Skip to main content
  • Systematic Review
  • Open access
  • Published:

Risk factors for esophageal anastomotic stricture after esophagectomy: a meta-analysis

Abstract

Background

The aim of this study was to assess the risk factors for anastomotic stricture in esophageal cancer patients undergoing esophagectomy. Esophageal anastomotic stricture is the most common long-term complication for esophagectomy. The risk factors for esophageal anastomotic stricture still remain controversial.

Methods

MEDLINE, Cochrane Library, and EMBASE were searched to identify observational studies reporting the risk factors for esophageal anastomotic stricture after esophagectomy. A meta-analysis was conducted to investigate the impact of various risk factors on esophageal anastomotic stricture. The GRADE [Grading of Recommendations Assessment, Development and Evaluation] approach was used for quality assessment of evidence on outcome levels.

Results

This review included 14 studies evaluating 5987 patients.The meta-analysis found that anastomotic leakage (odds ratio [OR]: 2.75; 95% confidence interval[CI]:2.16–3.49), cardiovascular disease [OR:1.62; 95% CI: 1.22–2.16],diabete [OR: 1.62; 95% CI: 1.20–2.19] may be risk factors for esophageal anastomotic stricture.There were no association between neoadjuvant therapy [OR: 0.78; 95% CI:0.62–0.97], wide gastric conduit [OR:0.98; 95% CI: 0.37–2.56],mechanical anastomosis [OR: 0.84; 95% CI:0.47–1.48],colonic interposition[OR:0.20; 95% CI: 0.12–0.35],and transhiatal approach[OR:1.16; 95% CI:0.81–1.64],with the risk of esophageal anastomotic stricture.

Conclusions

This meta-analysis provides some evidence that anastomotic leakage,cardiovascular disease and diabete may be associated with higher rates of esophageal anastomotic stricture.Knowledge about those risk factors may influence treatment and procedure-related decisions,and possibly reduce the anastomotic stricture rate.

Peer Review reports

Introduction

Esophageal cancer is the eighth most common malignancy in the world, with more than 570 000 new cases diagnosed each year [1]. In China, the incidence of esophageal cancer is the highest in the world, especially in the case of esophageal squamous cell carcinoma, which has been steadily increasing in recent decades [2]. China accounts for more than half of the total number of new cases of esophageal cancer in the world each year. The main treatment for esophageal cancer remains esophagectomy, with a five-year survival rate of around 50% [3]. Despite many improvements in treatment and perioperative care, esophagectomy is still associated with relatively high morbidity and mortality.

Complications of anastomosis are common with esophageal anastomotic stricture being one of the recognised complications affecting 0.5% to 42% of patients following esophageal anastomosis [4,5,6,7,8]. The wide range of reported incidence may be due to the different definition of stricture [4, 5, 7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29]. In this study, anastomotic stricture was defined as follows. Tumor recurrence at the anastomosis site was excluded after esophagectomy. The diameter of the stricture was less than 1 cm under endoscopy, or the conventional type of endoscope (about 1 cm in diameter) could not pass through, accompanied by different degrees of dysphagia.

Patients with esophageal anastomotic stricture had a significantly higher recurrence rate than patients without esophageal anastomotic stricture [30]. Therefore, esophageal anastomotic stricture should be evaluated separately from other complications. The risk factors for esophageal anastomotic stricture still remain controversial. Some of the risk factors associated with a higher incidence of esophageal anastomotic stricture include: Anastomotic leakage [4, 7,8,9, 12, 13], neoadjuvant therapy [4, 7,8,9], cardiovascular disease [4, 7,8,9, 13], diabete [12], colonic interposition [4, 31], wide gastric conduit [10, 12], mechanical anastomosis [11], female and distal location of strictures [30], American society of Aneshesiologists (ASA) grade, cervical anastomosis and transhiatal resections [13]. However, other studies have found that some of these factors are not associated with esophageal anastomotic stricture [7, 8, 31,32,33]. Therefore, it is important to identify the risk factors for esophageal anastomotic stricture to determine appropriate treatment strategies. Knowledge of these risk factors may help to tailor treatment to each individual patient.

Methods

Search strategy

The MEDLINE and EMBASE and Cochrane Library databases were searched [from inception to June 20, 2024].For MEDLINE,eligible trials used the followed medical subject heading (MeSH) terms and search formula:"anastomosis, surgical" [MeSH Terms] AND (stricture [tw] OR stenosis [tw] OR “benign strictures” [tw]) AND esophagectomy [MeSH Terms] AND "Risk Factors" [Title]. The searches were limited to articles published in English. Individually fitted search strategies with similar search terms were also performed in the EMBASE.Only published journal studies were included; unpublished data were not sought.The ‘related articles’ function from MEDLINE was used to broaden the search, and reference lists of included studies were searched for additional relevant studies. Manual searching of reference lists then identified further potentially relevant studies.For the study selection process, see Fig. 1.

Fig. 1
figure 1

The study selection process

Inclusion criteria

The inclusion criteria for the selected studies as follow.

  1. 1.

    Studies had to report risk factors for esophageal anastomotic stricture in patients who underwent esophagectomy.

  2. 2.

    Anastomotic stricture must have met postoperative dysphagia requiring endoscopic dilatation [34, 35].

  3. 3.

    Anastomotic stricture was benign rather than tumor recurrence.

  4. 4.

    Multivariate regression analysis had to be used for the analysis of risk factors to reduce the risk of confounding in observational studies.

Quality assessment

The Newcastle–Ottawa scale [36] was used to assess the quality of the studies included. Studies that received seven stars or more were considered to be of higher quality.

The GRADE [Grading of Recommendations Assessment, Development and Evaluation] method was used to assess the quality of evidence at the level of the analytical results [37]. Quality can be rated as high, moderate, low or very low. The GRADE assessment was carried out using GRADEpro software, version 3.6.1 for Windows.

Data extraction

The following data were extracted from all eligible studies: first author, year of publication, country of origin, study design, number of subjects and risk factors associated with esophageal anastomotic stricture. For risk factors, the focus was on factors previously identified as independent factors, including: anastomotic leakage, cardiovascular disease, diabetes, neoadjuvant therapy, mechanical anastomosis, wide gastric conduit, colonic interposition and transhiatal approach.

Statistical analysis

The meta-analysis was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and meta-analysis [PRISMA] statement [38]. The odds ratio [OR] was used as a statistical indicator of dichotomous results. ORs were calculated from the original data and presented with 95% confidence intervals [CIs]. For the effect of each variable on the incidence of esophageal anastomotic stricture, the combined odds ratios were calculated. Pooled outcome measures were determined using random-effects models as described by Der Simonian and Laird [39].

Heterogeneity was quantified using I [2]. Slight heterogeneity can account for less than 25% of the variance in point estimates, while significant heterogeneity can account for more than 50% [40].

Statistical analysis was performed using STATA software, version 12.0 [Stata Corporation, College Station, TX, USA].

Results

Search results

Our pre-defined search strategy identified 391 studies.Duplicate studies were excluded. Studies that were not available in full text and original data were also excluded. The remaining 25 studies were searched for full-text articles. Three reviews and three case reports were excluded. In total, 19 studies were identified for further analysis. Five of these studies did not meet the inclusion criteria and were excluded from further analysis. Thus, 14 studies that met the inclusion criteria were included in the final meta-analysis. A total of 5987 patients were included. And the overall population was 1571 patients with anastomotic stricture. The overall proportion of anastomotic stenosis in the total study population was 26.2%.

Quality assessment

The included studies were assessed for risk of bias using the Newcastle–Ottawa scale. The mean score for all studies was 7.6 [range: 7–8]. For most studies, the risk of bias was low and the quality of outcome assessment was good (Table 1).

Table 1 Characteristics of the 14 studies included in the meta-analysis to assess the risk of esophageal anastomotic stricture after esophagectomy

The GRADE method was used to assess the quality of the evidence at the level of the analysis results. The quality of the evidence was assessed as high, medium or low for these risk factors (Table 2).

Table 2 Summary finding of risk factors eligible for meta-analysis

Risk factors for esophageal anastomotic stricture

Anastomotic leakage

Total 11 studies [4, 5, 7, 8, 10, 11, 13,14,15,16, 29] [n = 5257] assessed anastomotic leakage as a risk factor. A meta-analysis of these studies using a random-effects model found an overall OR of 2.75 [95% CI: 2.16–3.49], indicating that anastomotic leakage was associated with a high risk of esophageal anastomotic stricture (Fig. 2a). Heterogeneity between studies was mild [p = 0.202; I2 = 25.4%] and using funnel plots and Egger's test [p = 0.408], there was no evidence of significant publication bias. Using the GRADE method, the quality of the evidence was judged to be high.

Fig. 2
figure 2

Forest plots for risk factor eligible for meta-analysis. a Anastomotic leakage. b Neoadjuvant therapy. c Cardiovascular disease. d Diabete. e Wide gastric conduit. f Mechanical anastomosis. g Colonic interposition. h Transhiatal approach

Neoadjuvant treatment

Seven studies [4, 5, 8, 10, 11, 14, 15] [n = 3507] of these trials found an overall OR of 0.78 [95% CI: 0.62–0.97] in a random-effects model, suggesting that neoadjuvant treatment did not increase the risk of esophageal anastomotic stricture (Fig. 2b). Heterogeneity between studies was mild [p = 0.218; I2 = 27.6%] and using funnel plots and Egger's test [p = 0.665], there was no evidence of significant publication bias. Using the GRADE approach, the quality of the evidence was assessed as moderate.

Cardiovascular diseases

Seven studies [4, 5, 7, 8, 10, 11, 14] [n = 2362] assessed cardiovascular disease as a risk factor. A meta-analysis of these studies using a random-effects model found an overall OR of 1.62 [95% CI: 1.22–2.16], indicating that cardiovascular disease was associated with the risk of esophageal anastomotic stricture (Fig. 2c). Heterogeneity between studies was mild [p = 0.185; I2 = 31.8%] and no significant evidence of publication bias was found using the funnel plot and Egger's test [p = 0.822]. The quality of the evidence was assessed as high using the GRADE method.

Diabete

Seventh studies [4, 5, 10, 11, 14, 15, 29] [n = 3827] assessed diabete as a risk factor. In meta-analysis of these studies using a random-effects model, an overall OR of 1.62 [95% CI: 1.20–2.19] was found, suggesting that diabetes was a risk factor for esophageal anastomotic stricture (Fig. 2d). Heterogeneity between studies was low [p = 0.329; I2 = 13.2%], and there was no significant evidence of publication bias using the funnel plot and Egger's test [p = 0.829]. The quality of the evidence was rated high using the GRADE approach.

Wide gastric conduit

Only four studies [10, 12, 14, 15] [n = 2367] evaluated wide gastric conduit as a risk factor. A meta-analysis of these studies using a random-effects model found an overall OR of 0.98 [95% CI: 0.37–2.56], indicating that wide gastric conduit was not a risk factor for esophageal anastomotic stricture (Fig. 2e). However, the heterogeneity was very high [p = 0.000; I2 = 94.9%]. The results were not reliable. The quality of the evidence was regarded as low based on the GRADE approach.

Mechanical anastomosis

Four studies [4, 7, 15, 29] [n = 2756] evaluated the effect of anastomosis techniques. A meta-analysis of these studies using a random-effects model showed an overall OR of 0.84 [95% CI: 0.47–1.48], indicating that mechanical anastomosis was not a risk factor for esophageal anastomotic stricture (Fig. 2f). Heterogeneity was, however, very high [p = 0.002; I2 = 80.5%]. The results were not reliable. Based on the GRADE approach, the quality of the evidence was assessed as low.

Colonic interposition

Colonic interposition was assessed as a risk factor in only three studies [4, 5, 16] [n = 1183]. In meta-analysis of these studies using a random-effects model, an overall OR of 0.20 [95% CI: 0.12–0.35] was found, suggesting that colonic interposition was not a risk factor for esophageal anastomotic stricture (Fig. 2g). There was no heterogeneity between studies [p = 0.802; I2 = 0.0%], and no significant evidence of publication bias was found using funnel plot and Egger's test [p = 0.401]. The GRADE approach was used to assess the quality of the evidence as high.

Transhiatal approach

Only three studies [4, 10, 11] [n = 1525] evaluated transhiatal approach as a risk factor. A meta-analysis of these studies using a random effects model found an overall OR of 1.16 [95% CI: 0.81–1.64], suggesting that transhiatal approach was not a risk factor for stricture of the esophageal anastomosis (Fig. 2h). Study heterogeneity was low [p = 0.293; I2 = 18.6%] and there was no significant evidence of publication bias by funnel plot and Egger's test [p = 0.856]. The quality of evidence was assessed as high using the GRADE method.

Discussion

This is the first meta-analysis of observational studies designed to assess risk factors for esophageal anastomotic stricture after esophagectomy. The meta-analysis showed that anastomotic leakage, cardiovascular disease and diabete are associated with esophageal anastomotic stricture. No association was found between neoadjuvant therapy, wide gastric conduit, mechanical anastomosis, colonic interposition and transhiatal approach with the risk of esophageal anastomotic stricture. Based on the results of the meta-analysis of GRADE approaches, the quality of the evidence was assessed as low to high. The studies included in the meta-analysis were judged by the NOS to be of good quality for the assessment of outcomes. Thus, the low quality of the results was not due to the bias of the studies, but mainly to the observational nature of the studies, which was initially rated as low by GRADE.

In this meta-analysis, anastomotic leakage is an important risk factor for anastomotic stricture. The transition of anastomotic leakage to a later benign stricture, although not fully investigated, may be associated with concomitant fibrosis leading to scar formation and subsequent stricture formation. This secondary healing process results in strictures that are more likely to be refractory [11]. Low oxygen supply and poor perfusion in the anastomosis, which are likely to be the cause of leakage, also lead directly to the formation of anastomotic strictures [4]. The local infection caused by anastomotic leakage, as well as repetitive trauma and stimulus, can promote fibrous tissue proliferation and scar formation, leading to gradual narrowing of the anastomosis [11, 41, 42].

In this meta-analysis, neoadjuvant treatment did not increase the risk of anastomotic stricture. Interestingly, patients who received neoadjuvant chemotherapy were less likely to develop stricture. Prior chemotherapy reduced the likelihood of anastomotic stricture by almost 70% [30]. One hypothesis is that the phenomenon of a low inflammatory response to foreign bodies, based on the patient's compromised immune system, may lead to a lower incidence of anastomotic strictures due to the foreign body response [30, 43]. Another hypothesis is that chemotherapy reduces the postoperative inflammatory response (fibrosis and intimal hyperplasia) leading to luminal stricturing [30, 44,45,46].

The meta-analysis found that a history of cardiovascular disease is an important risk factor for anastomotic stricture [7, 33]. A leakage at the anastomosis site can lead to local ischemia and hypoxia, which can progress to anastomotic stenosis. Similarly, postoperative gastric tube anastomosis circulate has already been damaged, and patients with a history of cardiovascular disease will exacerbate this insufficient circulate and further worsen the condition [47]. Although significant atherosclerosis is rarely observed in the right gastroepiploic artery, it is possible that some degree of luminal stenosis may be sufficient to cause conduit ischaemia if accompanied by impaired oxygen exchange or any cardiovascular or hemodynamic factors affecting perfusion [48]. Taken together, these findings suggest that arterial atherosclerosis or microvascular stenosis may be sufficient to cause conduction hypoperfusion in the presence of oxygen exchange disturbances or cardiovascular or hemodynamic factors affecting systemic circulation.

Diabete has also been shown to increase the incidence of anastomotic stricture after esophagectomy. Its aetiology is similar to that of cardiovascular disease. Diabete can lead to atherosclerosis and microcirculatory disorders. Blood supply to the proximal part of the gastric conduit depends on intragastric collateral flow and microvascular perfusion. If diabete causes hyalinosis and microcirculatory disturbances, ischaemia of this part of the duct may occur [31, 49]. Therefore, it is important to monitor the patient's blood glucose levels to maintain uninterrupted microcirculation. Recent studies suggest that anastomosis reconstruction 4–5 days after vascular dissection may promote neovascularisation and prevent anastomosis-related complications [31, 50, 51].

It is not clear from the meta-analysis what the effect of wide gastric conduit is on anastomotic stricture. In some studies, an extensive gastric conduit was associated with high risk [14, 15, 52,53,54], but not in others [10, 11, 55,56,57,58]. In contrast to the wide gastric conduit, the diameter of the narrow band is similar to the diameter of the natural esophagus. The narrow conduit allowed the greater curvature to be extended as much as possible, which reduces the strain of the anastomosis and allows the anastomosis to be performed close to an area where the blood supply from the right gastroepiploic artery is intact [15].

Mechanical anastomosis instead of hand-sewn anastomosis accelerating anastomotic strictures are not well-defined. A common endoscopic finding in patients with esophagogastric anastomosis is the presence of protruding sutures and/or staples in the esophageal lumen. The presence of a foreign body may promote inflammation and scarring, which complicates treatment [44]. Differences in the incidence of strictures between manually sutured and stapled anastomoses may be due to inadequate attachment of the mucosa to the staples [59].

In this meta-analysis, colonic interposition did not increase the risk of stricture. Anastomotic stricture are more frequent after gastric reconstruction. The blood supply to the colon used for colonic interposition is completely vascularized by arteries, which may explain the relatively large difference in the incidence of stricture [4, 32, 60]. There's another explanation that gastric pull-up suggests that reflux of gastric juices may contribute to stricture formation [16]. The observation that late stricture (more than 1 year) occurred only in patients with gastric pull-up additionally supports a reflux etiology.

The transhiatal approach was not an independent risk factor for anastomotic stricture. During transhiatal esophagectomy, the esophagus was removed directly through an extended diaphragmatic hiatus in a straight line of sight to the lower pulmonary vein. After mobilization of the right or left esophagus, the intrathoracic esophagus was squatted proximally to distally using a vein cutter [4]. The transthoracic approach was similar to the transhiatal approach, as was the creation of a 3 to 4 cm wide gastric tube. In a meta-analysis, the transhiatal approach was not statistically associated with the number of anastomotic complications [49, 50, 61].

The present meta-analysis has some limitations: the risk factor for esophageal anastomotic stricture can only be examined in observational studies, which poses a risk of bias that cannot be eliminated by a tailored analysis. Observational studies were considered to be of lower quality than randomised controlled trials, so the conclusions we present are based on studies of lower quality. The included studies differed in terms of risk factors for disease severity, location and duration, surgical indication and type of surgical procedure. We therefore used random-effects models in the meta-analysis, which provided more conservative conclusions in the presence of heterogeneity. For observational studies, we sought to reduce the risk of confounding factors by selecting studies using multivariate regression. However, without including all known and unknown confounders, this risk cannot be completely eliminated.

In conclusion, the prevalence of anastomotic leakage, cardiovascular disease and diabetes was associated with a higher incidence of esophageal anastomotic stricture in patients undergoing esophagectomy. This may help surgeons to better decide on appropriate treatment strategies for individual patients.

Availability of data and materials

Data is provided within the manuscript or supplementary information files.

References

  1. Arnold M, Ferlay J, Van Berge Henegouwen MI, et al. Global burden of oesophageal and gastric cancer by histology and subsite in 2018. Gut. 2020;69:1564–71.

    Article  PubMed  Google Scholar 

  2. Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108.

    Article  PubMed  Google Scholar 

  3. Tachimori Y, Ozawa S, Numaski H, et al. Comprehensive registry of esophageal cancer in Japan, 2010. Esophagus. 2017;14(3):189–214.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Mark van Heijl M, Gooszen JA, Fockens P, et al. Risk factors for development of benign cervical strictures after esophagectomy. Ann Surg 2010;251:1064–69.

  5. Tanaka K, Makino T, Yamasaki M, et al. An analysis of the risk factors of anastomotic stricture after esophagectomy. Surg Today. 2018;48:449–54.

    Article  CAS  PubMed  Google Scholar 

  6. Heitmiller RF, Fischer A, Liddicoat JR. Cervical esophagogastric anastomosis: results following esophagectomy for carcinoma. Dis Esophagus. 1999;12:264–9.

    Article  CAS  PubMed  Google Scholar 

  7. Honkoop P, Siersema PD, Tilanus HW, et al. Benign anastomotic strictures after transhiatal esophagectomy and cervical esophagogastrostomy: risk factors and management. Thorac Cardiovasc Surg. 1996;111:1141–6.

    Article  CAS  Google Scholar 

  8. Sutcliffe RP, Forshaw MJ, Tandon R, et al. Anastomotic strictures and delayed gastric emptying after esophagectomy: incidence, risk factors and management. Dis Esophagus. 2008;21:712–7.

    Article  CAS  PubMed  Google Scholar 

  9. Dewar L, Gelfand G, Finley RJ, et al. Factors affecting cervical anastomotic leak and stricture formation following esophagogastrectomy and gastric tube interposition. Am J Surg. 1992;163:484–9.

    Article  CAS  PubMed  Google Scholar 

  10. Renol M Koshy, Joshua M Brown, Jakub Chmelo, et al. Anastomotic stricture after Ivor Lewis esophagectomy:An evaluation of incidence, risk factors, and treatment. Surgery. 2022;171(2):393–8.

  11. Nishikawa Katsunori, Fujita Tetsuji, Yuda Masami, et al. Early prediction of complex benign anastomotic stricture after esophagectomy using early postoperative endoscopic fndings. Surg Endosc. 2020;34(8):3460–9.

    Article  PubMed  Google Scholar 

  12. Hosoi Takahiro, Abe Tetsuya, Uemura Norihisa, et al. The impact of circular stapler size on the incidence of cervical anastomotic stricture after esophagectomy. World J Surg. 2019;43(7):1746–55.

    Article  PubMed  Google Scholar 

  13. Ahmed Zuhair, Elliott Jessie A, King Sinead, et al. Risk factors for anastomotic stricture post-esophagectomy with a standardized sutured anastomosis. World J Surg. 2017;41(2):487–97.

    Article  PubMed  Google Scholar 

  14. Dongshan Zhu, Jian-wei Cao, Ming-fei Geng, et al. Wide gastric conduit increases the risk of benign anastomotic stricture after esophagectomy. American Surgeon. 2020;86(6):621–7.

  15. Jiang Haoyao, Hua Rong, Sun Yifeng, et al. Risk factors for anastomotic complications after radical Mckeown esophagectomy. Ann Thorac Surg. 2021;112(3):944–51.

    Article  PubMed  Google Scholar 

  16. John W Briel, Anand P Tamhankar, Jeffrey A Hagen, et al. Prevalence and risk factors for ischemia, leak, and stricture of esophageal anastomosis:gastric pull-up versus colon interposition. J Am Coll Surg. 2004;198(4):536–41; discussion 541–2.

  17. Fok M, Wong J. Oesophageal cancer treatment: curative modalities. Eur J Gastroenterol Hepatol. 1994;6:676–83.

    Article  Google Scholar 

  18. McManus KG, Ritchie AJ, McGuigan J, et al. Sutures, staplers, leaks and strictures:a review of anastomoses in oesophageal resection at Royal Victoria Hospital, Belfast 1977–1986. Eur J Cardiothorac Surg. 1990;4:97–100.

    Article  CAS  PubMed  Google Scholar 

  19. Tilanus HW, Langenhorst BLAM. Morbidity and mortality after oesophageal resection without thoracotomy. Gullet. 1991;1:122–5.

    Google Scholar 

  20. Griffin SM, Woods SDS, Chan A, et al. Earlyand late surgical complications of subtotal oesophagectomy for squamous carcinoma of the oesophagus. J R Coll Surg Edinb. 1991;36:170–3.

    CAS  PubMed  Google Scholar 

  21. Fok M, Ah-Chong AK, Chenh SWK, et al. Comparison of a single layer continuous hand-sewn method and circular stapling in 580 oesophageal anastomoses. Br J Surg. 1991;78:342–5.

    Article  CAS  PubMed  Google Scholar 

  22. Wang LS, Huang MH, Huang BS, et al. Gastric substitution for resectable carcinoma of the esophagus:an analysis of 368 cases. Ann Thorac Surg. 1992;53:289–94.

    Article  CAS  PubMed  Google Scholar 

  23. Lam TCF, Fok M, Cheng SWK, et al. Anastomotic complications after esophagectomy for cancer: acomparison of neck and chest anastomoses. J Thorac Cardiovasc Surg. 1992;104:395–400.

    Article  CAS  PubMed  Google Scholar 

  24. Pierie JPEN, de Graaf PW, Poen H, et al. Incidence and management of benign anastomotic stricture after cervical oesophagogastrostomy. Br J Surg. 1993;80:471–4.

    Article  CAS  PubMed  Google Scholar 

  25. Zieren HU, Mtiller JM, Pichhnaler H. Prospective randomized study of one or two layer anastomosis following oesophageal resection and cervical oesophagogastrostomy. Br J Surg. 1993;80:608–11.

    Article  CAS  PubMed  Google Scholar 

  26. Bardini R, Bonavina L, Asolati M, et al. Single-layered cervical esophageal anastomoses:a prospective study of two suturing techniques. Ann Thorac Surg. 1994;58:1087–90.

    Article  CAS  PubMed  Google Scholar 

  27. Leonie Haverkamp, Pieter C van der Sluis, Roy J J Verhage, et al. End to end cervical esophagogastric anastomoses are associated with a higher number of strictures compared with end-to-side anastomoses. Gastrointest Surg. 2013;17(5):872–6.

  28. Tyler Robert, Nair Amit, Lau Meagan, et al. Incidence of anastomotic stricture after Ivor-Lewis oesophagectomy using a circular stapling device. World J Gastrointest Surg. 2019;11(11):407–13.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Yimin Gu, Yang Yushang, Shang Qixin, et al. Risk factors for benign anastomotic stricture post-oesophagectomy: single centre analysis of 702 oesophagectomies with squamous cell carcinoma. Transl Cancer Res. 2019;8(3):828–35.

    Article  Google Scholar 

  30. Mendelson Aaron H, Small Aaron J, Agarwalla Anant, et al. Esophageal anastomotic strictures:outcomes of endoscopic dilation, risk of recurrence and refractory stenosis, and effect of foreign body removal. Clin Gastroenterol Hepatol. 2015;13:263–71.

    Article  PubMed  Google Scholar 

  31. Briel JW, Tamhankar AP, Hagen JA, et al. Prevalence and risk factors for ischemia, leak, and stricture of esophageal anastomosis: gastric pull-up versus colon interposition. J Am Coll Surg. 2004;198:536–41.

    Article  PubMed  Google Scholar 

  32. Urschel JD, Blewett CJ, Bennett WF, et al. Handsewn or stapled esophagogastric anastomoses after esophagectomy for cancer:meta-analyses of randomized controlled trials. Dis Esophagus. 2001;14:212–7.

    Article  CAS  PubMed  Google Scholar 

  33. Petrin G, Ruol A, Battaglia G, et al. Anastomotic stenoses occurring after circular stapling in esophageal cancer surgery. Surg Endosc. 2000;14:670–4.

    Article  CAS  PubMed  Google Scholar 

  34. Low DE, Kuppusamy MK, Alderson D, et al. Benchmarking complications associated with esophagectomy. Ann Surg. 2019;269:291–8.

    Article  PubMed  Google Scholar 

  35. Dresner SM, Lamb PJ, Wayman J, et al. Benign anastomotic stricture following transthoracic subtotal oesophagectomy and stapled oesophagogastrostomy:risk factors and management (Abstract only). Br J Surg. 2000;87:370–1.

    Article  Google Scholar 

  36. Wells G, Shea B, O’Connell D, et al. The Newcastle-Ottawa Scale [NOS] for assessing the quality of nonrandomised studies in meta-analyses. Ottawa: Ottawa Hospital Research Institute; 2013.

    Google Scholar 

  37. Balshem H, Helfand M, Schunemann HJ, et al. GRADE guidelines: 3. Rating the quality of evidence. J Clin Epidemiol. 2011; 64:401–6.

  38. Liberati A, Altman DG, Tetzlaff J, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions:explanation and elaboration. BMJ. 2009;339:b2700.

    Article  PubMed  PubMed Central  Google Scholar 

  39. DerSimonian R, Laird N. meta-analyses in clinical trials. Control Clin Trials. 1986;7:177–88.

    Article  CAS  PubMed  Google Scholar 

  40. Higgins JP, Thompson SG. Quantifying heterogeneity in a meta-analyses. Stat Med. 2002;21:1539–58.

    Article  PubMed  Google Scholar 

  41. Cassivi SD. Leaks, strictures, and necrosis:a review of anastomotic complications following esophagectomy. Semin Thorac Cardiovasc Surg. 2004;16:124–32.

    Article  PubMed  Google Scholar 

  42. Urschel JD. Esophagogastric anastomotic leaks:the importance of gastricischemia and therapeutic applications of gastric conditioning. J Invest Surg. 1998;11:245–50.

    Article  CAS  PubMed  Google Scholar 

  43. Hachim D, Wang N, Lopresti ST, et al. Effects of aging upon the host response to implants. J Biomed Mater Res A. 2017;105:1281–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lew RJ, Kochman ML. A review of endoscopic methods of esophageal dilatation. J Clin Gastroenterol. 2002;35:117–26.

    Article  PubMed  Google Scholar 

  45. Kochhar R, Ray JD, Sriram PVJ, et al. Intralesional steroids augment the effects of endoscopic dilation in corrosive esoph ageal strictures. Gastrointest Endosc. 1999;49:509–13.

    Article  CAS  PubMed  Google Scholar 

  46. Hirdes MM, van Hooft JE, Koornstra JJ, et al. Endoscopic corticosteroid injections do not reduce dysphagia after endo scopic dilation therapy in patients with benign esophagogastric anastomotic strictures. Clin Gastroenterol Hepatol. 2013;11:795–801.

    Article  CAS  PubMed  Google Scholar 

  47. Cooper GJ, Sherry KM, Thorpe JA. Changes in gastric tissue oxygenation during mobilization for oesophageal replacement. Eur J Cardiothorac Surg. 1995;9:158–60.

    Article  CAS  PubMed  Google Scholar 

  48. Suma H, Takanashi R. Arteriosclerosis of the gastroepiploic and internal thoracic arteries. Ann Thorac Surg. 2017;50(3):413–6.

    Article  Google Scholar 

  49. Salo JA, Perhoniemi VJ, Heikkinen LO, et al. Pulse oximetry for the assessment of gastric tube circulation in esophageal replacements. Am J Surg. 1992;163:446–7.

    Article  CAS  PubMed  Google Scholar 

  50. Holscher AH, Schneider PM, Gutschow C, et al. Laparoscopic ischemic conditioning of the stomach for esophageal replacement. Ann Surg. 2007;245:241–6.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Oezcelik A, Banki F, DeMeester SR, et al. Delayed esophagogastrostomy: a safe strategy for management of patients with ischemic gastric conduit at time of esophagectomy. J Am Coll Surg. 2009;208:1030–4.

    Article  PubMed  Google Scholar 

  52. Zhang CB, Li J, Zheng J, Wang Q, et al. Feasible study for construction of gastric tube in easophageal reconstruction. J Henan Med Univ. 2005;2(3):175–9.

    Google Scholar 

  53. Heitmiller RF. Impact of gastric tube diameter on upper mediastinal anatomy after transhiatal esophagectomy. Dis Esophagus. 2000;13(4):288–92.

    Article  CAS  PubMed  Google Scholar 

  54. Buunen M, Rooijens PPGM, Smaal HJ, et al. Vascular anat omy of the stomach related to gastric tube construction. Dis Esophagus. 2008;21(3):272–4.

    Article  CAS  PubMed  Google Scholar 

  55. Shu Y-S, Sun C, Shi W-P, et al. Tubular stomach or whole stomach for esophagectomy through cervicothoraco-abdominal approach:a comparative clinical study on anastomotic leakage. Ir J Med Sci. 2013;182(3):477–80.

    Article  PubMed  Google Scholar 

  56. Tabira Y, Sakaguchi T, Kuhara H, et al. The width of a gastric tube has no impact on outcome after esophagectomy. Am J Surg. 2004;187(3):417–21.

    Article  PubMed  Google Scholar 

  57. Shen Y, Wang H, Feng M, et al. The effect of narrowed gastric conduits on anastomotic leakage following minimally invasive oesophagectomy. Interact Cardiovasc Thorac Surg. 2014;19(2):263–8.

    Article  PubMed  Google Scholar 

  58. Ndoye J-M, Dia A, Ndiaye A, et al. Arteriography of three models of gastric oesophagoplasty: the whole stomach, a wide gastric tube and a narrow gastric tube. Surg Radiol Anat. 2006;28(5):429–37.

    Article  PubMed  Google Scholar 

  59. Polglase AL, Hughes ESR, McDermott FT, et al. A comparison of end-to-end staple and suture colorectal anastomosis in the dog. Surg Gynecol Obstet. 1981;152:792–6.

    CAS  PubMed  Google Scholar 

  60. Knezevic JD, Radovanovic NS, Simic AP, et al. Colon interposition in the treatment of esophageal caustic strictures:40 years of experience. Dis Esophagus. 2007;20:530–4.

    Article  CAS  PubMed  Google Scholar 

  61. Akiyama H, Tsurumaru M, Kawamura T, et al. Principles of surgical treatment for carcinoma of the esophagus: analysis of lymph node involvement. Ann Surg. 1981;194:438–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

Not available.

Author information

Authors and Affiliations

Authors

Contributions

Zhong Yuan and Zhai Chunbo were responsible for study design, data acquisition, and analysis, interpretation of the data, and preparation of the draft of the manuscript. Sun Ruijuan, Li Wei and Wang Weiqian was responsible for statistical analysis. Che Jianpeng, Ji Linlin and Guo Bingrong was responsible for study concept and design, interpretation of the data, study supervision, and revision of the manuscript.

Corresponding author

Correspondence to Chunbo Zhai.

Ethics declarations

Ethics approval and consent to participate

This study is a meta-analysis, and ethics approval and consent to participate are not available.

Consent for publication

Not available.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhong, Y., Sun, R., Li, W. et al. Risk factors for esophageal anastomotic stricture after esophagectomy: a meta-analysis. BMC Cancer 24, 872 (2024). https://doi.org/10.1186/s12885-024-12625-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-024-12625-8

Keywords