosa. Biomaterials. 2013; 34:6729-37. Publisher Full Text | DOI | PubMed
  124. Wainwright JM, Czajka CA, Patel UB, Freytes DO, Tobita K, Gilbert TW, et al. Preparation of cardiac extracellular matrix from an intact porcine heart. Tissue Eng Part C Methods. 2010; 16:525-32. Publisher Full Text | DOI | PubMed
  125. Wolf MT, Daly KA, Reing JE, Badylak SF. Biologic scaffold composed of skeletal muscle extracellular matrix. Biomaterials. 2012; 33:2916-25. Publisher Full Text | DOI | PubMed
  126. Brown BN, Valentin JE, Stewart-Akers AM, McCabe GP, Badylak SF. Macrophage phenotype and remodeling outcomes in response to biologic scaffolds with and without a cellular component. Biomaterials. 2009; 30:1482-91. Publisher Full Text | DOI | PubMed
  127. Keane TJ, Londono R, Turner NJ, Badylak SF. Consequences of ineffective decellularization of biologic scaffolds on the host response. Biomaterials. 2012; 33:1771-81. DOI | PubMed
  128. DeQuach JA, Lin JE, Cam C, Hu D, Salvatore MA, Sheikh F, et al. Injectable skeletal muscle matrix hydrogel promotes neovascularization and muscle cell infiltration in a hindlimb ischemia model. Eur Cell Mater. 2012; 23:400-12. Publisher Full Text | DOI | PubMed
  129. Johnson TD, Christman KL. Injectable hydrogel therapies and their delivery strategies for treating myocardial infarction. Expert Opin Drug Deliv. 2013; 10:59-72. DOI | PubMed
  130. Londono R, Badylak SF. Regenerative Medicine Strategies for Esophageal Repair. Tissue Eng Part B Rev. 2015; 21:393-410. Publisher Full Text | DOI | PubMed
  131. Seif-Naraghi SB, Horn D, Schup-Magoffin PJ, Christman KL. Injectable extracellular matrix derived hydrogel provides a platform for enhanced retention and delivery of a heparin-binding growth factor. Acta Biomater. 2012; 8:3695-703. Publisher Full Text | DOI | PubMed
  132. Singelyn JM, Sundaramurthy P, Johnson TD, Schup-Magoffin PJ, Hu DP, Faulk DM, et al. Catheter-deliverable hydrogel derived from decellularized ventricular extracellular matrix increases endogenous cardiomyocytes and preserves cardiac function post-myocardial infarction. J Am Coll Cardiol. 2012; 59:751-63. Publisher Full Text | DOI | PubMed
  133. Valentin JE, Stewart-Akers AM, Gilbert TW, Badylak SF. Macrophage participation in the degradation and remodeling of extracellular matrix scaffolds. Tissue Eng Part A. 2009; 15:1687-94. Publisher Full Text | DOI | PubMed
  134. Wolf MT, Daly KA, Brennan-Pierce EP, Johnson SA, Carruthers CA, D’Amore A, et al. A hydrogel derived from decellularized dermal extracellular matrix. Biomaterials. 2012; 33:7028-38. Publisher Full Text | DOI | PubMed
  135. Sicari BM, Johnson SA, Siu BF, Crapo PM, Daly KA, Jiang H, et al. The effect of source animal age upon the in vivo remodeling characteristics of an extracellular matrix scaffold. Biomaterials. 2012; 33:5524-33. Publisher Full Text | DOI | PubMed
  136. Barnes CA, Brison J, Michel R, Brown BN, Castner DG, Badylak SF, et al. The surface molecular functionality of decellularized extracellular matrices. Biomaterials. 2011; 32:137-43. Publisher Full Text | DOI | PubMed
  137. Brown BN, Barnes CA, Kasick RT, Michel R, Gilbert TW, Beer-Stolz D, et al. Surface characterization of extracellular matrix scaffolds. Biomaterials. 2010; 31:428-37. Publisher Full Text | DOI | PubMed
  138. Badylak SF. Decellularized allogeneic and xenogeneic tissue as a bioscaffold for regenerative medicine: factors that influence the host response. Ann Biomed Eng. 2014; 42:1517-27. DOI | PubMed
  139. Bhrany AD, Beckstead BL, Lang TC, Farwell DG, Giachelli CM, Ratner BD. Development of an esophagus acellular matrix tissue scaffold. Tissue Eng. 2006; 12:319-30. DOI | PubMed
  140. Bhrany AD, Lien CJ, Beckstead BL, Futran ND, Muni NH, Giachelli CM, et al. Crosslinking of an oesophagus acellular matrix tissue scaffold. J Tissue Eng Regen Med. 2008; 2:365-72. DOI | PubMed
  141. Urita Y, Komuro H, Chen G, Shinya M, Kaneko S, Kaneko M, et al. Regeneration of the esophagus using gastric acellular matrix: an experimental study in a rat model. Pediatr Surg Int. 2007; 23:21-6. DOI | PubMed
  142. Faulk DM, Londono R, Wolf MT, Ranallo CA, Carruthers CA, Wildemann JD, et al. ECM hydrogel coating mitigates the chronic inflammatory response to polypropylene mesh. Biomaterials. 2014; 35:8585-95. Publisher Full Text | DOI | PubMed
  143. Wolf MT, Carruthers CA, Dearth CL, Crapo PM, Huber A, Burnsed OA, et al. Polypropylene surgical mesh coated with extracellular matrix mitigates the host foreign body response. J Biomed Mater Res A. 2014; 102:234-46. Publisher Full Text | DOI | PubMed
  144. Wolf MT, Dearth CL, Ranallo CA, LoPresti ST, Carey LE, Daly KA, et al. Macrophage polarization in response to ECM coated polypropylene mesh. Biomaterials. 2014; 35:6838-49. Publisher Full Text | DOI | PubMed
  145. Purushotham AD, Carachi R, Gorham SD, French DA, Shivas AA. Use of a collagen coated vicryl tube in reconstruction of the porcine esophagus. Eur J Pediatr Surg. 1991; 1:80-4. DOI | PubMed
  146. Takimoto Y, Nakamura T, Yamamoto Y, Kiyotani T, Teramachi M, Shimizu Y. The experimental replacement of a cervical esophageal segment with an artificial prosthesis with the use of collagen matrix and a silicone stent. J Thorac Cardiovasc Surg. 1998; 116:98-106. DOI | PubMed
  147. Kuppan P, Sethuraman S, Krishnan UM. PCL and PCL-gelatin nanofibers as esophageal tissue scaffolds: optimization, characterization and cell-matrix interactions. J Biomed Nanotechnol. 2013; 9:1540-55. DOI | PubMed
  148. Jensen T, Blanchette A, Vadasz S, Dave A, Canfarotta M, Sayej WN, et al. Biomimetic and synthetic esophageal tissue engineering. Biomaterials. 2015; 57:133-41. DOI | PubMed
  149. Wu B, Takeshita N, Wu Y, Vijayavenkataraman S, Ho KY, Lu WF, et al. Pluronic F127 blended polycaprolactone scaffolds via e-jetting for esophageal tissue engineering. J Mater Sci Mater Med. 2018; 29:140. DOI | PubMed
  150. Chung EJ, Ju HW, Park HJ, Park CH. Three-layered scaffolds for artificial esophagus using poly(varepsilon-caprolactone) nanofibers and silk fibroin: An experimental study in a rat model. J Biomed Mater Res A. 2015; 103:2057-65. DOI | PubMed
  151. Kim IG, Wu Y, Park SA, Cho H, Choi JJ, Kwon SK, et al. Tissue-Engineered Esophagus via Bioreactor Cultivation for Circumferential Esophageal Reconstruction. Tissue Eng Part A. 2019; 25:1478-92. DOI | PubMed
  152. Tan YJ, Yeong WY, Tan X, An J, Chian KS, Leong KF. Characterization, mechanical behavior and in vitro evaluation of a melt-drawn scaffold for esophageal tissue engineering. J Mech Behav Biomed Mater. 2016; 57:246-59. DOI | PubMed
  153. Barron MR, Blanco EW, Aho JM, Chakroff J, Johnson J, Cassivi SD, et al. Full-thickness oesophageal regeneration in pig using a polyurethane mucosal cell seeded graft. J Tissue Eng Regen Med. 2018; 12:175-85. DOI | PubMed
  154. Zhuravleva M, Gilazieva Z, Grigoriev TE, Shepelev AD, Kh Tenchurin T, Kamyshinsky R, et al. In vitro assessment of electrospun polyamide-6 scaffolds for esophageal tissue engineering. J Biomed Mater Res B Appl Biomater. 2019; 107:253-68. DOI | PubMed
  155. La Francesca S, Aho JM, Barron MR, Blanco EW, Soliman S, Kalenjian L, et al. Long-term regeneration and remodeling of the pig esophagus after circumferential resection using a retrievable synthetic scaffold carrying autologous cells. Sci Rep. 2018; 8:4123. Publisher Full Text | DOI | PubMed
  156. Park SY, Choi JW, Park JK, Song EH, Park SA, Kim YS, et al. Tissue-engineered artificial oesophagus patch using three-dimensionally printed polycaprolactone with mesenchymal stem cells: a preliminary report. Interact Cardiovasc Thorac Surg. 2016; 22:712-7. Publisher Full Text | DOI | PubMed
  157. Nam H, Jeong HJ, Jo Y, Lee JY, Ha DH, Kim JH, et al. Multi-layered Free-form 3D Cell-printed Tubular Construct with Decellularized Inner and Outer Esophageal Tissue-derived Bioinks. Sci Rep. 2020; 10:7255. Publisher Full Text | DOI | PubMed
  158. Park H, Kim IG, Wu Y, Cho H, Shin JW, Park SA, et al. Experimental investigation of esophageal reconstruction with electrospun polyurethane nanofiber and 3D printing polycaprolactone scaffolds using a rat model. Head Neck. 2021; 43:833-48. DOI | PubMed