Skip to main content
Erschienen in: BMC Surgery 1/2018

Open Access 01.12.2018 | Research article

Predictive factors for major postoperative complications related to gastric conduit reconstruction in thoracoscopic esophagectomy for esophageal cancer: a case control study

verfasst von: Shinichiro Kobayashi, Kengo Kanetaka, Yasuhiro Nagata, Masahiko Nakayama, Ryo Matsumoto, Mitsuhisa Takatsuki, Susumu Eguchi

Erschienen in: BMC Surgery | Ausgabe 1/2018

Abstract

Background

Regardless of developments in thoracoscopic esophagectomy (TE), postoperative complications relative to gastric conduit reconstruction are common after esophagectomy. The aim of the present study was to evaluate the predictive factors of major complications related to gastric conduit after TE.

Methods

From 2006 to 2015, 75 patients with esophageal cancer who underwent TE were evaluated to explore the predictive factors of major postoperative complications related to gastric conduit.

Results

Patients with major complications related to gastric conduit had a significantly longer postoperative hospital stay than patients without these complications (P <  0.01). Multivariate analysis demonstrated that three-field lymph node dissection (3FLND) and high serum levels of creatine phosphokinase (CPK) and C-reactive protein (CRP) at 1 postoperative day (1POD) after TE were significant predictive factors of major complications related to gastric conduit [odds ratio (OR) 5.37, 95% confidence interval (CI) 1.41–24.33, P = 0.02; OR 5.40, 95% CI 1.60–20.20, P <  0.01; OR 5.07, 95% CI 1.47–20.25, P = 0.01, respectively]. The incidence rates of major complications related to gastric conduit for 0, 1, 2, and 3 predictive factors were 5.3%, 18.8%, 58.8%, and 85.7%, respectively (P <  0.01).

Conclusions

Two or more factors in 3FLND and the high levels of CPK and CRP at 1POD after TE were identified as the risk model for major complications related to gastric conduit after TE.

Trial registration

UMIN Clinical Trials Registry, ID: UMIN000024436, Registered date: Oct/17/2016.
Abkürzungen
3FLND
Three-field lymph node dissection
AUC
The area under the ROC curve
BMI
Body mass index
CI
Confidence interval
CPK
Creatine phosphokinase
CRP
C-reactive protein
IQR
Interquartile ranges
OR
Odds ratio
POD
Postoperative day
ROC
Receiver operating characteristic
SD
Standard deviation
TE
Thoracoscopic esophagectomy

Background

Although esophagectomy remains the curative treatment for patients with esophageal cancer, this procedure is accompanied by high incidences of complications [1, 2]. The rates of morbidity and mortality after esophagectomy in large national databases were reported to be from 42% to 50% and 2.85% to 4.3%, respectively [37]. Recent developments and improvements in thoracoscopic esophagectomy (TE) have reduced severe pulmonary complications after esophagectomy [8]. However, postoperative complications related to gastric conduit reconstruction are still common after esophagectomy [9]. Regarding cervical anastomotic complications after esophagectomy, leak and stricture formation are major issues [10, 11]. In particular, ischemia of the proximal portion of the graft predisposes these patients to a high incidence of anastomotic complications after esophagectomy [12]. Less commonly, severe graft ischemia can lead to transmural necrosis. Thus, early diagnosis of an ischemic reaction may facilitate appropriate postoperative management and therapeutic intervention to prevent leakage, strictures and necrosis. The aim of the present study was to determine the predictive factors of severe gastric conduit-related postoperative complications.

Methods

Patient population and operations

From 2006 to 2015, 105 patients with esophageal cancer underwent esophagectomy and lymph node dissection at the Department of Surgery at Nagasaki University Hospital. Treatment plans for each patient were provided according to the clinical guidelines edited by the Japan Esophageal Society [13]. We chose open esophagectomy for the patients with severe adhesions in the chest or invasive neoplasia with lymph node involvement. Thirty patients were excluded because they required open esophagectomy with lymph node dissection. Seventy-five consecutive patients with esophageal cancer who underwent TE were retrospectively studied to evaluate the predictive factors for major complications related to gastric conduit after TE. The rules for classification and staging corresponded to the 7th edition of the International Union Against Cancer (UICC)/American Joint Committee on Cancer (AJCC) Tumor Node Metastasis (TNM) staging system [14].
TE was performed from the right side in the left lateral position. Esophagectomy with lymphadenectomy in the mediastinum and around both recurrent nerves were performed. In the abdominal section, hand-assisted laparoscopic gastrectomy was performed to remove the mobilized esophagus with lymphadenectomy around the left gastric artery and aorta. After mobilization of the full stomach and esophagus, a gastric conduit was created by dividing the lesser curve of the stomach. The right gastric and right gastroepiploic artery provided the vascular supply to the created gastric conduit. In 73 patients, the gastric conduit was pulled up in the post-sternal route; in 2 patients, it was pulled up in the post-mediastinal route. The esophagogastrostomy was performed in the neck by end-to-side anastomosis. A 21-mm or 25-mm intraluminal stapler was used as the stapling device (CDH21, CDH25, Ethicon Ltd., Edinburgh, United Kingdom). The inserted part of the gastric conduit was crossed by linear stapling. All staple lines were oversewn.
Three-field lymph node dissection (3FLND) was performed in patients who had upper thoracic esophageal cancer or middle or lower thoracic esophageal cancer with lymph node metastasis in the neck region or around the right recurrent nerve [15].
This study was approved by the Ethics Committee of Nagasaki University Hospital (16082215). The written informed consent from the patients was waved from the Ethics Committee because the information on the opportunity to opt out was presented on the web site (http://​www.​mh.​nagasaki-u.​ac.​jp/​research/​rinsho/​patients/​open_​surgery2.​html). This study was registered in the UMIN Clinical Trials Registry as UMIN000024436.

Postoperative management

The nasogastric tube was removed before anastomosis. On the first postoperative day (1POD), transintestinal nutrition was started from a jejunostomy feeding tube to prevent postoperative complications [16]. In the first three postoperative days, the patients without hoarseness and aspiration pneumonia started to drink fluids, followed by a soft diet.

Definition of major postoperative complications related to gastric conduit reconstruction

Major complications related to gastric conduit after TE were defined as anastomotic leakage, refractory anastomotic strictures, and gastric conduit necrosis. Anastomotic leakage was defined as fistula formation that required any invasive treatment (Clavien-Dindo classification of grade III or more). Anastomotic strictures were defined as the presence of a lumen requiring endoscopic balloon dilatation for the passage of a normal endoscope (9.2 mm diameter) with symptomatic dysphagia. Refractory esophageal strictures were defined as more than 5 sessions of balloon dilation 6 months after the operation [17, 18]. Gastric conduit necrosis was defined as a severe ischemic condition that required resection of the gastric graft.

Statistical analysis

The data are expressed as the means ± standard deviation (SD) or medians and interquartile ranges (IQR). The relationships among major complications related to gastric conduit and age, body mass index (BMI), total operation time, operation time of thoracic surgery, and C-reactive protein (CRP) at 1POD were evaluated using Student’s t-tests. The relationships between major postoperative complications related to gastric conduit reconstruction and other values were evaluated using Wilcoxon’s tests. The relationships among anastomotic leakage and age, body mass index (BMI), total operation time, operation time of thoracic surgery, and C-reactive protein (CRP) at 1POD were evaluated using Student’s t-tests. The relationships between anastomotic leakage and other values were evaluated using Wilcoxon’s tests. The relationships among refractory anastomotic strictures and age, body mass index (BMI), total operation time, operation time of thoracic surgery, and C-reactive protein (CRP) at 1POD were evaluated using Student’s t-tests. The relationships between refractory anastomotic strictures and other values were evaluated using Wilcoxon’s tests. Receiver operating characteristic (ROC) curves and the area under the ROC curve (AUC) were used to assess the feasibility of using CRP and creatine phosphokinase (CPK) at 1POD as diagnostic tools for major complications related to gastric conduit [19]. The 95% CI values greater than 0.5 for AUC indicated that prediction was better than chance [20]. The patients were divided into two groups according to the cut-off values of CRP and CPK at 1POD. The relationships of categorical clinical factors between the groups were analyzed using chi-square tests or Fisher’s exact tests. A Fisher’s exact test was applied if the theoretical frequency was less than five. Probability values (P) less than 0.05 were considered statistically significant. Multiple logistic regression (stepwise) models were developed, and odds ratios (OR) were used to evaluate predictive factors associated with major complications related to gastric conduit. The Cochrane-Armitage trend test was used to test for a linear trend in the proportion of patients who developed major postoperative complications related to gastric conduit reconstruction according to numbers of predictive factors. All statistical analyses were performed using SAS-JMP programs for Windows (SAS Institute Inc., Cary, NC).

Results

Patient characteristics

The clinical characteristics of the 75 patients, which included 18 females and 57 males, are summarized in Table 1. The average age of all patients was 61.3 ± 8.1 years. The average BMI of all patients was 21.3 ± 2.7. Preoperative chemotherapy was performed in 51 patients (68.0%). Three patients (4.0%) were diagnosed with adenocarcinoma, and 72 patients (96.0%) were diagnosed with squamous cell carcinoma. According to the TNM classification, 47 patients (62.7%) had tumors more advanced than stage I. 3FLND was performed in 23 patients (30.7%). The average operating time was 605 ± 114 min. The median estimated blood loss was 370 g (IQR 270–600). Blood transfusion was performed in 7 patients (9.3%). The median length of the postoperative hospital stay was 27 days (IQR 20–39).
Table 1
Patients’ characteristics
Characteristic
Values
Age (year)
61.3 ± 8.1
Gender (Male, Female)
57, 18
BMI
21.3 ± 2.7
Preoperative chemotherapy
51 (68.0%)
TNM Stage (I, II(IIA, IIB), III(IIIA, IIIB, IIIC), IV)
28, 19 (7, 12), 24 (13, 7, 4), 4
Total operating time (min)
605 ± 114
Operation time of thoracic surgery (min)
331 ± 73
Blood loss (g)
370 (270–600)
Blood transfusions
7 (9.3%)
3-field lymph node dissection
23 (30.7%)
Paroxysmal atrial fibrillation
13 (17.3%)
Vasopressor agents
8 (10.7%)
WBC (10^3/μl) at 1POD
9.4 (7.7–12.3)
CRP (10^4 μg/L) at 1POD
9.2 ± 2.4
Lactic acid (mmol/L) at 1POD
1.8 ± 1.2
CPK (IU/L) at 1POD
961 (670–1504)
Postoperative hospital stay (days)
27 (20, 39)
The major complications related to gastric conduit after TE are summarized in Fig. 1. Twenty-three patients (30.7%) developed major complications related to gastric conduit reconstruction after TE. Anastomotic leakage occurred in 17 patients who required drainage to manage infectious conditions. No patients died within 30 days after the operation due to anastomotic leakage. A stricture occurred in 33 patients who required endoscopic balloon dilation. Twenty patients developed simple esophageal strictures without other gastric conduit-related complications. Seven patients developed anastomotic leakage followed by simple esophageal strictures. Six patients developed refractory esophageal strictures. All patient with refractory strictures developed symptomatic strictures within 2 months after TE (28.0 ± 7.0 days). Two patients developed anastomotic leakage followed by refractory esophageal strictures. Two patients had gastric conduit necrosis, and one of these two patients died due to non-occlusive mesenteric ischemia after resection of the necrotic gastric conduit. The length of postoperative hospital stay after TE in the patients with major complications related to gastric conduit (39 days, IQR 28–47) was significantly longer than in those without these complications (22 days, IQR 19–28) (P <  0.01).

Predictive factors for the development of major complications related to gastric conduit after TE

The predictive factors for developing major complications related to gastric conduit are shown in Table 2. In a univariate analysis, the predictive factors for developing major complications related to gastric conduit included age, 3FLND and levels of CRP and CPK at 1POD. The AUC for CRP and CPK at 1POD was 0.684 (95%CI; 0.546–0.796) and 0.670 (95%CI; 0.514–0.796). ROC curve analysis also identified the following cut-off values for CRP and CPK at 1POD: 9.6 × 10^4 μg/L and 1164 IU/L, respectively (Fig. 2). At a threshold of 9.6 × 10^4 μg/L for CRP at 1POD, the optimal sensitivity and specificity were 73.9% and 65.4%, respectively, in patients developing major complications related to gastric conduit. At a threshold of 1164 IU/L for CPK at 1POD, the sensitivity and the specificity were 69.6% and 75.0%, respectively.
Table 2
Univariate analysis for factors predicting major complications related to gastric conduit after TE
 
Postoperative complications related to gastric conduit reconstruction
P-value
Negative (n = 52)
Positive (n = 23)
Age (years)
62.9 ± 7.2
57.5 ± 8.7
<  0.01
Gender (Male, Female)
37, 15
20, 3
N.S.
BMI
21.4 ± 2.7
21.1 ± 2.7
N.S.
Preoperative chemotherapy
21 (40.4%)
13 (56.5%)
N.S.
TNM Stage (I, II, III, IV)
22, 10, 18, 2
6, 9, 6, 2
N.S.
Total operation time (min)
604 ± 113
606 ± 116
N.S.
Operation time of thoracic surgery (min)
337 ± 77
318 ± 64
N.S.
Blood loss (g)
380 (303–623)
340 (200–500)
N.S.
Blood Transfusion
5 (9.6%)
2 (5.2%)
N.S.
3-field lymph node dissection
12 (23.1%)
11 (47.8%)
0.03
Paroxysmal atrial fibrillation
10 (19.2%)
3 (13.0%)
N.S.
Vasopressor agents
5 (9.6%)
3 (13.0%)
N.S.
WBC (10^3/μl) at 1POD
9.7 (8.2–12.8)
8.9 (7.0–11.5)
N.S.
CRP (10^4 μg/L) at 1POD
8.7 ± 0.3
10.3 ± 0.5
<  0.01
Lactic acid (mmol/L) at 1POD
1.4 (1.1–1,8)
2.1 (1.2–2.7)
N.S.
CPK (IU/L) at 1POD
890 (620–1309)
1277 (675–2041)
0.02
Postoperative hospital stay (days)
22 (19–28)
39 (28–47)
<  0.01
The predictive factors for developing major complications related to gastric conduit are shown in Tables 3 and 4. In a univariate analysis, the predictive factors for developing anastomotic leakage related to gastric conduit included levels of CRP and CPK at 1POD.
Table 3
Univariate analysis for factors predicting anastomotic leakage after TE
 
Anastomotic leakage
P-value
Negative (n = 58)
Positive (n = 17)
Age (years)
62.3 ± 7.8
57.8 ± 8.3
N.S.
Gender (Male, Female)
43, 15
14, 3
N.S.
BMI
21.3 ± 2.7
21.1 ± 2.8
N.S.
Preoperative chemotherapy
25 (43.1%)
9 (52.9%)
N.S.
TNM Stage (I, II, III, IV)
22, 13, 20, 3
6, 6, 4, 1
N.S.
Total operation time (min)
608 ± 112
594 ± 122
N.S.
Operation time of thoracic surgery (min)
338 ± 76
305 ± 60
N.S.
Blood loss (g)
380 (290–608)
350 (235–615)
N.S.
Blood Transfusion
6 (10.3%)
1 (5.9%)
N.S.
3-field lymph node dissection
15 (25.9%)
8 (47.1%)
N.S.
Paroxysmal atrial fibrillation
10 (17.2%)
3 (17.7%)
N.S.
Vasopressor agents
7 (12.1%)
1 (5.9%)
N.S.
WBC (10^3/μl) at 1POD
9.7 (8.2–12.8)
8.9 (7.0–11.5)
N.S.
CRP (10^4 μg/L) at 1POD
8.7 ± 2.4
10.5 ± 2.0
<  0.01
Lactic acid (mmol/L) at 1POD
1.5 (1.1–2.0)
2.0 (0.7–2.6)
N.S.
CPK (IU/L) at 1POD
919.5 (629–1400)
1232 (683–2177)
<  0.05
Postoperative hospital stay (days)
22 (19–28)
42 (30–47)
<  0.01
Table 4
Univariate analysis for factors predicting refractory anastomotic strictures after TE
 
Refractory anastomotic strictures
P-value
Negative (n = 69)
Positive (n = 6)
Age (years)
61.4 ± 8.2
59.0 ± 6.4
N.S.
Gender (Male, Female)
51, 18
6, 0
N.S.
BMI
21.4 ± 2.7
21.1 ± 2.7
N.S.
Preoperative chemotherapy
39 (56.5%)
2 (33.3%)
N.S.
TNM Stage (I, II, III, IV)
27, 16, 22, 4
1, 4, 1, 0
N.S.
Total operation time (min)
604 ± 113
606 ± 116
N.S.
Operation time of thoracic surgery (min)
331 ± 75
329 ± 55
N.S.
Blood loss (g)
380 (303–623)
340 (200–500)
N.S.
Blood Transfusion
6 (8.7%)
1 (16.7%)
N.S.
3-field lymph node dissection
22 (29.3%)
1 (16.7%)
N.S.
Paroxysmal atrial fibrillation
12 (17.4%)
1 (16.7%)
N.S.
Vasopressor agents
7 (10.1%)
1 (16.7%)
N.S.
WBC (10^3/μl) at 1POD
9.7 (8.2–12.8)
8.9 (7.0–11.5)
N.S.
CRP (10^4 μg/L) at 1POD
9.0 ± 2.3
10.9 ± 3.6
N.S.
Lactic acid (mmol/L) at 1POD
1.4 (1.1–1,8)
2.1 (1.2–2.7)
N.S.
CPK (IU/L) at 1POD
890 (620–1309)
1214 (675–2041)
N.S.
Postoperative hospital stay (days)
20 (20–35)
41 (25–61)
N.S.
When a multiple logistic regression analysis was performed to evaluate confounding factors, 3FLND and the levels of CPK and CRP at 1POD were found to be significantly associated with developing major complications related to gastric conduit (Table 5). The incidence rates of these complications for 0, 1, 2, and 3 predictive factors were 5.3% (1/19), 18.8% (6/32), 58.8% (10/17), and 85.7% (6/7), respectively (Fig. 3). There was a strong trend toward increasing the prevalence of major complications related to gastric conduit based on the number of predictive factors (P <  0.01). The accuracy of 2 or more factors for major complications related to gastric conduit after TE was 0.800.
Table 5
Multivariate analysis for factors predicting major complications related to gastric conduit after TE
 
Odds ratio
P-value
95% CI
Age (years)
0.92
0.06
(0.85–1.00)
3-field lymph node dissection
5.37
0.02
(1.41–24.33)
CRP at 1POD (high / low)
5.07
0.01
(1.47–20.25)
CPK at 1POD (high / low)
5.40
<  0.01
(1.60–20.20)

Discussion

Our results showed that major postoperative complications related to gastric conduit were frequently present after TE and significantly prolonged the length of the postoperative hospital stay. We first evaluated the predictive factors for major complications related to gastric conduit after TE. Our study investigated the number of factors that could predict these complications after TE.
TE has been shown to reduce pulmonary complications and the length of postoperative hospital stay [8, 21, 22]. These results indicate a faster postoperative recovery in patients after TE than in patients after open esophagectomy. However, the morbidity after TE remains high (approximately 30–40%) [1, 2, 23], and preventing gastric conduit-related complications after TE remains difficult. Our data are similar to the Japanese nationwide web-based database and other articles regarding BMI, operation time, the rate of anastomotic leakage, and postoperative days [5, 7, 24, 25]. There is a therapeutic benefit in predicting the postoperative complications of gastric conduit reconstruction after TE. The 3 predictive factors identified in this study may facilitate the decision to delay oral intake and perform early interventional treatments, such as re-operation, drainage and dilation, after TE.
Our study showed that 3FLND led to major complications related to gastric conduit. Lymph node dissection for thoracic esophageal cancer is controversial, and whether 3FLND or 2-field lymph node dissection (2FLND) is better remains a subject of debate [2629]. The advantages and disadvantages of 3FLND remain controversial when compared to 2FLND of esophagectomy [30, 31]. One meta-analysis showed that 3FLND improves the overall survival rate but leads to more major complications than 2FLND [32, 33]. Anastomotic leakage is likely linked to cervical lymph node dissection due to inflammation and reduced angiogenesis around the anastomotic area [34], which strongly supports our results. However, future studies should determine whether 3FLND or 2FLND is better according to the patient’s physical condition and tumor staging.
The retrosternal route in almost all cases was applied to the gastric conduit of reconstruction after esophagectomy. There are several advantages of this method for the management of local recurrence, including fewer complications in gastric conduit and a short route in the retrosternal route of reconstruction [13, 35, 36]. In RCT studies, both posterior and anterior mediastinal routes of reconstruction were associated with similar surgical outcomes after esophagectomy for cancer [37]. In the Japanese registry, the retrosternal route of reconstruction was selected in 34% of patients, although the posterior mediastinal route was used for reconstruction in 41.3% of patients [38]. Thus, the route of reconstruction remains controversial.
High CRP levels after esophagectomy are reported to precede the clinical diagnosis of postoperative infectious complications [39, 40]. With regard to postoperative infectious complications, there is no difference between patients with and without postoperative infectious complications on 1POD [39, 40]. Consistent with previous reports, our results showed that some infectious complications developed but hardly affected the serum CRP levels on 1POD after TE. Moreover, TE minimizes lung injury and severe pulmonary complications after esophagostomy. Thus, high CRP levels on 1POD may be induced in response to surgical trauma and gastric conduit ischemic conditions after TE. We also identified high CPK levels as a predictive factor for major complications related to gastric conduit after TE. CPK was also reported as a biomarker of ischemic small bowel disease in animal models [41, 42]. CPK may reflect not only ischemic changes in the muscle layer of the gastric conduit but also inflammation around the muscle layers of the neck, as the CPK level is not generally a good biomarker for bowel ischemia [43]. In open esophagectomy, high CPK levels may be observed in patients without major complications related to gastric conduit because of the large incision in the thoracic field.
Postoperative endoscopic examination is a highly accurate method to evaluate reconstruction of the gastric conduit after esophagectomy [12, 4446]. However, endoscopic examination is complex and invasive after esophagectomy. Thus, these predictive factors after TE are useful to select patients who may benefit from endoscopic examination.
Published results have been inconclusive as to which anastomotic technique is ideal for esophagectomy [9, 4751]. Thus, surgeons base their choice of anastomotic technique on personal preference. We applied end-to-side anastomosis with an intraluminal stapler in this study. Cervical anastomosis using a stapler more frequently causes anastomotic strictures than other techniques [9, 52]. However, almost all patients show improved anastomotic strictures after three or fewer dilatations within several months [52]. Thus, the ischemic condition of the gastric conduit may influence anastomotic healing in patients who develop refractory strictures [49, 52].
Our study has several limitations. First, our study was performed at a single institution, and further prospective studies are needed at multiple institutions. Second, the accuracy of the predictive factors is somewhat low. Third, almost all patients were diagnosed with squamous cell carcinoma. Thus, transthoracic extended radical esophagectomy with 3-field lymph node dissection is included in our data. The invasive procedure caused delayed recovery of the patients and resulted in a relatively long postoperative stay. 2-field lymphadenectomy using the Ivor Lewis procedure or trans-hiatal esophagectomy is more commonly performed for esophageal adenocarcinoma in Western countries [5]. Because differences in oncological features and health insurance systems may result in differences in surgical procedures and postoperative stay, it remains unclear whether the predictive factors are applicable to assess patients in Western countries [7, 53]. Fourth, delayed emptying of the gastric conduit was eliminated because there were no patients with endoscopic pyloric dilation and surgical intervention [54, 55]. Despite these limitations, this study is the first to address major complications related to gastric conduit after TE.

Conclusions

In conclusion, 3FLND and the levels of CPK and CRP at 1POD after TE were predictive factors for major complications related to gastric conduit. Two or more factors in 3FLND and the high levels of CPK and CRP at 1POD after TE were identified as the risk model for major complications related to gastric conduit after TE.

Acknowledgements

The authors thank Shuntaro Sato, who helped with the statistical analysis.

Funding

This study was partially supported in  writing and editing the manuscript by a Grant-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (15H06503 and 17 K16569 Shinichiro Kobayashi).

Availability of data and materials

The datasets generated and analyzed during the current study are available from the corresponding author on reasonable request.
This study was approved by the Ethics Committee of Nagasaki University Hospital (16082215). The written informed consent from the patients was waved from the Ethics Committee because the information on the opportunity to opt out was presented on the web site (http://​www.​mh.​nagasaki-u.​ac.​jp/​research/​rinsho/​patients/​open_​surgery2.​html).
Not applicable. Individual identifying data were not included in this manuscript.

Competing interests

Shinichiro Kobayashi, Kengo Kanetaka, Yasuhiro Nagata, Masahiko Nakayama, Ryo Matsumoto, Mitsuhisa Takatsuki, and Susumu Eguchi do not have any financial relationships relevant to this publication to disclose.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.
Literatur
1.
Zurück zum Zitat Verhage RJ, Hazebroek EJ, Boone J, Van Hillegersberg R. Minimally invasive surgery compared to open procedures in esophagectomy for cancer: a systematic review of the literature. Minerva Chir. 2009;64(2):135–46.PubMed Verhage RJ, Hazebroek EJ, Boone J, Van Hillegersberg R. Minimally invasive surgery compared to open procedures in esophagectomy for cancer: a systematic review of the literature. Minerva Chir. 2009;64(2):135–46.PubMed
2.
Zurück zum Zitat Blencowe NS, Strong S, McNair AG, Brookes ST, Crosby T, Griffin SM, Blazeby JM. Reporting of short-term clinical outcomes after esophagectomy: a systematic review. Ann Surg. 2012;255(4):658–66.CrossRefPubMed Blencowe NS, Strong S, McNair AG, Brookes ST, Crosby T, Griffin SM, Blazeby JM. Reporting of short-term clinical outcomes after esophagectomy: a systematic review. Ann Surg. 2012;255(4):658–66.CrossRefPubMed
3.
Zurück zum Zitat Dhungel B, Diggs BS, Hunter JG, Sheppard BC, Vetto JT, Dolan JP. Patient and peri-operative predictors of morbidity and mortality after esophagectomy: American College of Surgeons National Surgical Quality Improvement Program (ACS-NSQIP), 2005-2008. J Gastrointest Surg. 2010;14(10):1492–501.CrossRefPubMed Dhungel B, Diggs BS, Hunter JG, Sheppard BC, Vetto JT, Dolan JP. Patient and peri-operative predictors of morbidity and mortality after esophagectomy: American College of Surgeons National Surgical Quality Improvement Program (ACS-NSQIP), 2005-2008. J Gastrointest Surg. 2010;14(10):1492–501.CrossRefPubMed
4.
Zurück zum Zitat Mamidanna R, Bottle A, Aylin P, Faiz O, Hanna GB. Short-term outcomes following open versus minimally invasive esophagectomy for cancer in England: a population-based national study. Ann Surg. 2012;255(2):197–203.CrossRefPubMed Mamidanna R, Bottle A, Aylin P, Faiz O, Hanna GB. Short-term outcomes following open versus minimally invasive esophagectomy for cancer in England: a population-based national study. Ann Surg. 2012;255(2):197–203.CrossRefPubMed
5.
Zurück zum Zitat Takeuchi H, Miyata H, Gotoh M, Kitagawa Y, Baba H, Kimura W, Tomita N, Nakagoe T, Shimada M, Sugihara K, et al. A risk model for esophagectomy using data of 5354 patients included in a Japanese nationwide web-based database. Ann Surg. 2014;260(2):259–66.CrossRefPubMed Takeuchi H, Miyata H, Gotoh M, Kitagawa Y, Baba H, Kimura W, Tomita N, Nakagoe T, Shimada M, Sugihara K, et al. A risk model for esophagectomy using data of 5354 patients included in a Japanese nationwide web-based database. Ann Surg. 2014;260(2):259–66.CrossRefPubMed
6.
Zurück zum Zitat Yerokun BA, Sun Z, Yang CJ, Gulack BC, Speicher PJ, Adam MA, D'Amico TA, Onaitis MW, Harpole DH, Berry MF, et al. Minimally invasive versus open Esophagectomy for esophageal cancer: a population-based analysis. Ann Thorac Surg. 2016;102(2):416–23.CrossRefPubMedPubMedCentral Yerokun BA, Sun Z, Yang CJ, Gulack BC, Speicher PJ, Adam MA, D'Amico TA, Onaitis MW, Harpole DH, Berry MF, et al. Minimally invasive versus open Esophagectomy for esophageal cancer: a population-based analysis. Ann Thorac Surg. 2016;102(2):416–23.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Takeuchi H, Miyata H, Ozawa S, Udagawa H, Osugi H, Matsubara H, Konno H, Seto Y, Kitagawa Y. Comparison of short-term outcomes between open and minimally invasive Esophagectomy for esophageal cancer using a Nationwide database in Japan. Ann Surg Oncol. 2017;24(7):1821–7.CrossRefPubMed Takeuchi H, Miyata H, Ozawa S, Udagawa H, Osugi H, Matsubara H, Konno H, Seto Y, Kitagawa Y. Comparison of short-term outcomes between open and minimally invasive Esophagectomy for esophageal cancer using a Nationwide database in Japan. Ann Surg Oncol. 2017;24(7):1821–7.CrossRefPubMed
8.
Zurück zum Zitat Findlay JM, Gillies RS, Millo J, Sgromo B, Marshall RE, Maynard ND. Enhanced recovery for esophagectomy: a systematic review and evidence-based guidelines. Ann Surg. 2014;259(3):413–31.CrossRefPubMed Findlay JM, Gillies RS, Millo J, Sgromo B, Marshall RE, Maynard ND. Enhanced recovery for esophagectomy: a systematic review and evidence-based guidelines. Ann Surg. 2014;259(3):413–31.CrossRefPubMed
9.
Zurück zum Zitat Honda M, Kuriyama A, Noma H, Nunobe S, Furukawa TA. Hand-sewn versus mechanical esophagogastric anastomosis after esophagectomy: a systematic review and meta-analysis. Ann Surg. 2013;257(2):238–48.CrossRefPubMed Honda M, Kuriyama A, Noma H, Nunobe S, Furukawa TA. Hand-sewn versus mechanical esophagogastric anastomosis after esophagectomy: a systematic review and meta-analysis. Ann Surg. 2013;257(2):238–48.CrossRefPubMed
10.
Zurück zum Zitat Cassivi SD. Leaks, strictures, and necrosis: a review of anastomotic complications following esophagectomy. Semin Thorac Cardiovasc Surg. 2004;16(2):124–32.CrossRefPubMed Cassivi SD. Leaks, strictures, and necrosis: a review of anastomotic complications following esophagectomy. Semin Thorac Cardiovasc Surg. 2004;16(2):124–32.CrossRefPubMed
11.
Zurück zum Zitat Nishikawa K, Fujita T, Yuda M, Yamamoto S, Tanaka Y, Matsumoto A, Tanishima Y, Yano F, Mitsumori N, Yanaga K. Early postoperative endoscopy for targeted management of patients at risks of anastomotic complications after esophagectomy. Surgery. 2016;160(5):1294–301.CrossRefPubMed Nishikawa K, Fujita T, Yuda M, Yamamoto S, Tanaka Y, Matsumoto A, Tanishima Y, Yano F, Mitsumori N, Yanaga K. Early postoperative endoscopy for targeted management of patients at risks of anastomotic complications after esophagectomy. Surgery. 2016;160(5):1294–301.CrossRefPubMed
12.
Zurück zum Zitat Fujiwara H, Nakajima Y, Kawada K, Tokairin Y, Miyawaki Y, Okada T, Nagai K, Kawano T. Endoscopic assessment 1 day after esophagectomy for predicting cervical esophagogastric anastomosis-relating complications. Surg Endosc. 2016;30(4):1564–71.CrossRefPubMed Fujiwara H, Nakajima Y, Kawada K, Tokairin Y, Miyawaki Y, Okada T, Nagai K, Kawano T. Endoscopic assessment 1 day after esophagectomy for predicting cervical esophagogastric anastomosis-relating complications. Surg Endosc. 2016;30(4):1564–71.CrossRefPubMed
13.
Zurück zum Zitat Kuwano H, Nishimura Y, Oyama T, Kato H, Kitagawa Y, Kusano M, Shimada H, Takiuchi H, Toh Y, Doki Y, et al. Guidelines for diagnosis and treatment of carcinoma of the esophagus April 2012 edited by the Japan esophageal society. Esophagus. 2015;12(1):1–30.CrossRefPubMed Kuwano H, Nishimura Y, Oyama T, Kato H, Kitagawa Y, Kusano M, Shimada H, Takiuchi H, Toh Y, Doki Y, et al. Guidelines for diagnosis and treatment of carcinoma of the esophagus April 2012 edited by the Japan esophageal society. Esophagus. 2015;12(1):1–30.CrossRefPubMed
14.
Zurück zum Zitat Edge SB BD, Compton CC, Fritz AG, Greene FL, Trotti A. AJCC cancer staging manual. 7th ed. New York: Springer-Verlag; 2009. p.103–15. Edge SB BD, Compton CC, Fritz AG, Greene FL, Trotti A. AJCC cancer staging manual. 7th ed. New York: Springer-Verlag; 2009. p.103–15.
15.
Zurück zum Zitat Li H, Yang S, Zhang Y, Xiang J, Chen H. Thoracic recurrent laryngeal lymph node metastases predict cervical node metastases and benefit from three-field dissection in selected patients with thoracic esophageal squamous cell carcinoma. J Surg Oncol. 2012;105(6):548–52.CrossRefPubMed Li H, Yang S, Zhang Y, Xiang J, Chen H. Thoracic recurrent laryngeal lymph node metastases predict cervical node metastases and benefit from three-field dissection in selected patients with thoracic esophageal squamous cell carcinoma. J Surg Oncol. 2012;105(6):548–52.CrossRefPubMed
16.
Zurück zum Zitat Lewis SJ, Andersen HK, Thomas S. Early enteral nutrition within 24 h of intestinal surgery versus later commencement of feeding: a systematic review and meta-analysis. J Gastrointest Surg. 2009;13(3):569–75.CrossRefPubMed Lewis SJ, Andersen HK, Thomas S. Early enteral nutrition within 24 h of intestinal surgery versus later commencement of feeding: a systematic review and meta-analysis. J Gastrointest Surg. 2009;13(3):569–75.CrossRefPubMed
17.
Zurück zum Zitat Kochman ML, McClave SA, Boyce HW. The refractory and the recurrent esophageal stricture: a definition. Gastrointest Endosc. 2005;62(3):474–5.CrossRefPubMed Kochman ML, McClave SA, Boyce HW. The refractory and the recurrent esophageal stricture: a definition. Gastrointest Endosc. 2005;62(3):474–5.CrossRefPubMed
18.
Zurück zum Zitat Yano T, Yoda Y, Nomura S, Toyosaki K, Hasegawa H, Ono H, Tanaka M, Morimoto H, Horimatsu T, Nonaka S, et al. Prospective trial of biodegradable stents for refractory benign esophageal strictures after curative treatment of esophageal cancer. Gastrointest Endosc. 2017;86(3):492–9.CrossRefPubMed Yano T, Yoda Y, Nomura S, Toyosaki K, Hasegawa H, Ono H, Tanaka M, Morimoto H, Horimatsu T, Nonaka S, et al. Prospective trial of biodegradable stents for refractory benign esophageal strictures after curative treatment of esophageal cancer. Gastrointest Endosc. 2017;86(3):492–9.CrossRefPubMed
19.
Zurück zum Zitat Hanley JA, McNeil BJ. The meaning and use of the area under a receiver operating characteristic (ROC) curve. Radiology. 1982;143(1):29–36.CrossRefPubMed Hanley JA, McNeil BJ. The meaning and use of the area under a receiver operating characteristic (ROC) curve. Radiology. 1982;143(1):29–36.CrossRefPubMed
20.
Zurück zum Zitat Lasko TA, Bhagwat JG, Zou KH, Ohno-Machado L. The use of receiver operating characteristic curves in biomedical informatics. J Biomed Inform. 2005;38(5):404–15.CrossRefPubMed Lasko TA, Bhagwat JG, Zou KH, Ohno-Machado L. The use of receiver operating characteristic curves in biomedical informatics. J Biomed Inform. 2005;38(5):404–15.CrossRefPubMed
21.
Zurück zum Zitat Biere SS, van Berge Henegouwen MI, Maas KW, Bonavina L, Rosman C, Garcia JR, Gisbertz SS, Klinkenbijl JH, Hollmann MW, de Lange ES, et al. Minimally invasive versus open oesophagectomy for patients with oesophageal cancer: a multicentre, open-label, randomised controlled trial. Lancet. 2012;379(9829):1887–92.CrossRefPubMed Biere SS, van Berge Henegouwen MI, Maas KW, Bonavina L, Rosman C, Garcia JR, Gisbertz SS, Klinkenbijl JH, Hollmann MW, de Lange ES, et al. Minimally invasive versus open oesophagectomy for patients with oesophageal cancer: a multicentre, open-label, randomised controlled trial. Lancet. 2012;379(9829):1887–92.CrossRefPubMed
22.
Zurück zum Zitat Straatman J, van der Wielen N, Cuesta MA, Daams F, Roig Garcia J, Bonavina L, Rosman C, van Berge Henegouwen MI, Gisbertz SS, van der Peet DL. Minimally invasive versus open esophageal resection: three-year follow-up of the previously reported randomized controlled trial: the TIME trial. Ann Surg. 2017;266(2):232–6.CrossRefPubMed Straatman J, van der Wielen N, Cuesta MA, Daams F, Roig Garcia J, Bonavina L, Rosman C, van Berge Henegouwen MI, Gisbertz SS, van der Peet DL. Minimally invasive versus open esophageal resection: three-year follow-up of the previously reported randomized controlled trial: the TIME trial. Ann Surg. 2017;266(2):232–6.CrossRefPubMed
23.
Zurück zum Zitat Decker G, Coosemans W, De Leyn P, Decaluwe H, Nafteux P, Van Raemdonck D, Lerut T. Minimally invasive esophagectomy for cancer. Eur J Cardiothorac Surg. 2009;35(1):13–21. Decker G, Coosemans W, De Leyn P, Decaluwe H, Nafteux P, Van Raemdonck D, Lerut T. Minimally invasive esophagectomy for cancer. Eur J Cardiothorac Surg. 2009;35(1):13–21.
24.
Zurück zum Zitat Hirahara N, Matsubara T, Mizota Y, Ishibashi S, Tajima Y. Prognostic value of preoperative inflammatory response biomarkers in patients with esophageal cancer who undergo a curative thoracoscopic esophagectomy. BMC Surg. 2016;16(1):66.CrossRefPubMedPubMedCentral Hirahara N, Matsubara T, Mizota Y, Ishibashi S, Tajima Y. Prognostic value of preoperative inflammatory response biomarkers in patients with esophageal cancer who undergo a curative thoracoscopic esophagectomy. BMC Surg. 2016;16(1):66.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Baba Y, Yoshida N, Shigaki H, Iwatsuki M, Miyamoto Y, Sakamoto Y, Watanabe M, Baba H. Prognostic impact of postoperative complications in 502 patients with surgically resected esophageal squamous cell carcinoma: a retrospective single-institution study. Ann Surg. 2016;264(2):305–11.CrossRefPubMed Baba Y, Yoshida N, Shigaki H, Iwatsuki M, Miyamoto Y, Sakamoto Y, Watanabe M, Baba H. Prognostic impact of postoperative complications in 502 patients with surgically resected esophageal squamous cell carcinoma: a retrospective single-institution study. Ann Surg. 2016;264(2):305–11.CrossRefPubMed
26.
Zurück zum Zitat Tsurumaru M, Kajiyama Y, Udagawa H, Akiyama H. Outcomes of extended lymph node dissection for squamous cell carcinoma of the thoracic esophagus. Ann Thorac Cardiovasc Surg. 2001;7(6):325–9.PubMed Tsurumaru M, Kajiyama Y, Udagawa H, Akiyama H. Outcomes of extended lymph node dissection for squamous cell carcinoma of the thoracic esophagus. Ann Thorac Cardiovasc Surg. 2001;7(6):325–9.PubMed
27.
Zurück zum Zitat Tachibana M, Kinugasa S, Yoshimura H, Shibakita M, Tonomoto Y, Dhar DK, Nagasue N. Clinical outcomes of extended esophagectomy with three-field lymph node dissection for esophageal squamous cell carcinoma. Am J Surg. 2005;189(1):98–109.CrossRefPubMed Tachibana M, Kinugasa S, Yoshimura H, Shibakita M, Tonomoto Y, Dhar DK, Nagasue N. Clinical outcomes of extended esophagectomy with three-field lymph node dissection for esophageal squamous cell carcinoma. Am J Surg. 2005;189(1):98–109.CrossRefPubMed
28.
Zurück zum Zitat Hsu WH, Hsu PK, Hsieh CC, Huang CS, Wu YC. The metastatic lymph node number and ratio are independent prognostic factors in esophageal cancer. J Gastrointest Surg. 2009;13(11):1913–20.CrossRefPubMed Hsu WH, Hsu PK, Hsieh CC, Huang CS, Wu YC. The metastatic lymph node number and ratio are independent prognostic factors in esophageal cancer. J Gastrointest Surg. 2009;13(11):1913–20.CrossRefPubMed
29.
Zurück zum Zitat Japan Esophageal Society. Japanese classification of esophageal cancer. 11th edition, part I. Esophagus. 2017;17(1):1–36. Japan Esophageal Society. Japanese classification of esophageal cancer. 11th edition, part I. Esophagus. 2017;17(1):1–36.
30.
Zurück zum Zitat Lerut T, Nafteux P, Moons J, Coosemans W, Decker G, De Leyn P, Van Raemdonck D, Ectors N. Three-field lymphadenectomy for carcinoma of the esophagus and gastroesophageal junction in 174 R0 resections: impact on staging, disease-free survival, and outcome: a plea for adaptation of TNM classification in upper-half esophageal carcinoma. Ann Surg. 2004;240(6):962–74. Lerut T, Nafteux P, Moons J, Coosemans W, Decker G, De Leyn P, Van Raemdonck D, Ectors N. Three-field lymphadenectomy for carcinoma of the esophagus and gastroesophageal junction in 174 R0 resections: impact on staging, disease-free survival, and outcome: a plea for adaptation of TNM classification in upper-half esophageal carcinoma. Ann Surg. 2004;240(6):962–74.
31.
Zurück zum Zitat Chen J, Wu S, Zheng X, Pan J, Zhu K, Chen Y, Li J, Liao L, Lin Y, Liao Z. Cervical lymph node metastasis classified as regional nodal staging in thoracic esophageal squamous cell carcinoma after radical esophagectomy and three-field lymph node dissection. BMC Surg. 2014;14:110.CrossRefPubMedPubMedCentral Chen J, Wu S, Zheng X, Pan J, Zhu K, Chen Y, Li J, Liao L, Lin Y, Liao Z. Cervical lymph node metastasis classified as regional nodal staging in thoracic esophageal squamous cell carcinoma after radical esophagectomy and three-field lymph node dissection. BMC Surg. 2014;14:110.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Ye T, Sun Y, Zhang Y, Chen H. Three-field or two-field resection for thoracic esophageal cancer: a meta-analysis. Ann Thorac Surg. 2013;96(6):1933–41.CrossRefPubMed Ye T, Sun Y, Zhang Y, Chen H. Three-field or two-field resection for thoracic esophageal cancer: a meta-analysis. Ann Thorac Surg. 2013;96(6):1933–41.CrossRefPubMed
33.
Zurück zum Zitat Ma GW, Situ DR, Ma QL, Long H, Zhang LJ, Lin P, Rong TH. Three-field vs two-field lymph node dissection for esophageal cancer: a meta-analysis. World J Gastroenterol. 2014;20(47):18022–30.CrossRefPubMedPubMedCentral Ma GW, Situ DR, Ma QL, Long H, Zhang LJ, Lin P, Rong TH. Three-field vs two-field lymph node dissection for esophageal cancer: a meta-analysis. World J Gastroenterol. 2014;20(47):18022–30.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Zheng QF, Wang JJ, Ying MG, Liu SY. Omentoplasty in preventing anastomotic leakage of oesophagogastrostomy following radical oesophagectomy with three-field lymphadenectomy. Eur J Cardiothorac Surg. 2013;43(2):274–8.CrossRefPubMed Zheng QF, Wang JJ, Ying MG, Liu SY. Omentoplasty in preventing anastomotic leakage of oesophagogastrostomy following radical oesophagectomy with three-field lymphadenectomy. Eur J Cardiothorac Surg. 2013;43(2):274–8.CrossRefPubMed
35.
Zurück zum Zitat Chen H, Lu JJ, Zhou J, Zhou X, Luo X, Liu Q, Tam J. Anterior versus posterior routes of reconstruction after esophagectomy: a comparative anatomic study. Ann Thorac Surg. 2009;87(2):400–4.CrossRefPubMed Chen H, Lu JJ, Zhou J, Zhou X, Luo X, Liu Q, Tam J. Anterior versus posterior routes of reconstruction after esophagectomy: a comparative anatomic study. Ann Thorac Surg. 2009;87(2):400–4.CrossRefPubMed
36.
Zurück zum Zitat Hu H, Ye T, Tan D, Li H, Chen H. Is anterior mediastinum route a shorter choice for esophageal reconstruction? A comparative anatomic study. Eur J Cardiothorac Surg. 2011;40(6):1466–9.PubMed Hu H, Ye T, Tan D, Li H, Chen H. Is anterior mediastinum route a shorter choice for esophageal reconstruction? A comparative anatomic study. Eur J Cardiothorac Surg. 2011;40(6):1466–9.PubMed
37.
Zurück zum Zitat Urschel JD, Urschel DM, Miller JD, Bennett WF, Young JE. A meta-analysis of randomized controlled trials of route of reconstruction after esophagectomy for cancer. Am J Surg. 2001;182(5):470–5.CrossRefPubMed Urschel JD, Urschel DM, Miller JD, Bennett WF, Young JE. A meta-analysis of randomized controlled trials of route of reconstruction after esophagectomy for cancer. Am J Surg. 2001;182(5):470–5.CrossRefPubMed
38.
Zurück zum Zitat Tachimori Y, Ozawa S, Numasaki H, Ishihara R, Matsubara H, Muro K, Oyama T, Toh Y, Udagawa H, Uno T. Comprehensive registry of esophageal cancer in Japan, 2010. Esophagus. 2017;14(3):189–214.CrossRefPubMedPubMedCentral Tachimori Y, Ozawa S, Numasaki H, Ishihara R, Matsubara H, Muro K, Oyama T, Toh Y, Udagawa H, Uno T. Comprehensive registry of esophageal cancer in Japan, 2010. Esophagus. 2017;14(3):189–214.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Miki Y, Toyokawa T, Kubo N, Tamura T, Sakurai K, Tanaka H, Muguruma K, Yashiro M, Hirakawa K, Ohira M. C-reactive protein indicates early stage of postoperative infectious complications in patients following minimally invasive Esophagectomy. World J Surg. 2017;41(3):796–803.CrossRefPubMed Miki Y, Toyokawa T, Kubo N, Tamura T, Sakurai K, Tanaka H, Muguruma K, Yashiro M, Hirakawa K, Ohira M. C-reactive protein indicates early stage of postoperative infectious complications in patients following minimally invasive Esophagectomy. World J Surg. 2017;41(3):796–803.CrossRefPubMed
40.
Zurück zum Zitat Hoeboer SH, Groeneveld AB, Engels N, van Genderen M, Wijnhoven BP, van Bommel J. Rising C-reactive protein and procalcitonin levels precede early complications after esophagectomy. J Gastrointest Surg. 2015;19(4):613–24.CrossRefPubMedPubMedCentral Hoeboer SH, Groeneveld AB, Engels N, van Genderen M, Wijnhoven BP, van Bommel J. Rising C-reactive protein and procalcitonin levels precede early complications after esophagectomy. J Gastrointest Surg. 2015;19(4):613–24.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Kanda T, Nakatomi Y, Ishikawa H, Hitomi M, Matsubara Y, Ono T, Muto T. Intestinal fatty acid-binding protein as a sensitive marker of intestinal ischemia. Dig Dis Sci. 1992;37(9):1362–7.CrossRefPubMed Kanda T, Nakatomi Y, Ishikawa H, Hitomi M, Matsubara Y, Ono T, Muto T. Intestinal fatty acid-binding protein as a sensitive marker of intestinal ischemia. Dig Dis Sci. 1992;37(9):1362–7.CrossRefPubMed
42.
Zurück zum Zitat Kanda T, Tsukahara A, Ueki K, Sakai Y, Tani T, Nishimura A, Yamazaki T, Tamiya Y, Tada T, Hirota M, et al. Diagnosis of ischemic small bowel disease by measurement of serum intestinal fatty acid-binding protein in patients with acute abdomen: a multicenter, observer-blinded validation study. J Gastroenterol. 2011;46(4):492–500.CrossRefPubMed Kanda T, Tsukahara A, Ueki K, Sakai Y, Tani T, Nishimura A, Yamazaki T, Tamiya Y, Tada T, Hirota M, et al. Diagnosis of ischemic small bowel disease by measurement of serum intestinal fatty acid-binding protein in patients with acute abdomen: a multicenter, observer-blinded validation study. J Gastroenterol. 2011;46(4):492–500.CrossRefPubMed
43.
Zurück zum Zitat van der Voort PH, Westra B, Wester JP, Bosman RJ, van Stijn I, Haagen IA, Loupatty FJ, Rijkenberg S. Can serum L-lactate, D-lactate, creatine kinase and I-FABP be used as diagnostic markers in critically ill patients suspected for bowel ischemia. BMC Anesthesiol. 2014;14:111.CrossRefPubMedPubMedCentral van der Voort PH, Westra B, Wester JP, Bosman RJ, van Stijn I, Haagen IA, Loupatty FJ, Rijkenberg S. Can serum L-lactate, D-lactate, creatine kinase and I-FABP be used as diagnostic markers in critically ill patients suspected for bowel ischemia. BMC Anesthesiol. 2014;14:111.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Oezcelik A, Banki F, Ayazi S, Abate E, Zehetner J, Sohn HJ, Hagen JA, DeMeester SR, Lipham JC, Palmer SL, et al. Detection of gastric conduit ischemia or anastomotic breakdown after cervical esophagogastrostomy: the use of computed tomography scan versus early endoscopy. Surg Endosc. 2010;24(8):1948–51.CrossRefPubMed Oezcelik A, Banki F, Ayazi S, Abate E, Zehetner J, Sohn HJ, Hagen JA, DeMeester SR, Lipham JC, Palmer SL, et al. Detection of gastric conduit ischemia or anastomotic breakdown after cervical esophagogastrostomy: the use of computed tomography scan versus early endoscopy. Surg Endosc. 2010;24(8):1948–51.CrossRefPubMed
45.
Zurück zum Zitat Page RD, Asmat A, McShane J, Russell GN, Pennefather SH. Routine endoscopy to detect anastomotic leakage after esophagectomy. Ann Thorac Surg. 2013;95(1):292–8.CrossRefPubMed Page RD, Asmat A, McShane J, Russell GN, Pennefather SH. Routine endoscopy to detect anastomotic leakage after esophagectomy. Ann Thorac Surg. 2013;95(1):292–8.CrossRefPubMed
46.
Zurück zum Zitat Schaible A, Sauer P, Hartwig W, Hackert T, Hinz U, Radeleff B, Büchler MW, Werner J. Radiologic versus endoscopic evaluation of the conduit after esophageal resection: a prospective, blinded, intraindividually controlled diagnostic study. Surg Endosc. 2014;28(7):2078–85.CrossRefPubMed Schaible A, Sauer P, Hartwig W, Hackert T, Hinz U, Radeleff B, Büchler MW, Werner J. Radiologic versus endoscopic evaluation of the conduit after esophageal resection: a prospective, blinded, intraindividually controlled diagnostic study. Surg Endosc. 2014;28(7):2078–85.CrossRefPubMed
47.
Zurück zum Zitat Dewar L, Gelfand G, Finley RJ, Evans K, Inculet R, Nelems B. Factors affecting cervical anastomotic leak and stricture formation following esophagogastrectomy and gastric tube interposition. Am J Surg. 1992;163(5):484–9.CrossRefPubMed Dewar L, Gelfand G, Finley RJ, Evans K, Inculet R, Nelems B. Factors affecting cervical anastomotic leak and stricture formation following esophagogastrectomy and gastric tube interposition. Am J Surg. 1992;163(5):484–9.CrossRefPubMed
48.
Zurück zum Zitat Walther B, Johansson J, Johnsson F, Von Holstein CS, Zilling T. Cervical or thoracic anastomosis after esophageal resection and gastric tube reconstruction: a prospective randomized trial comparing sutured neck anastomosis with stapled intrathoracic anastomosis. Ann Surg. 2003;238(6):803–14. Walther B, Johansson J, Johnsson F, Von Holstein CS, Zilling T. Cervical or thoracic anastomosis after esophageal resection and gastric tube reconstruction: a prospective randomized trial comparing sutured neck anastomosis with stapled intrathoracic anastomosis. Ann Surg. 2003;238(6):803–14.
49.
Zurück zum Zitat Zhang YS, Gao BR, Wang HJ, Su YF, Yang YZ, Zhang JH, Wang C. Comparison of anastomotic leakage and stricture formation following layered and stapler oesophagogastric anastomosis for cancer: a prospective randomized controlled trial. J Int Med Res. 2010;38(1):227–33.CrossRefPubMed Zhang YS, Gao BR, Wang HJ, Su YF, Yang YZ, Zhang JH, Wang C. Comparison of anastomotic leakage and stricture formation following layered and stapler oesophagogastric anastomosis for cancer: a prospective randomized controlled trial. J Int Med Res. 2010;38(1):227–33.CrossRefPubMed
50.
Zurück zum Zitat Kim RH, Takabe K. Methods of esophagogastric anastomoses following esophagectomy for cancer: a systematic review. J Surg Oncol. 2010;101(6):527–33.CrossRefPubMed Kim RH, Takabe K. Methods of esophagogastric anastomoses following esophagectomy for cancer: a systematic review. J Surg Oncol. 2010;101(6):527–33.CrossRefPubMed
51.
Zurück zum Zitat Xu QR, Wang KN, Wang WP, Zhang K, Chen LQ. Linear stapled esophagogastrostomy is more effective than hand-sewn or circular stapler in prevention of anastomotic stricture: a comparative clinical study. J Gastrointest Surg. 2011;15(6):915–21.CrossRefPubMed Xu QR, Wang KN, Wang WP, Zhang K, Chen LQ. Linear stapled esophagogastrostomy is more effective than hand-sewn or circular stapler in prevention of anastomotic stricture: a comparative clinical study. J Gastrointest Surg. 2011;15(6):915–21.CrossRefPubMed
52.
Zurück zum Zitat Law S, Fok M, Chu KM, Wong J. Comparison of hand-sewn and stapled esophagogastric anastomosis after esophageal resection for cancer: a prospective randomized controlled trial. Ann Surg. 1997;226(2):169–73.CrossRefPubMedPubMedCentral Law S, Fok M, Chu KM, Wong J. Comparison of hand-sewn and stapled esophagogastric anastomosis after esophageal resection for cancer: a prospective randomized controlled trial. Ann Surg. 1997;226(2):169–73.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Muramatsu N, Liang J. Hospital length of stay in the United States and Japan: a case study of myocardial infarction patients. Int J Health Serv. 1999;29(1):189–209.CrossRefPubMed Muramatsu N, Liang J. Hospital length of stay in the United States and Japan: a case study of myocardial infarction patients. Int J Health Serv. 1999;29(1):189–209.CrossRefPubMed
54.
Zurück zum Zitat Akkerman RD, Haverkamp L, van Hillegersberg R, Ruurda JP. Surgical techniques to prevent delayed gastric emptying after esophagectomy with gastric interposition: a systematic review. Ann Thorac Surg. 2014;98(4):1512–9.CrossRefPubMed Akkerman RD, Haverkamp L, van Hillegersberg R, Ruurda JP. Surgical techniques to prevent delayed gastric emptying after esophagectomy with gastric interposition: a systematic review. Ann Thorac Surg. 2014;98(4):1512–9.CrossRefPubMed
55.
Zurück zum Zitat Poghosyan T, Gaujoux S, Chirica M, Munoz-Bongrand N, Sarfati E, Cattan P. Functional disorders and quality of life after esophagectomy and gastric tube reconstruction for cancer. J Visc Surg. 2011;148(5):e327–35.CrossRefPubMed Poghosyan T, Gaujoux S, Chirica M, Munoz-Bongrand N, Sarfati E, Cattan P. Functional disorders and quality of life after esophagectomy and gastric tube reconstruction for cancer. J Visc Surg. 2011;148(5):e327–35.CrossRefPubMed
Metadaten
Titel
Predictive factors for major postoperative complications related to gastric conduit reconstruction in thoracoscopic esophagectomy for esophageal cancer: a case control study
verfasst von
Shinichiro Kobayashi
Kengo Kanetaka
Yasuhiro Nagata
Masahiko Nakayama
Ryo Matsumoto
Mitsuhisa Takatsuki
Susumu Eguchi
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Surgery / Ausgabe 1/2018
Elektronische ISSN: 1471-2482
DOI
https://doi.org/10.1186/s12893-018-0348-9

Weitere Artikel der Ausgabe 1/2018

BMC Surgery 1/2018 Zur Ausgabe

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.