#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Optimizing ischemic preconditioning for esophagogastric anastomosis: A standardized rat model to evaluate histological changes


Authors: I. Kovac 1;  Z. Hribíková 2;  M. Miklosova 1;  K. Kovacova 2;  J. Gajdos 2;  D. Matiova 3;  D. Petrasova 3;  J. Belák 1
Authors‘ workplace: Department of Pathology, Louis Pasteur University Hospital and Pavol Jozef Safarik University, Kosice, Slovak Republic 2;  Laboratory of Research Bio-models, Pavol Jozef Safarik University, Kosice, Slovak Republic 3;  nd Department of Surgery, Louis Pasteur University Hospital and Pavol Jozef Safarik University, Kosice, Slovak Republic 12
Published in: Rozhl. Chir., 2024, roč. 103, č. 3, s. 84-90.
Category: Original articles
doi: https://doi.org/10.33699/PIS.2024.103.3.84–90

Overview

Introduction: A reproducible and simple model is essential for verifying gastric conduit vitality before esophagectomy. Ischemia is a major cause of esophagogastric anastomotic dehiscence and leakage. Ischemic conditioning of the stomach prior to esophageal surgery has been shown to lower the incidence of postoperative complications, including anastomotic leakage. However, the optimal timing and technique of ischemization remain uncertain.

Methods: Male Sprague-Dawley rats (n=24) were randomly divided into four groups: ischemic group – samples collected 1 hour after ischemia (I1H), ischemic group – samples collected 1 day after ischemia (I1D), ischemic group – samples collected 7 days after ischemia (I7D), and control group (C). Ischemia was induced by ligation of the left gastric (LGA) and short gastric arteries (SGA). The samples were verified using histological and macroscopic analysis, and the number and percentage of immunocompetent cells were determined.

Results: One hour after ischemization (I1H), ischemic denudation with mucosal erosion was observed, and the total number of eosinophils was significantly higher (p<0.05) in the I1H group compared to the I1D and I7D groups. One day after ischemia (I1D), there was a reduction in the inflammatory response with partial regeneration of gastric mucosa. In the I7D group, nearly complete architectural regeneration of mucosal epithelium was documented. The total count of polymorphonuclears was significantly lower (p<0.05) compared to the I1D group.

Conclusion: Ischemic mucosal injury after LGA and SGA ligation was observed dominantly in the I1H and I1D groups, but not in I7D group. In conclusion, this study presents a simple method for verifying gastric ischemic changes.

Keywords:

rat – Stomach – ischemia – esophagogastric anastomosis – histological changes

INTRODUCTION

Anastomotic dehiscence and leakage have been reported in 10% of patients after esophageal resection for cancer and represent a serious complication resulting in significant morbidity and mortality [1]. Depending on the site of the tumor, cervical or intrathoracic anastomoses are usually constructed using gastric conduit [2]. Although esophagogastric anastomosis is more fragile compared to all other anastomoses of the gastrointestinal tract, especially when cervical anastomosis for higher reconstruction is indicated, many surgeons prefer to use cervical anastomosis rather than intrathoracic [3]. Many factors are linked to the failure of esophagogastric anastomosis. One of the most delicate aspects is the ischemia of the gastric conduit. Ischemia is a major cause of anastomotic dehiscence leading to leakage and a sequence of fatal aberrances. Hence, the elimination of anastomotic leakages is essential for improving the morbidity and mortality after esophagectomy [4].

The stomach is supplied by the left gastric artery (LGA), right gastric artery (RGA), and gastroepiploic arteries. Three out of four main branches are closed by ligation during gastric mobilization, and the right gastroepiploic artery (RGEA) remains the principal supplier of the gastric conduit [5]. The upper part of the gastric tube is vascularized only by the intramural network and is thus the most vulnerable part for ischemia [6,7]. An alteration of venous drainage of the gastric tube may also increase the possibility of ischemia of this special conduit. The degree of ischemia could be amplified by forced manipulation with tension and strangulation during surgery [4].

A shortage in blood flow to any organ or tissue causes cellular dysfunction and death [8]. After a certain period of ischemia, the gastrointestinal tissue becomes vulnerable to damage due to reperfusion, in which the restored oxygen supply produces free radical species that lead to further tissue injury. Ischemic reperfusion damage after clamping the celiac artery induces gross damage to gastric mucosa, resulting in the formation of petechiae and ulcers deep down in the mucosa [9]. It is believed that ischemia after LGA, RGA, and left gastroepiploic artery (LGEA) ligation is the dominant factor resulting in future anastomotic leakage [10]. Therefore, numerous experimental and clinical studies have focused on the prevention of ischemia. Gastric ischemic conditioning prior to surgery was verified as a method to improve the vascularization of the gastric conduit [4,10−16]. However, available meta-analyses have confirmed conflicting results because of search strategy discrepancies, methodological incongruities due to inclusion of single arm studies and natural heterogeneity of the studies. A lack of a randomized study confirming the benefit of gastric ischemic conditioning (GIC) is also noticeable. On the other hand, there is still no consensus on the optimal timing of GIC [11,12,16].

Hence, this study aimed to establish a rat gastric ischemic conditioning model, to observe histological changes following gastric ischemia, and to standardize the evaluation of selected immunocompetent cell counts. The use of a reproducible model is necessary for a future investigation of GIC effect on esophagogastric anastomosis viability. Recording histological changes following GIC is assumed as an important factor in predicting anastomosis failure. Furthermore, sophisticated understanding of gastric conduit ischemia from the point of view of histological changes observation and immunocompetent cell counts may be useful in optimal timing for esophagogastric anastomosis creation.

 

METHODS

Ethical statement:

The experiment was approved on May 17th 2021 by the Ethics Committee of the Faculty of Medicine of Pavol Jozef Šafárik University and by the State Veterinary and Food Administration of Slovakia (48935/2021-220).

 

Animal model:

Male Sprague-Dawley rats (n=24) weighing 500–600 g were obtained from the Animal Facility of P. J. Šafárik University and used for the experiment. The animals were individually housed in plexiglass cages under standard conditions (55±5% humidity, 22±2 °C, 12/12 h light-dark cycle) with free access to standard laboratory diet and tap water. All rats were fasted for 12 hours (with free access to water) before the experiment. The rats were randomly divided into four groups: three ischemic groups and one control group. The ischemic groups were divided according to the time when samples were collected for evaluation (I1H – ischemic group, samples were collected one hour after ischemia (n=6); I1D – ischemic group, samples were collected one day after ischemia (n=6); I7D – ischemic group, samples were collected seven days after ischemia (n=6)). A sham laparotomy with no vessel ligation was done in the control group (n=6), and samples were collected on the seventh day.

 

Surgical procedure:

All procedures were done under aseptic conditions and general anesthesia (Zoletil, Tiletamine, Zolazepam, Virbac, Westlake, Texas), 20 mg/kg i.m., and 5 mg/kg of mabron (Tramadoliumchlorid, Medochemie, Limassol, Cyprus). Each animal was placed in the supine position, and the operation field was prepared using standard methods (shaved abdomen cleaned with Kodan antiseptic tincture, Schülke, Norderstedt, Germany). An upper median laparotomy was performed, and after mobilization of the stomach, ligations of the left gastric artery (LGA) and short gastric vessels (SGV) were done using absorbable sutures (3-0 Polysorb, Covidien). In the control group, rats underwent only mobilization of the stomach without LGA occlusion (shown in Fig. 1). The laparotomy was closed using continuous sutures in anatomical layers (3-0 Polysorb suture Covidien) for the musculofascial layer and peritoneum, Chiraflon 5/0 running suture (Chirmax, Prague, Czech Republic) for skin closure. The animals were allowed free access to water after the operation, and standard laboratory diet was given on the second day after surgery. Postoperative analgesia was performed on the second day with mabron i.m. (2.5 mg/kg Tramadoliumchlorid, Medochemie, Limassol, Cyprus). The animals were euthanized according to the group by an overdose of anesthetics at one hour, one day, and seven days after surgery. Macroscopic photographs of the gastric mucosa were taken, and samples were stained for histological evaluation.

 

Macroscopic assay:

The distal part of the abdominal esophagus, gastroesophageal junction (GE), and the whole stomach were removed, and dissection was made on the greater curvature. Samples were cleaned using a sterile solution. Photographs of the mucosa were taken using an Olympus E330 digital camera equipped with a digital ED 50 mm f 2.0 macro-objective and a ring set flash SRF-11 (Olympus, Tokyo, Japan). Macroscopic signs of mucosal ischemia and necrosis were evaluated.

 

Histological Analysis:

A tissue sample was processed for light microscopy using routine techniques including fixation in 4% buffered formaldehyde, dehydration, embedding, cutting, and staining with hematoxylin-eosin. Two sections were prepared from the proximal stomach and GE junction of each sample. An experienced pathologist, who was blinded to the operative protocol, evaluated a section of the cardiac region and esophagogastric junction under a light microscope and performed microscopic analysis. Additionally, thirty microscopic fields per condition were randomly selected, and the number of immunocompetent cells was counted.

 

Statistical Analysis:

Descriptive statistics were expressed as mean ±SD (standard deviation). The data obtained from cell counting were compared using one-way ANOVA followed by the Student-Newman-Keuls test. A p-value of less than 0.05 was considered statistically significant.

1. Stomach mobilization
Stomach mobilization
Left gastric artery (LGA) is shown by the arrow.

 

RESULTS

Macroscopic Analysis:

The results of the macroscopic assay are shown in Fig. 2. Limited hyperemia was observed only in group I1D. No erosions, necrotic areas, or significant changes were observed in any of the experimental groups.

2. Macroscopic view of gastric mucosa after stomach dissection
Macroscopic view of gastric mucosa after stomach dissection
a − one hour after ischemization – group I1H; b − one day after ischemization – group I1D; c − seven days after ischemization – group I7D; d − control group; hyperemia of fundal mucosa is shown in b with no significant macroscopic changes between the groups.

Microscopic Analysis:

I1H ischemic group: The dominant finding in this group was passive hyperemia, ischemic denudation with mucosal erosion (Fig. 3), and capillary leukostasis. Eosinophils (45.37%) and lymphocytes (23.53%) were the dominant inflammatory cells. The total number of eosinophils was significantly higher (p<0.05) in groups I1H and I7D compared to group I1D. However, the total lymphocyte count was significantly lower in the I1H group (p<0.05) compared to the I1D and I7D groups (Graph 1).

I1D ischemic group: One day after ischemia with reparative changes and fibroblast proliferation, the fundal mucosa was reduced to one-third of the height compared to the nonischemic group. The inflammatory response in the epithelial-subepithelial junction was reduced, and vascular proliferation, mitotic activity, and partial regeneration of glands were increased at this time after vessel ligation (Fig. 4). The increase in the number of polymorphonuclear cells (23.15%) and histiocytes (16.59%) indicated a non-specific immune response and stimulation of reparation. The total count of polymorphonuclear cells was significantly higher (p<0.05) at this time of healing (group I1D) compared to group I7D, where the samples were collected seven days after ischemia. Furthermore, the number of histiocytes was significantly higher (p<0.05) in the I1D group compared to the I1H group (Graph 1). The percentage of eosinophils (31.56%) was reduced.

I7D ischemic group: Nearly total architectural regeneration of the fundal mucosa and capillary hyperplasia were documented at this time. Inflammatory response was reduced at this time after ischemization. The total count of polymorphonuclear cells (16.65%) was decreased (Fig. 5).

1. Total count of immunocompetent cells
Total count of immunocompetent cells
Cell counts were compared using one-way ANOVA followed by Student-Newman-Keuls test. P<0.05 was considered to indicate a statistically significant difference. The total lymphocyte count was significantly lower in I1H group compared to I1D and I7D groups. Also, the number of histiocytes was significantly lower in I1H and I7D groups compared to I1D group. On the other hand, the total count of eosinophils was significantly higher in I1H and I7D groups compared to I1D group. The total count of polymorphonuclears was significantly higher in I1D group compared to I7D group, but there were no significant differences between I1D and I1H groups. (I1H – one hour after ischemization, I1D – one day after ischemization, I7D – seven days after ischemization).

3. I1H group, HE, 40×
I1H group, HE, 40×
Brief histological picture of one-hour ischemia in the esophagogastric junction. Note the beginning ischemia and necrosis of glandular cardiac mucosa (star) and increase in inflammatory cells in lamina propria (arrow).

4. I1D group, HE, 40×
I1D group, HE, 40×
Brief histological picture of one day ischemia in the esophagogastric junction. Note the fundal mucosa high reduction (star) and subepithelial edema and neovascularization (arrow).

5. I7D group, HE, 40×
I7D group, HE, 40×
Brief histological picture of seven days ischemia in the esophagogastric junction. Note the inflammatory and reparative changes (arrow) and the almost complete mucosal recovery. Remarkable is the fundic glandular tubulization (star).

 

DISCUSSION

As far as we know, this is the first study to summarize both histological changes and inflammatory cell counts following gastric ischemia. Various animal models have been used to assess the changes that occur in the gastric remnant and subsequent esophagogastric anastomosis after partial gastric devascularization [7,9,13]. Measurements in animals have shown that gastric perfusion drops substantially immediately after vessel ligation, with continuous increase over time. Additional benefit in the conduit blood flow was shown when the short gastric arteries were ligated in addition to the left gastric artery [17]. In our study, we used laparotomy with stomach mobilization and ligation of the left gastric artery and short gastric vessels. Monitoring of histological changes, especially neovascularization and capillary hyperplasia, are essential for assessment of adequate tissue perfusion after gastric conduit creation. On the other hand, presence of necrosis and apoptosis in histological view are believed to be a negative factor for future anastomosis healing. Here we present nearly total histological recovery with regeneration of fundal mucosa, capillary hyperplasia, and absence of necrotic tissue 7 days after ischemia with a total count of polymorphonuclear cells (PMNL) significantly lower (p<0.05) compared to the 1 day after ischemia group.

Neutrophils/polymorphonuclear cells are the predominant leukocyte population in human blood and among the first cells recruited to an inflammatory site. During sterile tissue injuries, neutrophils are also essential to participate in the clearance of cellular debris, returning tissue to homeostasis [18,19]. In our study, acute inflammatory response was reduced in the group I7D, presented by a significantly lower total count of PMNL (p<0.05) at this time of healing (group I7D) compared to the group I1D. From this point of view the decreased number of PMNL 7days after GIC could be interpreted as a positive prognostic factor for future anastomosis healing.

Reavis et al. [20] ligated left, distal right, and short gastric vascular pedicles in an opossum model, resulting in an immediate decrease of gastric fundus blood flow by 73%. The distance of full thickness muscularis propria atrophy was 0.53 mm in the immediate group compared to 0.10 mm in the delay group (4 weeks after ischemia). In a study with Sprague-Dawley rats, Lamas et al. [7] verified higher rates of apoptosis and necrosis after 24 hours of partial gastric ischemia. Similar findings are reported in our present study with ischemic denudation and mucosal erosions as the dominant finding one hour after ischemia. Additionally, eosinophils (45.37%) were the major inflammatory cells at this time of healing, and the total count of lymphocytes was significantly lower compared to other groups. Histological changes in a mean of mucosal erosions and ischemic denudation presented one hour after ischemia belong to negative factors, increasing the complication rate of anastomosis healing.

Mingol-Navarro et al. [4] described the anatomic and physiopathologic background of ischemic preconditioning in their complex review. Kechagias et al. [21] analyzed all currently available information dealing with ischemic conditioning of the stomach in the prevention of esophagogastric anastomotic leakage after esophagectomy. Benefits of ischemic preconditioning were also described by several experimental and clinical studies [13−17]. Significantly increased neovascularity with decreased inflammation was observed by Perry et al. 30 days after gastric ischemic preconditioning in their animal study [13]. Therefore, we report an easily reproducible animal model of GIC, following histological changes in different time intervals from ischemia and measuring total counts of immunocompetent cells. Existence of this model is important for future esophagogastric anastomosis studies and to help optimize the time interval between GIC and anastomosis creation. On the one hand presence of neovascularization and capillary hyperplasia with architectonic regeneration of fundal mucosa 7 days after ischemia promote healing, on the other hand verifying mucosal alterations and denudation one hour after ischemia impairs the process of future anastomosis healing.

In this part, the optimal time interval between ischemic preconditioning and definitive surgery with esophagogastric anastomosis creation is discussed, with most studies showing waiting periods of approximately 2 weeks to be optimal [22−24]. However, there is still controversy surrounding this issue, as most clinical reports that evaluated the effectiveness of the technique did not implement rigid and precise protocols with a standardized interval [4]. Despite several studies and reviews published on the concept of GIC, there is still no agreement regarding its utility. Proponents of GIC have previously reported positive results on the anastomotic leak rate. On the other hand, there is still no consensus on the optimal timing of GIC [12]. The authors’ own study found that histological recovery after partial gastric ischemization was verified already 7 days after vessel ligation.

The authors note that Drescher et al. have pointed out that there were no existing rodent models of wound healing for esophagogastric anastomosis [25]. Drescher et al. unique rat model proved to be technically feasible to perform reconstruction by gastric tube formation and esophagogastric anastomosis.

The main limitation of our current presented study is the absence of verification of GIC effect on esophagogastric anastomosis vitality. On the other hand, the authors presented an easily reproducible animal model of GIC supplemented by verifying the presence of histological changes at different time points after ischemia, including changes in immunocompetent cell counts. Presence of neovascularization, a lack of mucosal erosions, capillary hyperplasia, and decreased number of acute inflammation phase cells (PMNL) 7 days after ischemia can be used as a positive prognostic factor in a process of optimal esophagogastric anastomosis timing. Based on these results, further investigation is needed to verify the effect of GIC on esophagogastric anastomosis vitality and to determine optimal timing between GIC and anastomosis creation.

 

Acknowledgments:

The authors would like to thank Milan Stebnický MD, PhD, Peter Varga MD and Michal Chyla MD, PhD, for their involvement in the surgical part.

 

Statement of ethics:

The experiment was approved on May 17th 2021 by the Ethics Committee of the Faculty of Medicine of Pavol Jozef Šafárik University and by the State Veterinary and Food Administration of Slovakia (4893-5/2021-220).

 

Conflict of interests

The authors declare that they have not conflict of interest in connection with this paper and that the article has not been published in any other journal, except congress abstracts and clinical guidelines.


Sources
  1. Bonavina L. Progress in the esophagogastric anastomosis and the challenges of minimally invasive thoracoscopic surgery. Ann Transl Med. 2021 May;9(10):907. doi: 10.21037/atm.2020.03.66.
  2. Deng J, Su Q, Ren Z, et al. Comparison of short-term outcomes between minimally invasive McKeown and Ivor Lewis esophagectomy for esophageal or junctional cancer: a systematic review and meta-analysis. Onco Targets Ther. 2018 Sep 20;11:6057−6069. doi: 10.2147/OTT. S169488.
  3. Chidi AP, Etchill EW, Ha JS, et al. Effect of thoracic versus cervical anastomosis on anastomotic leak among patients who undergo esophagectomy after neoadjuvant chemoradiation. J Thorac Cardiovasc Surg. 2020 Oct;160(4):1088−1095. doi: 10.1016/j.jtcvs.2020.01.089. Epub 2020 Feb 20.
  4. Mingol-Navarro F, Ballester-Pla N, Jimenez-Rosellon R. Ischaemic conditioning of the stomach previous to esophageal surgery. J Thorac Dis. 2019 Apr;11(Suppl 5):S663−S674. doi: 10.21037/jtd. 2019.01.43.
  5. Thomas DM, Langford RM, Russell RC, et al. The anatomical basis for gastric mobilization in total oesophagectomy. Br J Surg. 1979 Apr;66(4):230−233. doi: 10.1002/bjs.1800660404. PMID: 454988.
  6. Liebermann-Meffert DM, Meier R, Siewert JR. Vascular anatomy of the gastric tube used for esophageal reconstruction. Ann Thorac Surg. 1992 Dec;54(6):1110−1115. doi: 10.1016/0003-4975(92)90077-h. PMID: 1449294.
  7. Lamas S, Azuara D, de Oca J, et al. Time course of necrosis/apoptosis and neovascularization during experimental gastric conditioning. Dis Esophagus. 2008;21(4):370−376. doi: 10.1111/j.1442-2050.2007.00772.x.
  8. Procházka V, Grolich T, Čan V, et al. Results of minimally invasive esophagectomy for esophageal cancer performed after ischemic gastric conditioning. Rozhl Chir. 2018 Winter;97(7):335-341. English.
  9. Kitano M, Bernsand M, Kishimoto Y, et al. Ischemia of rat stomach mobilizes ECL cell histamine. Am J Physiol Gastrointest Liver Physiol. 2005 May;288(5):G1084−1090. doi: 10.1152/ajpgi.00004.2004. Epub 2005 Jan 20.
  10. Bludau M, Hölscher AH, Vallböhmer D, et al. Ischemic conditioning of the gastric conduit prior to esophagectomy improves mucosal oxygen saturation. Ann Thorac Surg. 2010 Oct;90(4): 1121−1126. doi: 10.1016/j.athoracsur.2010.06.003.
  11. Michalinos A, Antoniou SA, Ntourakis D, et al. Gastric ischemic preconditioning may reduce the incidence and severity of anastomotic leakage after οesophagectomy: a systematic review and meta-analysis. Dis Esophagus 2020 Oct 12;33(10):doaa010. doi: 10.1093/dote/ doaa010.
  12. Jogiat UM, Sun WYL, Dang JT, et al. Gastric ischemic conditioning prior to esophagectomy reduces anastomotic leaks and strictures: a systematic review and meta-analysis. Surg Endosc. 2022 Jul;36(7):5398−5407. doi: 10.1007/ s00464-021-08866-4. Epub 2021 Nov 15.
  13. Perry KA, Banarjee A, Liu J, et al. Gastric ischemic conditioning increases neovascularization and reduces inflammation and fibrosis during gastroesophageal anastomotic healing. Surg Endosc. 2013 Mar;27(3):753−760. doi: 10.1007/s00464012-2535-6. Epub 2012 Dec 18.
  14. Prudius V, Procházka V, Pavlovský Z, et al. Neovascularization after ischemic conditioning of the stomach and the influence of follow-up neoadjuvant chemotherapy thereon. Wideochir Inne Tech Maloinwazyjne 2018 Sep;13(3):299−305. doi: 10.5114/wiitm.2018.75907. Epub 2018 May 22.
  15. Prochazka V, Marek F, Kunovsky L, et al. Comparison of cervical anastomotic leak and stenosis after oesophagectomy for carcinoma according to the interval of the stomach ischaemic conditioning. Ann R Coll Surg Engl. 2018 Sep;100(7):509−514. doi: 10.1308/rcsann.2018.0066. Epub 2018 Jun 18.
  16. Aiolfi A, Bona D, Bonitta G, et al. Gastric Ischemic Conditioning (GIC) International Collaborative Group. Effect of gastric ischemic conditioning prior to esophagectomy: systematic review and meta-analysis. Updates Surg. 2023 Sep;75(6):1633−1643. doi: 10.1007/ s13304-023-01601-9. Epub 2023 Jul 27.
  1. Beck SM, Malay MB, Gagné DJ, et al. Experimental model of laparoscopic gastric ischemic preconditioning prior to transhiatal esophagectomy. Surg Endosc. 2011 Aug;25(8):2470−2477. doi: 10.1007/ s00464-010-1568-y. Epub 2011 Feb 8.
  2. Wang J. Neutrophils in tissue injury and repair. Cell Tissue Res. 2018 Mar; 371(3):531−539. doi: 10.1007/s00441-017-2785-7. Epub 2018 Jan 30.
  3. Zahorec R. Neutrophil-to-lymphocyte ratio, past, present and future perspectives. Bratisl Lek Listy 2021;122(7):474−488. doi: 10.4149/BLL_2021_078.
  4. Reavis KM, Chang EY, Hunter JG, et al. Utilization of the delay phenomenon improves blood flow and reduces collagen deposition in esophagogastric anastomoses. Ann Surg. 2005 May;241(5):73645; discussion 745−747. doi: 10.1097/01. sla.0000160704.50657.32.
  5. Kechagias A, van Rossum PSN, Ruurda JP, et al. Ischemic conditioning of the stomach in the prevention of esophagogastric anastomotic leakage after esophagectomy. Ann Thorac Surg. 2016 Apr;101(4):1614−1623.  doi:  10.1016/j.athoracsur.2015.10.034. Epub 2016 Feb 5. PMID: 26857639.
  6. Berrisford RG, Veeramootoo D, Parameswaran R, et al. Laparoscopic ischaemic conditioning of the stomach may reduce gastric-conduit morbidity following total minimally invasive oesophagectomy. Eur J Cardiothorac Surg. 2009 Nov;36(5):888−893; discussion 893. doi: 10.1016/j.ejcts.2009.01.055. Epub 2009 Jul 16.
  7. Diana M, Hübner M, Vuilleumier H, et al. Redistribution of gastric blood flow by embolization of gastric arteries before esophagectomy. Ann Thorac Surg. 2011 May;91(5):1546−1551. doi: 10.1016/j.atho-racsur.2011.01.081. Epub 2011 Mar 21.
  8. Farran L, Miro M, Alba E, et al. Preoperative gastric conditioning in cervical gastroplasty. Dis Esophagus. 2011 May;24(4):205−210. doi: 10.1111/j.14422050.2010.01115.x. Epub 2010 Oct 11.
  9. Drescher DG, Vogt J, Gabriel M, et al. Model of wound healing for esophagogastric anastomoses in rats. Eur Surg Res. 2012;48(4):194−199. doi: 10.1159/ 000338625. Epub 2012 Jun 7.

doc. MUDr. Ivan Kováč, PhD.
2
nd Department of Surgery,
Louise Pasteur University Hospital and P. J. Šafárik University
Rastislavova 43, 04011 Košice, Slovak Republic
e-mail:
ivankovac.kovi@gmail.com
ORCID: 0000-0001-6717-5818

Labels
Surgery Orthopaedics Trauma surgery
Topics Journals
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#