Skip to main content
Erschienen in: World Journal of Surgical Oncology 1/2018

Open Access 01.12.2018 | Research

Loss of skeletal muscle mass during neoadjuvant treatments correlates with worse prognosis in esophageal cancer: a retrospective cohort study

verfasst von: Tommi Järvinen, Ilkka Ilonen, Juha Kauppi, Jarmo Salo, Jari Räsänen

Erschienen in: World Journal of Surgical Oncology | Ausgabe 1/2018

Abstract

Background

Nutritional deficits, cachexia, and sarcopenia are extremely common in esophageal cancer. The aim of this article was to assess the effect of loss of skeletal muscle mass during neoadjuvant treatment on the prognosis of esophageal cancer patients.

Methods

Esophageal cancer patients (N = 115) undergoing neoadjuvant therapy and surgery between 2010 and 2014 were identified from our surgery database and retrospectively analyzed. Computed tomography imaging of the total cross-sectional muscle tissue measured at the third lumbar level defined the skeletal muscle index, which defined sarcopenia (SMI < 52.4 cm2/m2 for men and < 38.5 cm2/m2 for women). Images were collected before and after neoadjuvant treatments.

Results

Sarcopenia in preoperative imaging was prevalent in 92 patients (80%). Median overall survival was 900 days (interquartile range 334–1447) with no difference between sarcopenic (median = 900) and non-sarcopenic (median = 914) groups (p = 0.872). Complication rates did not differ (26.1% vs 32.6%, p = 0.725). A 2.98% decrease in skeletal muscle index during neoadjuvant treatment correlated with poor 2-year survival (log-rank p = 0.04).

Conclusion

Loss of skeletal muscle tissue during neoadjuvant treatment correlates with worse overall survival.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12957-018-1327-4) contains supplementary material, which is available to authorized users.
Abkürzungen
AC
Adenocarcinoma
BMI
Body mass index
CCI
Charlson Comorbidity Index
CI
Confidence interval
CRT
Chemoradiotherapy
CT
Computed tomography
EC
Esophageal cancer
ECOG
The Eastern Cooperative Oncology Group
EMR
Endoscopic mucosal resection
EOX
Epirubicin–oxaliplatin–capecitabine
EUS
Endoscopic ultrasound
HU
Hounsfield unit
ICU
Intensive care unit
IQR
Interquartile range
MIE
Minimally invasive esophagectomy
OS
Overall survival
PEG
Percutaneous gastrostomy
PET
Positron emission tomography
RFA
Radiofrequency ablation
RFS
Recurrence-free survival
ROI
Region of interest
SCC
Squamous cell carcinoma
SD
Standard deviation
SEMS
Self-expandable metallic stent
SMI
Skeletal muscle index
TMA
Total muscle area
VATS
Video assisted thoracoscopic surgery

Background

Esophageal cancer (EC) is intimately related to weight changes and poor nutritional status, since the most common symptoms of EC are dysphagia and weight loss [1]. In locally advanced EC, baseline nutritional status has been linked to survival after definitive chemoradiotherapy [2]. Preoperative weight loss has also been linked to worse outcomes [3]. A high body mass index (BMI) has not been found to have a significant effect on survival in EC [4, 5].
In the recent years, there has been an increasing amount of studies on frailty and especially sarcopenia as prognostic factors in cancers. Sarcopenia is defined as the progressive loss of muscle related to aging or disease [6]. Sarcopenia has been associated with worse outcomes in many types of cancers such as hepatocellular carcinoma, colorectal cancer, and small cell lung cancer [79]. For esophageal and gastroesophageal junction cancers, there are conflicting reports. Worse long-term outcomes have been reported in resected esophageal or gastroesophageal junctional cancers [1012]. Sarcopenia has been linked to increased pulmonary and other complication rates [13, 14]. Decreased skeletal muscle area during neoadjuvant therapy has also been associated with poorer outcomes and risk of positive clinical resection margin [15, 16]. There are also reports of sarcopenia not being an independent prognostic risk factor for mortality, morbidity, or poor outcomes in EC after neoadjuvant chemoradiotherapy or chemotherapy [14, 1719]. One study found a significant correlation with lean psoas mass and survival in patients not undergoing neoadjuvant treatment, but no such effect on patients receiving neoadjuvant treatment [20].
The aim of this trial was to assess the effect of sarcopenia and loss of skeletal muscle index during neoadjuvant treatments in patients undergoing esophagectomy for EC. The primary end-point is overall survival, and secondary end points are recurrence-free survival and complication rates.

Methods

Patients

Patients who underwent surgical resection and neoadjuvant therapy for EC between 2010 and 2014 were identified in retrospect from our surgery database using type of surgery and diagnosis of esophageal or junctional cancer as identifiers (N = 118). Patients who had no eligible imaging for analysis of the skeletal muscle index were excluded (N = 3), leaving 115 patients. Collected data included patient characteristics, weight, and weight-loss data before and during the treatment and follow up, primary tumor characteristics and staging, specifics of neoadjuvant, endoscopic, surgical and adjuvant treatments, post-operative and long-term complications, and overall survival. Weight loss here is defined as unintentional deviation from healthy weight (weight 6 months before diagnosis). Complications were collected as recommended by an international consensus statement [21]. 30- and 90-day overall survival rates and 2-year overall survival and recurrence-free survival rates were also collected.
CT (computed tomography) scans were collected from time of initial staging, post-neodjuvant, 6-, and 18-month follow-up visits. CT scans were excluded if there was impaired visibility at third lumbar vertebra or over 1 month of time interval between the CT scan and associated event (start of neoadjuvant therapy, operation, 6-month follow-up visit, or 18-month follow-up visit).

Preoperative staging

All patients underwent gastroscopies with biopsies that confirmed the diagnosis of EC. All patients underwent CT scans of the thorax, abdomen, and pelvis and a routine total body PET-CT scan. Endoscopic ultrasound was done unless prevented by tumor obstruction or stent insertion and assessed the invasion depth of the tumor and identified regional enlarged lymph nodes. PET/CT scans were routinely repeated after neoadjuvant treatments for preoperative planning and to assess the radiologic response of the tumor.

Neoadjuvant treatment protocols

Treatment strategies were discussed and decided together with oncologists. Patients with nodal disease spread (cN+) or transmural tumor invasion (cT ≥ 3) underwent neoadjuvant treatment, unless contraindicated. Neoadjuvant therapy was epirubicin–oxaliplatin–capecitabine neoadjuvant chemotherapy as per MAGIC (Medical Research Council Adjuvant Gastric Infusional Chemotherapy) protocol for esophageal adenocarcinoma [22]. Squamous cell carcinoma was treated with neoadjuvant chemoradiotherapy consisting of 2 cycles of platin- and 5-fluorouracil-based therapy over 5–6 weeks. Chemosensitization was followed by a 45 Gy total dose of radiation to the tumor and regional nodes, in 1.8 Gy daily fraction.

Preoperative endoscopic procedures

Endoscopic mucosal resection (EMR) with or without radiofrequency ablation (RFA) was discussed as an option for patients with mucosal tumors or high-grade dysplasia. The initial decision whether to proceed with endoscopic treatments or to do an esophagectomy was a shared decision between the surgeon and the patient. If endoscopic mucosal resection showed submucosal spread or there was cancer recurrence, treatment proceeded to esophagectomy. One patient included in this study had a prior EMR. Patients with obstructing tumor growth and marked dysphagia preventing the ingestion of solid foods were treated with insertion of a self-expandable metallic stent (SEMS, N = 35) or a percutaneous endoscopic gastrostomy (PEG, N = 4) tube.

Surgical treatment and follow-up

Surgical techniques included minimally invasive esophagectomy, hybrid minimally invasive esophagectomy with either laparoscopy or video-assisted thoracoscopic surgery, Ivor-Lewis esophagectomy, 3-part esophagectomy, and transhiatal esophagectomy. These techniques are described elsewhere [23]. Post-operative stage of the tumor was reported according to eighth edition AJCC/IAUC staging [24]. Amount of positive lymph nodes and total lymph node count was recorded.
Patients were followed until death or January 2017, yielding a follow-up period of at least 24 months. Patients were met at an outpatient clinic 1 month after surgery to assess the recovery from surgery. Gastroscopies were done every 6 months for 2 years after surgery and annually for up to 5 years. CT scans were taken 6 months after surgery, 18 months after surgery, and annually up to 5 years.
All the treatments discussed here are standard approaches in our institution.

Measurement of muscle parameters and sarcopenia definition

Scans were coded in order to blind the researcher from outcome. Images were imported to Osirix® Version 3.3 (32-bit Pixmeo, Sarl, Switzerland). We selected a single image on the level of L3, with both transverse processes and delineated abdominal muscles by use of a semi-automated selection of region of interest. Psoas, quadratus lumborum, paraspinal, transverse abdominal, external oblique, internal oblique, and rectus abdominis muscles were included. The Hounsfield unit threshold range for skeletal muscle was − 29 to + 150. The images were manually corrected, if needed, by the propulsion and brush tools in Osirix©. The cross-sectional total muscle area at the level of L3 (cm2) was divided by the square of height (m2), which produced the skeletal muscle index (SMI). This method is suggested as the preferred method of measuring the muscle mass of cancer patients [25]. SMI limit for sarcopenia was < 52.4 cm2/m2 for men and < 38.5 cm2/m2 for women, based on a previous study by Prado et al. [26] For the survival and complication analyses, the preoperative SMI values were used, unless stated otherwise.
The process of delineating the abdominal muscle mass is shown in Fig. 1.

Statistical analysis

All statistical analysis was done with R Project. (R Core Team, 2016). R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. URL: https://​www.​R-project.​org/).
Continuous parameters were tested for normality using Shapiro-Wilk’s test and deemed normally distributed if p > 0.05. Normal continuous data is presented as mean and standard deviation (SD) whereas non-normal data is described with median and interquartile range (IQR). For comparing normal scalar variables between two groups, independent samples Student’s t test was used and for non-normal variables Mann-Whitney U test was used. The 2-tailed χ2 test served for categorical variables. Kaplan-Meier survival analysis and log-rank test demonstrated the possible difference of survival between groups.

Results

Baseline characteristics of the study population are displayed in Table 1. Included patients numbered 115, of which sarcopenia was found in 92 (80%). Median overall survival was 900 days (interquartile range 334–1447). Patients with and without preoperative sarcopenia are compared in Table 1. The sarcopenic patients were statistically significantly older, taller, weighed less, and had smaller prevalence of current smokers.
Table 1
Baseline characteristics of the study patient population
 
Overall
Pre-operative sarcopenia
p
 
No
Yes
Number of patients (%)
115
23
92
 
Sex (%)
 Female
29
(25.2)
9
(39.1)
20
(21.7)
0.147
 Male
86
(74.8)
14
(60.9)
72
(78.3)
 
ECOG (%)
 0
40
(35.1)
11
(47.8)
29
(31.9)
0.140
 1
69
(60.5)
10
(43.5)
59
(64.8)
 
 2
5
(4.4)
2
(8.7)
3
(3.3)
 
Smoking status (%)
 Current smoker
32
(27.8)
11
(47.8)
21
(22.8)
0.018
 Ex-smoker
40
(34.8)
3
(13.0)
37
(40.2)
 
 Non-smoker
43
(37.4)
9
(39.1)
34
(37.0)
 
T stage (%)
 1
2
(1.8)
0
(0.0)
2
(2.2)
0.894
 2
10
(8.9)
2
(8.7)
8
(9.0)
 
 3
86
(76.8)
17
(73.9)
69
(77.5)
 
 4
14
(12.5)
4
(17.4)
10
(11.2)
 
N stage (%)
 0
33
(29.5)
6
(26.1)
27
(30.3)
0.258
 1
74
(66.1)
15
(65.2)
59
(66.3)
 
 2
4
(3.6)
1
(4.3)
3
(3.4)
 
 3
1
(0.9)
1
(4.3)
0
(0.0)
 
Cancer type (%)
 Adenocarcinoma
88
(76.5)
21
(91.3)
67
(72.8)
0.061
 SCC*
27
(23.5)
2
(8.7)
25
(27.2)
 
Pathological grade** (%)
 1
9
(16.1)
2
(28.6)
7
(14.3)
0.723
 2
22
(39.3)
3
(42.9)
19
(38.8)
 
 3
25
(44.7)
2
(28.6)
23
(46.9)
 
Tumor location (%)
 Lower third
95
(82.6)
20
(87.0)
75
(81.5)
0.646
 Middle third
17
(14.8)
3
(13.0)
14
(15.2)
 
 Upper third
3
(2.6)
0
(0.0)
3
(3.3)
 
Neoadjuvant treatment (%)
 Chemoradiation
28
(24.3)
2
(8.7)
26
(28.3)
0.051
 Chemotherapy
87
(75.7)
21
(91.3)
66
(71.7)
 
Operation type (%)
 MIE
78
(67.8)
18
(78.3)
60
(65.2)
0.464
 Thoracotomy
26
(22.6)
2
(8.7)
24
(26.1)
 
 Hybrid-laparoscopy
5
(4.3)
1
(4.3)
4
(4.3)
 
 Hybrid-VATS
3
(2.6)
1
(4.3)
2
(2.2)
 
 Transhiatal
3
(2.6)
1
(4.3)
2
(2.2)
 
Preop. endo. treatment (%)
 Any
40
(34.8)
4
(17.4)
36
(39.1)
0.087
 Stent
35
(30.4)
3
(13.0)
32
(34.8)
0.076
 PEG§
4
(3.5)
1
(4.3)
3
(3.3)
1.000
 EMR
1
(0.9)
1
(4.3)
0
(0.0)
0.451
Age, years (mean [SD])
63
[9]
59
[8]
64
[9]
0.015
Height, cm (median [IQR])
174
[166,179]
171
[160,174]
175
[167,180]
0.009
Preop. weight, kg (mean [SD])
74
[16]
82
[18]
73
[15]
0.017
Weight loss, kg (median [IQR])
7
[0, 13]
6
[0, 12]
8
[2, 12]
0.509
FEV1% (mean [SD])
90
[18]
90
[21]
91
[18]
0.914
Creatinine, umol/l (mean [SD])
71
[19]
72
[18]
71
[20]
0.810
CCI (median [IQR])
5
[4, 6]
5
[4, 6]
5
[5, 6]
0.098
*Squamous cell carcinoma
**Grade not reported on all pathological reports
Minimally invasive esophagectomy
Video-assisted thoracoscopic surgery
§Percutaneous endoscopic gastrostomy
Endoscopic mucosal resection
The progression of mean weight- and body composition-related parameters during neoadjuvant treatments and follow-up is displayed in Fig. 2. Ninety-one (79.1%) patients had sarcopenia before neoadjuvant treatments, 92 (80%) were sarcopenic before esophagectomy, 82 of 99 (82.8%) were sarcopenic 6 months post-operatively, and 67 of 78 (85.9%) 18 months post-operatively.
Table 2 shows the relevant complication rates and Clavien-Dindo scores of patients. The amount of intraoperative bleeding and length of initial intensive care unit stay are also displayed. There was no statistically significant difference between the sarcopenic or non-sarcopenic groups in any complication groups. An additional table shows a more exhaustive table of complication rates [see Additional file 1].
Table 2
Analysis of complication rates by preoperative sarcopenia
 
Preoperative
  
Sarcopenia
 
Level
No
Yes
p
(N = 23)
(N = 92)
30-day mortality (%)
 
0
(0.0)
3
(3.3)
0.884
90-day mortality (%)
 
1
(4.3)
6
(6.5)
1.000
Any complication (%)
 
17
(73.9)
62
(67.4)
0.725
Clavien-Dindo score (%)
0
6
(26.1)
30
(32.6)
0.886
 
1
0
(0.0)
3
(3.3)
 
 
2
4
(17.4)
10
(10.9)
 
 
3a
3
(13.0)
13
(14.1)
 
 
3b
7
(30.4)
20
(21.7)
 
 
4a
2
(8.7)
11
(12.0)
 
 
4b
0
(0.0)
2
(2.2)
 
 
5
1
(4.3)
3
(3.3)
 
Chyle leak (%)
 
1
(4.3)
8
(8.7)
0.795
Anastomotic leak (%)
 
2
(8.7
13
(14.1)
0.729
Conduit necrosis (%)
 
1
(4.3)
2
(2.2)
1.000
Recurrent nerve palsy (%)
 
1
(4.3)
7
(7.6)
0.927
Intraoperative Complications (%)
 
3
(13.0)
9
(9.8)
0.939
Pulmonary complications (%)
 
6
(26.1)
26
(28.3)
1.000
Reoperation rate (%)
 
2
(8.7)
8
(8.7)
1.000
Operative bleeding, ml (median [IQR])
 
200
[150, 400]
400
[150, 700]
0.091
ICU stay, days (median [IQR])
 
3
[1, 5]
2
[1, 4]
0.535
Tracheostomy rate (%)
 
1
(4.3)
8
(8.7)
0.795
Intraoperative vessel, conduit or airway injury or conversion to open esophagectomy
Contains ARDS (acute respiratory distress syndrome), pneumonia, atelectasis requiring intervention, pleural effusion or pneumothorax requiring intervention, pulmonary embolism, and acute aspiration
There was no statistical difference in 2-year overall survival or recurrence-free survival between the preoperative sarcopenic and non-sarcopenic groups (p = 0.74 and p = 0.64, respectively). The Kaplan-Meier curves are shown in Fig. 3a, b. Neither preneoadjuvant SMI nor preoperative SMI had an effect on OS (p = 0.6023 and p = 0.3843) or RFS (p = 0.3241 and p = 0.9273).
Patients were divided into groups based on the change between preneoadjuvant measurement and preoperative measurement of SMI. Median percentual change of SMI (− 2.98%) was used as the cut-off value, as has been done in a previous study [15]. The baseline characteristics are displayed in Table 3. The group with more muscle loss had more preoperative stents inserted (p = 0.037). SMI change below the median correlated with 2-year overall survival (p = 0.022) but not 2-year RFS (p = 0.11), as shown in Fig. 4a–b. The change of SMI during neoadjuvant treatments was significantly different between 2-year survivors and non-survivors with mean changes of − 0.69 and − 6.20%, respectively (p = 0.01259).
Table 3
Characteristics by SMI change groups
 
SMI change
p
 
< − 2.98%
> − 2.98%
Number of patients (%)
57
 
58
  
Sex (%)
 Female
19
(33.3)
10
(17.2)
0.076
 Male
38
(66.7)
48
(82.8)
 
ECOG (%)
 0
15
(26.3)
25
(43.9)
0.108
 1
40
(70.2)
29
(50.9)
 
 2
2
(3.5)
3
(5.3)
 
Smoking status (%)
 Current smoker
20
(35.1)
12
(20.7)
0.086
 Ex-smoker
21
(36.8)
19
(32.8)
 
 Non-smoker
16
(28.1)
27
(46.6)
 
T stage
 1
2
(3.8)
0
(0.0)
0.549
 2
5
(9.3)
5
(8.6)
 
 3
42
(77.8)
44
(75.9)
 
 4
5
(9.3)
9
(15.5)
 
N stage
 0
14
(25.9)
19
(32.8)
0.445
 1
38
(70.4)
36
(62.1)
 
 2
1
(1.9)
3
(5.2)
 
 3
1
(1.9)
0
(0.0)
 
Cancer type (%)
 Adenocarcinoma
40
(70.2)
48
(82.8)
0.111
 SCC*
17
(29.8)
10
(17.2)
 
Pathological grade**
 1
3
(13.0)
6
(18.2)
0.790
 2
9
(39.1)
13
(39.4)
 
 3
12
(47.9)
14
(42.4)
 
Tumor location (%)
 Lower third
45
(78.9)
50
(86.2)
0.357
 Middle third
11
(19.3)
6
(10.3)
 
 Upper third
1
(1.8)
2
(3.4)
 
Neoadjuvant treatment (%)
 Chemoradiation
19
(33.3)
11
(19.0)
0.079
 Chemotherapy
38
(66.7)
47
(81.0)
 
Operation type (%)
 MIE
39
(68.4)
39
(67.2)
0.931
 Thoracotomy
13
(22.8)
13
(22.4)
 
 Hybrid-laparoscopy
2
(3.5)
3
(5.2)
 
 Hybrid-VATS
2
(3.5)
1
(1.7)
 
 Transhiatal
1
(1.8)
2
(3.4)
 
Preop. endo. treatment (%)
 Any
26
(45.6)
14
(24.1)
0.026
 Stent
23
(40.4)
12
(20.7)
0.037
 PEG§
2
(3.5)
2
(3.4)
1.000
 EMR
1
(1.8)
0
(0.0)
0.993
Age, years (mean [SD])
63
[9]
63
[9]
0.817
Height, cm (median [IQR])
173
[164,179]
174
[168,179]
0.350
Preop. weight, kg (mean [SD])
74
[17]
75
[16]
0.840
Weight loss, kg (median [IQR])
10
[0, 15]
6
[2, 10]
0.131
FEV1% (mean [SD])
89
[21]
92
[16]
0.527
Creatinine, umol/l (mean [SD])
70
[22]
72
[16]
0.484
CCI (median [IQR])
5
[5, 6]
5
[4, 6]
0.397
*Squamous cell carcinoma
**Grade not reported on all pathological reports
†Minimally invasive esophagectomy
‡Video-assisted thoracoscopic surgery
§Percutaneous endoscopic gastrostomy
¶Endoscopic mucosal resection
Finally, a multivariate Cox regression analysis was done using the backwards elimination method with a p value limit of 0.2. The model constructed is shown in Table 4. The model showed that the patients who had percentual SMI change over the median (− 2.98%) had better survival than those whose SMI had decreased more (p = 0.049; HR − 0.609; HR 95% CI 0.297–0.997). No other covariates reached significance (N stage, T stage, or CCI). There was 11.75 events per variable (47 events, 4 variables), which is over the suggested limit of 10 (Table 4) [27].
Table 4
Multivariate Cox regression analysis of the covariates affecting OS
 
HR
95% CI
p
ΔSMI%* during neoadj. (>median vs. <median)
-0.609
0.297–0.997
0.049
N stage (per level)
0.486
0.940–2.801
0.082
T stage (per level)
0.496
0.993–2.892
0.086
CCI** (per level)
0.175
0.973–1.459
0.091
*The percentual change in skeletal muscle index during neoadjuvant treatments
**Charlson comorbidity index

Discussion

Our findings suggest that loss of skeletal muscle mass during neoadjuvant treatment of EC is a marker of poor prognosis. Sarcopenia itself was not correlated with poorer oncological outcomes; however, its prevalence is high in this population and increases post-esophagectomy.
The amount of skeletal muscle lost during neoadjuvant treatment seems to predict a poorer prognosis. A median cut-off of − 2.98% produced significantly different 2-year overall survival rates as seen in Fig. 4a. To the authors’ best knowledge, this finding has not been previously reported in published literature, although similar findings have been reported in squamous cell carcinoma patients [15]. A previous study failed to show this correlation, but concluded that the amount of skeletal muscle lost during neoadjuvant therapy differed significantly between survivors and non-survivors, which was confirmed by our study [16]. Skeletal muscle wasting post-operatively has been associated with worse outcomes in thoracic esophageal cancer [28].
The progression of sarcopenia is not stopped by resection of the tumor according to our data as shown in Fig. 2. The prevalence of sarcopenia increases with time in follow-up. Similar findings have been reported in the literature [14]. Esophagectomy seems to affect the body composition of many patients. An interventional randomized study did not see statistically significant change in the weight of patients’ post-esophagectomy at 7 days, regardless of the method of nutritional support [29]. A previous study has shown that malnutrition and weight loss is common even years after EC surgery [30]. Whether this change in body mass post-esophagectomy contributes to the morbidity or mortality of operatively treated EC patients is unknown.
Sarcopenia was not correlated with worse overall survival or recurrence-free survival in our study. This finding contradicts many previous studies and is supported by some previous studies [1018]. Complication rates in any of the complication subgroups did not differ significantly between sarcopenic and non-sarcopenic groups. There was a statistically non-significant increase in operative bleeding with sarcopenic patients.
Our study has a number of limitations. The study is retrospective in nature without randomization. This increases the risk for systemic errors and selection bias. The number of patients included in the study is of adequate size taking into account the incidence of this disease, but statistical power is of concern especially in regard to complication rates and specific complication where the number of events is low or non-existent.

Conclusions

In conclusion, our study found an interesting correlation between the loss of skeletal muscle during neoadjuvant treatment and worse oncological outcomes in surgically treated EC patients. This finding should guide further investigation into the interventions for nutritional support in esophageal cancer and into the significance of indirect measurements of body composition in the prognosis of EC patients.

Acknowledgements

We thank Ms. Yvonne Sundström for her invaluable secretarial work and Dr. Kirsi Volmonen for introducing us to the radiologic methods used in our work.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
This study was approved by local medical ethical committees and was done according to the revised version of the Declaration of Helsinki (October 2008, Seoul).
Not applicable.

Competing interests

The authors declare that they have no competing interests.

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 Daly JM, Fry WA, Little AG, Winchester DP, McKee RF, Stewart AK, et al. Esophageal cancer: results of an American College of Surgeons Patient Care Evaluation Study. J Am Coll Surg. 2000;190(5):562–72. discussion 72-3CrossRefPubMed Daly JM, Fry WA, Little AG, Winchester DP, McKee RF, Stewart AK, et al. Esophageal cancer: results of an American College of Surgeons Patient Care Evaluation Study. J Am Coll Surg. 2000;190(5):562–72. discussion 72-3CrossRefPubMed
2.
Zurück zum Zitat Di Fiore F, Lecleire S, Pop D, Rigal O, Hamidou H, Paillot B, et al. Baseline nutritional status is predictive of response to treatment and survival in patients treated by definitive chemoradiotherapy for a locally advanced esophageal cancer. Am J Gastroenterol. 2007;102(11):2557–63.CrossRefPubMed Di Fiore F, Lecleire S, Pop D, Rigal O, Hamidou H, Paillot B, et al. Baseline nutritional status is predictive of response to treatment and survival in patients treated by definitive chemoradiotherapy for a locally advanced esophageal cancer. Am J Gastroenterol. 2007;102(11):2557–63.CrossRefPubMed
3.
Zurück zum Zitat van der Schaaf MK, Tilanus HW, van Lanschot JJB, Johar AM, Lagergren P, Lagergren J, et al. The influence of preoperative weight loss on the postoperative course after esophageal cancer resection. J Thorac Cardiov Sur. 2014;147(1):490–5.CrossRef van der Schaaf MK, Tilanus HW, van Lanschot JJB, Johar AM, Lagergren P, Lagergren J, et al. The influence of preoperative weight loss on the postoperative course after esophageal cancer resection. J Thorac Cardiov Sur. 2014;147(1):490–5.CrossRef
4.
Zurück zum Zitat Grotenhuis BA, Wijnhoven BP, Hotte GJ, van der Stok EP, Tilanus HW, van Lanschot JJ. Prognostic value of body mass index on short-term and long-term outcome after resection of esophageal cancer. World J Surg. 2010;34(11):2621–7.CrossRefPubMedPubMedCentral Grotenhuis BA, Wijnhoven BP, Hotte GJ, van der Stok EP, Tilanus HW, van Lanschot JJ. Prognostic value of body mass index on short-term and long-term outcome after resection of esophageal cancer. World J Surg. 2010;34(11):2621–7.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Blom R, Lagarde S, Klinkenbijl J, Busch O, MIV H. A high body mass index in esophageal cancer patients does not influence postoperative outcome or long-term survival. Ann Surg Oncol. 2012;19(3):766–71.CrossRefPubMed Blom R, Lagarde S, Klinkenbijl J, Busch O, MIV H. A high body mass index in esophageal cancer patients does not influence postoperative outcome or long-term survival. Ann Surg Oncol. 2012;19(3):766–71.CrossRefPubMed
6.
Zurück zum Zitat Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, et al. Sarcopenia: European consensus on definition and diagnosis: report of the European working group on sarcopenia in older people. Age Ageing. 2010;39(4):412–23.CrossRefPubMedPubMedCentral Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, et al. Sarcopenia: European consensus on definition and diagnosis: report of the European working group on sarcopenia in older people. Age Ageing. 2010;39(4):412–23.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Harimoto N, Shirabe K, Yamashita YI, Ikegami T, Yoshizumi T, Soejima Y, et al. Sarcopenia as a predictor of prognosis in patients following hepatectomy for hepatocellular carcinoma. Brit J Surg. 2013;100(11):1523–30.CrossRefPubMed Harimoto N, Shirabe K, Yamashita YI, Ikegami T, Yoshizumi T, Soejima Y, et al. Sarcopenia as a predictor of prognosis in patients following hepatectomy for hepatocellular carcinoma. Brit J Surg. 2013;100(11):1523–30.CrossRefPubMed
8.
Zurück zum Zitat Miyamoto Y, Baba Y, Sakamoto Y, Ohuchi M, Tokunaga R, Kurashige J, et al. Sarcopenia is a negative prognostic factor after curative resection of colorectal cancer. Ann Surg Oncol. 2015;22(8):2663–8.CrossRefPubMed Miyamoto Y, Baba Y, Sakamoto Y, Ohuchi M, Tokunaga R, Kurashige J, et al. Sarcopenia is a negative prognostic factor after curative resection of colorectal cancer. Ann Surg Oncol. 2015;22(8):2663–8.CrossRefPubMed
9.
Zurück zum Zitat Go SI, Park MJ, Song HN, Kang MH, Park HJ, Jeon KN, et al. Sarcopenia and inflammation are independent predictors of survival in male patients newly diagnosed with small cell lung cancer. Support Care Cancer. 2016;24(5):2075–84.CrossRefPubMed Go SI, Park MJ, Song HN, Kang MH, Park HJ, Jeon KN, et al. Sarcopenia and inflammation are independent predictors of survival in male patients newly diagnosed with small cell lung cancer. Support Care Cancer. 2016;24(5):2075–84.CrossRefPubMed
10.
Zurück zum Zitat Tamandl D, Paireder M, Asari R, Baltzer PA, Schoppmann SF, Ba-Ssalamah A. Markers of sarcopenia quantified by computed tomography predict adverse long-term outcome in patients with resected oesophageal or gastro-oesophageal junction cancer. Eur Radiol. 2016;26(5):1359–67.CrossRefPubMed Tamandl D, Paireder M, Asari R, Baltzer PA, Schoppmann SF, Ba-Ssalamah A. Markers of sarcopenia quantified by computed tomography predict adverse long-term outcome in patients with resected oesophageal or gastro-oesophageal junction cancer. Eur Radiol. 2016;26(5):1359–67.CrossRefPubMed
11.
Zurück zum Zitat Kudou K, Saeki H, Nakashima Y, Edahiro K, Korehisa S, Taniguchi D, et al. Prognostic significance of sarcopenia in patients with Esophagogastric junction cancer or upper gastric cancer. Ann Surg Oncol. 2017;24(7):1804–10. Kudou K, Saeki H, Nakashima Y, Edahiro K, Korehisa S, Taniguchi D, et al. Prognostic significance of sarcopenia in patients with Esophagogastric junction cancer or upper gastric cancer. Ann Surg Oncol. 2017;24(7):1804–10.
12.
Zurück zum Zitat Paireder M, Asari R, Kristo I, Rieder E, Tamandl D, Ba-Ssalamah A, et al. Impact of sarcopenia on outcome in patients with esophageal resection following neoadjuvant chemotherapy for esophageal cancer. Eur J Surg Oncol. 2017;43(2):478–84.CrossRefPubMed Paireder M, Asari R, Kristo I, Rieder E, Tamandl D, Ba-Ssalamah A, et al. Impact of sarcopenia on outcome in patients with esophageal resection following neoadjuvant chemotherapy for esophageal cancer. Eur J Surg Oncol. 2017;43(2):478–84.CrossRefPubMed
13.
Zurück zum Zitat Nishigori T, Okabe H, Tanaka E, Tsunoda S, Hisamori S, Sakai Y. Sarcopenia as a predictor of pulmonary complications after Esophagectomy for thoracic esophageal cancer. J Surg Oncol. 2016;113(6):678–84.CrossRefPubMed Nishigori T, Okabe H, Tanaka E, Tsunoda S, Hisamori S, Sakai Y. Sarcopenia as a predictor of pulmonary complications after Esophagectomy for thoracic esophageal cancer. J Surg Oncol. 2016;113(6):678–84.CrossRefPubMed
14.
Zurück zum Zitat Elliott JA, Doyle SL, Murphy CF, King S, Guinan EM, Beddy P, et al. Sarcopenia: prevalence, and impact on operative and oncologic outcomes in the multimodal management of locally advanced esophageal cancer. Ann Surg. 2017;266(5):822–30.CrossRefPubMed Elliott JA, Doyle SL, Murphy CF, King S, Guinan EM, Beddy P, et al. Sarcopenia: prevalence, and impact on operative and oncologic outcomes in the multimodal management of locally advanced esophageal cancer. Ann Surg. 2017;266(5):822–30.CrossRefPubMed
15.
Zurück zum Zitat Liu JJ, Motoyama S, Sato Y, Wakita A, Kawakita Y, Saito H, et al. Decreased skeletal muscle mass after neoadjuvant therapy correlates with poor prognosis in patients with esophageal cancer. Anticancer Res. 2016;36(12):6677–85.CrossRefPubMed Liu JJ, Motoyama S, Sato Y, Wakita A, Kawakita Y, Saito H, et al. Decreased skeletal muscle mass after neoadjuvant therapy correlates with poor prognosis in patients with esophageal cancer. Anticancer Res. 2016;36(12):6677–85.CrossRefPubMed
16.
Zurück zum Zitat Reisinger KW, Bosmans JWAM, Uittenbogaart M, Alsoumali A, Poeze M, Sosef MN, et al. Loss of skeletal muscle mass during Neoadjuvant chemoradiotherapy predicts postoperative mortality in esophageal cancer surgery. Ann Surg Oncol. 2015;22(13):4445–52.CrossRefPubMedPubMedCentral Reisinger KW, Bosmans JWAM, Uittenbogaart M, Alsoumali A, Poeze M, Sosef MN, et al. Loss of skeletal muscle mass during Neoadjuvant chemoradiotherapy predicts postoperative mortality in esophageal cancer surgery. Ann Surg Oncol. 2015;22(13):4445–52.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Grotenhuis BA, Shapiro J, van Adrichem S, de Vries M, Koek M, Wijnhoven BP, et al. Sarcopenia/muscle mass is not a prognostic factor for short- and long-term outcome after esophagectomy for cancer. World J Surg. 2016;40(11):2698–704.CrossRefPubMedPubMedCentral Grotenhuis BA, Shapiro J, van Adrichem S, de Vries M, Koek M, Wijnhoven BP, et al. Sarcopenia/muscle mass is not a prognostic factor for short- and long-term outcome after esophagectomy for cancer. World J Surg. 2016;40(11):2698–704.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Awad S, Tan BH, Cui H, Bhalla A, Fearon KCH, Parsons SL, et al. Marked changes in body composition following neoadjuvant chemotherapy for oesophagogastric cancer. Clin Nutr. 2012;31(1):74–7.CrossRefPubMed Awad S, Tan BH, Cui H, Bhalla A, Fearon KCH, Parsons SL, et al. Marked changes in body composition following neoadjuvant chemotherapy for oesophagogastric cancer. Clin Nutr. 2012;31(1):74–7.CrossRefPubMed
19.
Zurück zum Zitat Yip C, Goh V, Davies A, Gossage J, Mitchell-Hay R, Hynes O, et al. Assessment of sarcopenia and changes in body composition after neoadjuvant chemotherapy and associations with clinical outcomes in oesophageal cancer. Eur Radiol. 2014;24(5):998–1005.CrossRefPubMed Yip C, Goh V, Davies A, Gossage J, Mitchell-Hay R, Hynes O, et al. Assessment of sarcopenia and changes in body composition after neoadjuvant chemotherapy and associations with clinical outcomes in oesophageal cancer. Eur Radiol. 2014;24(5):998–1005.CrossRefPubMed
20.
Zurück zum Zitat Sheetz KH, Zhao L, Holcombe SA, Wang SC, Reddy RM, Lin J, et al. Decreased core muscle size is associated with worse patient survival following esophagectomy for cancer. Dis Esophagus. 2013;26(7):716–22.PubMedPubMedCentral Sheetz KH, Zhao L, Holcombe SA, Wang SC, Reddy RM, Lin J, et al. Decreased core muscle size is associated with worse patient survival following esophagectomy for cancer. Dis Esophagus. 2013;26(7):716–22.PubMedPubMedCentral
21.
Zurück zum Zitat Low DE, Alderson D, Cecconello I, Chang AC, Darling GE, D'Journo XB, et al. International consensus on standardization of data collection for complications associated with esophagectomy esophagectomy complications consensus group (ECCG). Ann Surg. 2015;262(2):286–94.CrossRefPubMed Low DE, Alderson D, Cecconello I, Chang AC, Darling GE, D'Journo XB, et al. International consensus on standardization of data collection for complications associated with esophagectomy esophagectomy complications consensus group (ECCG). Ann Surg. 2015;262(2):286–94.CrossRefPubMed
22.
Zurück zum Zitat Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJH, Nicolson M, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. New Engl J Med. 2006;355(1):11–20.CrossRefPubMed Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJH, Nicolson M, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. New Engl J Med. 2006;355(1):11–20.CrossRefPubMed
23.
Zurück zum Zitat Stiles BM, Altorki NK. Traditional techniques of esophagectomy. Surg Clin North Am. 2012;92(5):1249–63.CrossRefPubMed Stiles BM, Altorki NK. Traditional techniques of esophagectomy. Surg Clin North Am. 2012;92(5):1249–63.CrossRefPubMed
24.
Zurück zum Zitat Rice TW, Ishwaran H, Hofstetter WL, Kelsen DP, Apperson-Hansen C, Blackstone EH, et al. Recommendations for pathologic staging (pTNM) of cancer of the esophagus and esophagogastric junction for the 8th edition AJCC/UICC staging manuals. Dis Esophagus. 2016;29(8):897–905.CrossRefPubMedPubMedCentral Rice TW, Ishwaran H, Hofstetter WL, Kelsen DP, Apperson-Hansen C, Blackstone EH, et al. Recommendations for pathologic staging (pTNM) of cancer of the esophagus and esophagogastric junction for the 8th edition AJCC/UICC staging manuals. Dis Esophagus. 2016;29(8):897–905.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011;12(5):489–95.CrossRefPubMed Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011;12(5):489–95.CrossRefPubMed
26.
Zurück zum Zitat Prado CMM, Liefers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L, et al. Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study. Lancet Oncol. 2008;9(7):629–35.CrossRefPubMed Prado CMM, Liefers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L, et al. Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study. Lancet Oncol. 2008;9(7):629–35.CrossRefPubMed
27.
Zurück zum Zitat Vittinghoff E, Mc Culloch CE. Relaxing the rule of ten events per variable in logistic and cox regression. Am J Epidemiol. 2007;165(6):710–8.CrossRefPubMed Vittinghoff E, Mc Culloch CE. Relaxing the rule of ten events per variable in logistic and cox regression. Am J Epidemiol. 2007;165(6):710–8.CrossRefPubMed
28.
Zurück zum Zitat Mayanagi S, Tsubosa Y, Omae K, Niihara M, Uchida T, Tsushima T, et al. Negative impact of skeletal muscle wasting after neoadjuvant chemotherapy followed by surgery on survival for patients with thoracic esophageal cancer. Ann Surg Oncol. 2017;24(12):3741–7.CrossRefPubMedPubMedCentral Mayanagi S, Tsubosa Y, Omae K, Niihara M, Uchida T, Tsushima T, et al. Negative impact of skeletal muscle wasting after neoadjuvant chemotherapy followed by surgery on survival for patients with thoracic esophageal cancer. Ann Surg Oncol. 2017;24(12):3741–7.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Page RD, Oo AY, Russell GN, Pennefather SH. Intravenous hydration versus naso-jejunal enteral feeding after esophagectomy: a randomised study. Eur J Cardio-Thorac. 2002;22(5):666–72.CrossRef Page RD, Oo AY, Russell GN, Pennefather SH. Intravenous hydration versus naso-jejunal enteral feeding after esophagectomy: a randomised study. Eur J Cardio-Thorac. 2002;22(5):666–72.CrossRef
30.
Zurück zum Zitat Martin L, Lagergren J, Lindblad M, Rouvelas I, Lagergren P. Malnutrition after oesophageal cancer surgery in Sweden. Br J Surg. 2007;94(12):1496–500.CrossRefPubMed Martin L, Lagergren J, Lindblad M, Rouvelas I, Lagergren P. Malnutrition after oesophageal cancer surgery in Sweden. Br J Surg. 2007;94(12):1496–500.CrossRefPubMed
Metadaten
Titel
Loss of skeletal muscle mass during neoadjuvant treatments correlates with worse prognosis in esophageal cancer: a retrospective cohort study
verfasst von
Tommi Järvinen
Ilkka Ilonen
Juha Kauppi
Jarmo Salo
Jari Räsänen
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
World Journal of Surgical Oncology / Ausgabe 1/2018
Elektronische ISSN: 1477-7819
DOI
https://doi.org/10.1186/s12957-018-1327-4

Weitere Artikel der Ausgabe 1/2018

World Journal of Surgical Oncology 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.