Skip to main content
Erschienen in: Annals of Surgical Oncology 8/2017

Open Access 21.03.2017 | Gastrointestinal Oncology

Prognostic Value of Computed Tomography: Measured Parameters of Body Composition in Primary Operable Gastrointestinal Cancers

verfasst von: Douglas Black, MBChB, MSc, MRCS, Craig Mackay, MBChB, MRCS, PhD, George Ramsay, MBChB, MRCS, PhD, Zaid Hamoodi, MBChB, Shayanthan Nanthakumaran, MBChB, FRCS, MD, Kenneth G. M. Park, MBChB, FRCS, MD, Malcolm A. Loudon, MBChB, FRCS, MD, Colin H. Richards, MBChB, FRCS, MD

Erschienen in: Annals of Surgical Oncology | Ausgabe 8/2017

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

Previous reports suggest that body composition parameters can be used to predict outcomes for patients with gastrointestinal (GI) cancers. However, evidence for an association with long-term survival is conflicting, with much of the data derived from patients with advanced disease. This study examined the effect of body composition on survival in primary operable GI cancer.

Methods

Patients with resectable adenocarcinoma of the GI tract (esophagus, stomach, colon, rectum) between 2006 and 2014 were identified from a prospective database. Computed tomography (CT) scans were analyzed using a transverse section at L3 to calculate sex-specific body composition indices for skeletal muscle, visceral fat, and subcutaneous fat. Kaplan–Meier and log-rank analysis were used to compare unadjusted survival. Multivariate survival analyses were performed using a proportional hazards model.

Results

The study enrolled 447 patients (191 woman and 256 men) with esophagogastric (OG) (n = 108) and colorectal (CR) (n = 339) cancer. Body composition did not predict survival for the OG cancer patients. Among the CR cancer patients, survival was shorter for those with sarcopenia (p = 0.017) or low levels of subcutaneous fat (p = 0.005). Older age (p = 0.046) and neutrophilia (p = 0.013) were associated with sarcopenia in patients with CR. Tumor stage (p = 0.033), neutrophil count (p = 0.011), and hypoalbuminemia (p = 0.023) were associated with sarcopenia in OG cancer patients. In the multivariate analysis, no single measure of body composition was an independent predictor of reduced survival.

Conclusion

Sarcopenia and reduced subcutaneous adiposity are associated with reduced survival for patients with primary operable CR cancer. However, in this study, no parameter of body composition was an independent prognostic marker when considered with age, tumor stage, and systemic inflammation.
An increasing number of reports have suggested that body composition parameters may be used to predict outcomes for patients with cancer.17 In particular, depletion of skeletal muscle mass, termed “sarcopenia,” is widely reported to confer a poor prognosis for patients with tumors of the gastrointestinal (GI) tract, associated with an increased rate of postoperative complications2 and impaired response to chemotherapy.1 A smaller number of studies also have reported relationships between subcutaneous or visceral adiposity and outcomes for several tumor types, including esophageal,8 pancreatic,9 and colorectal cancers.10,11 The majority of these studies have used image analysis of computed tomography (CT) scans to measure parameters of body composition, and the accuracy of this technique is now widely accepted.12 This approach has considerable practical appeal because most patients with GI cancers undergo CT scanning as part of routine staging.
Despite consistent reports regarding short-term outcomes, the evidence that body composition parameters relate to long-term survival for patients with GI cancers has been conflicting. Studies to date have tended to focus exclusively on one parameter of body composition such as skeletal muscle mass,3,4,6 and much of the survival data has been derived from small cohorts of patients with locally advanced or metastatic disease.4,6,7
To investigate this topic further, the current study aimed to analyze CT-measured parameters of body composition in a large cohort of patients with primary operable GI cancers and to examine their relationships with long-term survival.

Methods

Patients with confirmed adenocarcinoma of the gastrointestinal tract (esophagus, stomach, colon, and rectum) who underwent surgical resection with curative intent between 1 January 2006 and 31 December 2014 at Aberdeen Royal Infirmary were identified from a prospectively maintained regional database. Of these patients, only those who had preoperative CT images stored in an electronic format suitable for image analysis were included in the study.
All tumors were confirmed histologically and staged according to conventional American Joint Committee on Cancer (AJCC) Tumor, Node, and Metastases (TNM) Classification (6th edition). Additional pathologic data, including the presence or absence of lymphovascular invasion, were recorded from reports issued at the time of resection.
Patient variables recorded retrospectively from medical records included age, sex, and preoperative blood results recorded within 30 days before surgery. Using local reference values, anemia was defined as hemoglobin concentrations lower than 130 g/L in males and lower than 115 g/L in females. The systemic inflammatory response was assessed by differential serum white cell count (total white cell count, neutrophil count, and lymphocyte count) in line with published thresholds.13,14
The standard oncologic treatment for potentially resectable esophagogastric (OG) cancers was three cycles of neoadjuvant combination chemotherapy with epirubicin, cisplatin and capecitabine (ECX), followed by surgical resection and adjuvant chemotherapy with the same agents. Colon cancer was generally managed by surgical resection followed by adjuvant combination (fluorouracil- and oxaliplatin-based) chemotherapy for patients with involved lymph nodes or other pathologic indicators of a poor prognosis such as extramural venous invasion (EMVI). Locally advanced or margin-threatened rectal cancer was treated with “long course” chemoradiotherapy followed by surgery 8–10 weeks later, with adjuvant chemotherapy offered selectively for those with a good or partial response to preoperative treatment. Individual regimens changed over time and were dependent on patient fitness, inclusion in contemporary clinical trials, and multidisciplinary team (MDT) preference.
To perform the body composition analysis, staging computed tomography (CT) scans were first accessed through the hospital’s Picture Archiving and Communication System (PACS). Preoperative staging CTs before the start of neoadjuvant therapy were selected. A single slice at the level of the third lumbar vertebra (L3) was analyzed using medical imaging software (ImageJ; The National Institutes of Health, Washington, MD, USA; version 1.47), and the total fat area (cm2), subcutaneous fat area (cm2), visceral fat area (cm2), and skeletal muscle area (cm2) were measured using accepted Hounsfield unit (HU) thresholds (adipose tissue, −190 to −30; skeletal muscle, −29 to +150). Finally, each parameter was normalized for patient stature and designated as total fat index (cm2/m2), subcutaneous fat index (cm2/m2), visceral fat index (cm2/m2), and skeletal muscle index (cm2/m2) in line with accepted methodology.15,16 Sarcopenia was defined as a skeletal muscle index lower than 43 cm2/m2 for males and lower than 41 cm2/m2 for females using previously published cutoff values.6
The primary end point of the study was overall survival, which was measured in months from the date of surgery to the date of death from any cause. The date of death was obtained from patients’ electronic medical records. All survival analyses were performed after exclusion of 30-day postoperative deaths. Ethical guidance was sought from the regional Caldicott Guardian, who confirmed that the study fulfilled the criteria of a clinical audit, negating the requirement for further ethical committee approval.

Statistical Analysis

All variables were grouped according to clinically relevant or previously published thresholds. All statistical tests were two-sided, and a p value lower than 0.05 was considered to indicate statistical significance. χ 2 and Mann–Whitney U tests were used to compare clinical characteristics between groups. Kaplan–Meier analysis and the log-rank test were used to compare unadjusted survival differences. Uni- and multivariate survival analyses were performed using a Cox proportional hazards model. Statistical analysis was performed using SPSS, version 22 (SPSS, Chicago, IL, USA).

Results

During the study period, 608 patients with primary operable gastrointestinal cancers who had undergone surgical resection with curative intent were identified. Of these patients, 161 were excluded from the study (108 patients did not have a documented height and weight; 34 patients did not have CT images suitable for analysis; and 19 patients underwent a palliative procedure after more extensive disease had been diagnosed intraoperatively), leaving 447 patients (191 women and 256 men) included in the final analysis. A flow diagram of the study selection process is shown in Fig. 1.
The baseline clinicopathologic characteristics and body composition parameters of the cohort are shown in Table 1. Of the 447 patients included in the study, 108 had eophagogastric (OG) cancers (43 esophageal; 65 gastric), and 339 had colorectal (CR) cancers (253 colonic; 86 rectal). More than 40% of the patients were anemic preoperatively, and 18% exhibited a systemic inflammatory response, as evidenced by an elevated neutrophil count. There were significant differences between upper GI and colorectal cancer in terms of age (p < 0.001), sex (p = 0.003), and lymphovascular invasion (p < 0.001).
Table 1
Clinical, pathologic, and body composition parameters of the included patients
Variable
All patients (n = 447) n (%)
OG cancer n (%)
CR cancer n (%)
p valuea
Age (years)
 ≤65
133 (30)
46 (43)
87 (26)
<0.001
 65–74
148 (33)
40 (37)
108 (32)
 
 ≥75
166 (37)
22 (20)
144 (42)
 
Sex
 Female
191 (43)
33 (31)
158 (47)
0.003
 Male
256 (57)
74 (69)
181 (53)
 
Neoadjuvant therapy
 No
316 (71)
43 (40)
273 (81)
<0.001
 Yes
131 (29)
65 (60)
66 (19)
 
Adjuvant therapy
 No
343 (77)
66 (61)
277 (82)
<0.001
 Yes
104 (23)
42 (39)
62 (18)
 
TNM stage
 1
88 (20)
30 (28)
58 (17)
0.052
 2
196 (44)
43 (40)
153 (45)
 
 3
163 (36)
35 (32)
128 (38)
 
Lymphovascular invasion
 Yes
111 (25)
51 (47)
60 (18)
<0.001
 No
336 (75)
57 (53)
279 (82)
 
Anemiab,c
 Yes
186 (42)
44 (42)
142 (42)
0.873
 No
255 (58)
62 (58)
193 (58)
 
White cell count (× 109/L)c
 <8.5
280 (63)
70 (66)
210 (63)
0.711
 8.5–11
109 (25)
23 (22)
86 (26)
 
 >11
52 (12)
13 (12)
39 (12)
 
Neutrophil count (× 109/L)c
 <7.5
362 (82)
87 (82)
275 (82)
0.997
 ≥7.5
79 (18)
19 (18)
60 (18)
 
Lymphocyte count (× 109/L)c
 <1.0
94 (21)
18 (17)
76 (23)
0.211
 ≥1.0
347 (79)
88 (83)
259 (77)
 
Albumin (g/L)c
 ≥35
387 (88)
89 (84)
298 (89)
0.172
 <35
54 (12)
17 (16)
37 (11)
 
Subcutaneous fat index (cm2/m2)
 Median
66.2
64.9
70.0
0.114
 Range
200.5
193.4
191.9
 
 Low d
152 (34)
38 (35)
114 (34)
 
 Mediumd
148 (33)
33 (31)
115 (34)
 
 Highd
147 (33)
37 (34)
110 (32)
 
Visceral fat index (cm2/m2)
 Median
61.3
63.4
61.0
0.886
 Range
198.4
155.0
198.4
 
 Lowe
152 (34)
38 (35)
114 (34)
 
 Mediume
146 (33)
38 (35)
108 (32)
 
 Highd
149 (33)
32 (30)
117 (35)
 
Skeletal muscle index (cm2/m2)
 Median
47.4
47.7
47.3
0.888
 Range
80.1
44.2
80.1
 
 Sarcopeniaf
104 (23)
23 (21)
81 (24)
 
 Normal
343 (77)
85 (79)
258 (76)
 
OG esophagogastric, CR colorectal, TNM tumor-node-metastasis
a p Values represent X 2 tests for a linear trend in categorical variables and Mann–Whitney U tests for continuous variables
bAnemia is defined as <13 g/dL in males, <11.5 g/dL in females
cData are missing in six cases
dSex-specific tertiles for subcutaneous fat index
eSex-specific tertiles for visceral fat index
fSarcopenia is defined as <43 cm2/m2 in males and <41 cm2/m2 in females
To account for the differences in body composition distribution between the men and women, the subcutaneous fat index and the visceral fat index were classified into sex-specific tertiles, whereas previously published sex-specific cutoff values for skeletal muscle index were used to define sarcopenia in the men (<43 cm2/m2) and the women (<41 cm2/m2). According to these definitions, 23 patients (21%) with esophagogastric cancer and 81 patients (24%) with colorectal cancer showed evidence of sarcopenia on their staging CT scan (Table 1).
Figure 2 shows the relationships between body composition parameters and long-term survival. Levels of subcutaneous fat, visceral fat, and skeletal muscle did not influence overall survival for the patients with esophagogastric cancer. Among the patients with colorectal cancer, survival was significantly shorter for those with low levels of subcutaneous fat (p = 0.005, log-rank test) or evidence of sarcopenia (p = 0.017, log-rank test).
To investigate these relationships further, the associations between body composition and clinicopathologic variables were examined. An association between sarcopenia and advanced T stage (p = 0.033), elevated neutrophil count (p = 0.011), and hypoalbuminemia (p = 0.023) was observed in the patients with esophagogastric cancer (Table 2). In the patients with colorectal cancer, associations between sarcopenia and older age (p = 0.046) and elevated neutrophil count (p = 0.026) were demonstrated. Similar relationships were seen between low levels of subcutaneous fat and older age (p < 0.001) and elevated neutrophil count (p = 0.013) (Table 3).
Table 2
Associations between body composition parameters and clinicopathologic variables for patients with esophagogastric cancer
Variable
Subcutaneous fat index
p valuea
Visceral fat index
p valuea
Skeletal muscle index
p valuea
Low n (%)
Medium n (%)
High n (%)
Low n (%)
Medium n (%)
High n (%)
Normal n (%)
Sarcopenia n (%)
Age (years)
 ≤64
13 (34)
15 (45)
18 (49)
0.560
20 (53)
14 (37)
12 (38)
0.533
37 (44)
9 (39)
0.744
 65–74
16 (42)
10 (30)
14 (38)
 
11 (29)
17 (45)
12 (38)
 
32 (38)
8 (35)
 
 ≥75
9 (24)
8 (24)
5 (14)
 
7 (18)
7 (18)
8 (25)
 
16 (19)
6 (26)
 
Tumour (T) stage
 0/1
8 (21)
5 (15)
9 (24)
0.742
6 (16)
7 (18)
9 (28)
0.534
21 (25)
1 (4)
0.033
 2
4 (11)
9 (27)
5 (14)
 
5 (13)
9 (24)
4 (13)
 
13 (15)
5 (22)
 
 3
22 (58)
17 (52)
20 (54)
 
23 (61)
18 (47)
18 (56)
 
47 (55)
12 (52)
 
 4
4 (11)
2 (6)
3 (8)
 
4 (11)
4 (11)
1 (3)
 
4 (5)
5 (22)
 
Nodal (N) stage
 0
17 (45)
13 (39)
20 (54)
0.776
13 (34)
15 (39)
22 (69)
0.103
41 (48)
9 (39)
0.362
 1
12 (32)
13 (39)
10 (27)
 
14 (37)
15 (39)
6 (19)
 
29 (34)
6 (26)
 
 2
9 (24)
7 (21)
7 (19)
 
11 (29)
8 (21)
4 (13)
 
15 (18)
8 (35)
 
TNM stage
 I
9 (24)
10 (30)
11 (30)
0.846
6 (16)
11 (29)
13 (41)
0.136
27 (32)
3 (13)
0.175
 II
16 (42)
11 (33)
16 (43)
 
15 (39)
16 (42)
12 (38)
 
33 (39)
10 (43)
 
 III
13 (34)
12 (36)
10 (27)
 
17 (45)
11 (29)
7 (22)
 
25 (29)
10 (43)
 
Neoadjuvant therapy
 Yes
18 (47)
8 (24)
17 (46)
0.090
9 (24)
20 (53)
14 (44)
0.031
34 (40)
9 (39)
0.940
 No
20 (53)
25 (76)
20 (54)
 
29 (76)
18 (47)
18 (56)
 
51 (60)
14 (61)
 
Adjuvant therapy
 Yes
17 (45)
10 (30)
15 (41)
0.446
18 (47)
13 (34)
11 (34)
0.412
30 (35)
12 (52)
0.141
 No
21 (55)
23 (70)
22 (59)
 
20 (53)
25 (66)
21 (66)
 
55 (65)
11 (48)
 
Lymphovascular invasion
 Yes
17 (45)
13 (39)
21 (57)
0.324
21 (55)
16 (42)
14 (44)
0.463
38 (45)
13 (57)
0.314
 No
21 (55)
20 (61)
16 (43)
 
17 (45)
22 (58)
18 (56)
 
47 (55)
10 (43)
 
Anemiab
 Yes
17 (45)
14 (45)
13 (35)
0.621
19 (50)
11 (30)
14 (45)
0.181
33 (39)
11 (50)
0.364
 No
21 (55)
17 (55)
24 (65)
 
19 (50)
26 (70)
17 (55)
 
51 (61)
11 (50)
 
White cell count (×109/L)
 <8.5
23 (61)
21 (68)
26 (70)
0.840
24 (63)
23 (62)
23 (74)
0.736
61 (73)
9 (41)
0.011
 8.5–11
10 (26)
7 (23)
6 (16)
 
8 (21)
10 (27)
5 (16)
 
16 (19)
7 (32)
 
 >11
5 (13)
3 (10)
5 (14)
 
6 (16)
4 (11)
3 (10)
 
7 (8)
6 (27)
 
Neutrophil count (×109/L)
 <7.5
29 (76)
26 (84)
32 (86)
0.493
30 (79)
31 (84)
26 (84)
0.821
73 (87)
14 (64)
0.011
 ≥7.5
9 (24)
5 (16)
5 (14)
 
8 (21)
6 (16)
5 (16)
 
11 (13)
8 (36)
 
Lymphocyte count (× 109/L)
 <1.0
7 (18)
6 (19)
5 (14)
0.781
7 (18)
6 (16)
5 (16)
0.957
14 (17)
4 (18)
0.866
 ≥1.0
31 (82)
25 (81)
32 (86)
 
31 (82)
31 (84)
26 (84)
 
70 (83)
18 (82)
 
Albumin (g/L)
 ≥35
31 (82)
24 (77)
34 (92)
0.238
31 (82)
30 (81)
28 (90)
0.517
74 (88)
15 (68)
0.023
 <35
7 (18)
7 (23)
3 (8)
 
7 (18)
7 (19)
3 (10)
 
10 (12)
7 (32)
 
TNM tumor-node-metastasis
a p Values represent X 2 tests for a linear trend in categorical variables and Mann–Whitney U tests for continuous variables
bAnemia is defined as <13 g/dL in males, <11.5 g/dL in females
Table 3
The associations between body composition parameters and clinicopathologic variables in patients with colorectal cancer
Variable
Subcutaneous fat index
p valuea
Visceral fat index
p valuea
Skeletal muscle index
p Valuea
Low n (%)
Medium n (%)
High n (%)
Low n (%)
Medium n (%)
High n (%)
Normal n (%)
Sarcopenia n (%)
Age (years)
 ≤64
25 (22)
23 (20)
39 (35)
<0.001
43 (38)
21 (19)
23 (20)
<0.001
70 (27)
17 (21)
0.046
 65–74
27 (23)
39 (34)
42 (38)
18 (16)
39 (36)
51 (44)
88 (34)
20 (25)
 ≥75
62 (54)
53 (46)
29 (26)
53 (46)
48 (44)
43 (37)
100 (39)
44 (54)
Tumour (T) stage
 0/1
9 (8)
5 (4)
8 (7)
0.432
7 (6)
10 (9)
5 (4)
0.219
17 (7)
5 (6)
0.118
 2
12 (11)
13 (11)
20 (18)
13 (11)
13 (12)
19 (16)
40 (16)
5 (6)
 3
72 (63)
76 (66)
69 (63)
73 (64)
74 (69)
70 (60)
164 (64)
53 (65)
 4
21 (18)
21 (18)
13 (12)
21 (18)
11 (10)
23 (20)
37 (14)
18 (22)
Nodal (N) stage
 0
69 (61)
65 (57)
77 (70)
0.099
69 (61)
62 (57)
80 (68)
0.482
168 (65)
43 (53)
0.099
 1
24 (21)
35 (30)
19 (17)
29 (25)
27 (25)
22 (19)
57 (22)
21 (26)
 2
21 (18)
15 (13)
14 (13)
16 (14)
19 (18)
15 (13)
33 (13)
17 (21)
TNM stage
 1
19 (17)
13 (11)
26 (24)
0.094
16 (14)
18 (17)
24 (21)
0.398
50 (19)
8 (10)
0.058
 2
50 (44)
52 (45)
51 (46)
53 (46)
44 (41)
56 (48)
118 (46)
35 (43)
 3
45 (39)
50 (43)
33 (30)
45 (39)
46 (43)
37 (32)
90 (35)
38 (47)
Neoadjuvant therapy
 Yes
89 (78)
92 (80)
92 (84)
0.566
88 (77)
88 (81)
97 (83)
0.524
204 (79)
69 (85)
0.225
 No
25 (22)
23 (20)
18 (16)
26 (23)
20 (19)
20 (17)
54 (21)
12 (15)
Adjuvant therapy
 Yes
15 (13)
22 (19)
25 (23)
0.173
26 (23)
16 (15)
20 (17)
0.281
49 (19)
13 (16)
0.550
 No
99 (87)
93 (81)
85 (77)
88 (77)
92 (85)
97 (83)
209 (81)
68 (84)
Lymphovascular invasion
 Yes
23 (20)
20 (17)
17 (15)
0.648
20 (18)
18 (17)
22 (19)
0.914
45 (17)
15 (19)
0.825
 No
91 (80)
95 (83)
93 (85)
94 (82)
90 (83)
95 (81)
213 (83)
66 (81)
Anemiab
 Yes
53 (47)
53 (46)
36 (33)
0.069
49 (44)
44 (41)
49 (42)
0.925
105 (41)
37 (46)
0.423
 No
60 (53)
61 (54)
72 (67)
63 (56)
63 (59)
67 (58)
150 (59)
43 (54)
White cell count (×109/L)
 <8.5
64 (57)
73 (64)
73 (68)
0.241
76 (68)
64 (60)
70 (60)
0.110
162 (64)
48 (60)
0.561
 8.5–11
31 (27)
32 (28)
23 (21)
24 (21)
25 (23)
37 (32)
66 (26)
20 (25)
 >11
18 (16)
9 (8)
12 (11)
12 (11)
18 (17)
9 (8)
27 (11)
12 (15)
Neutrophil count (×109/L)
 <7.5
83 (73)
99 (87)
93 (86)
0.013
94 (84)
85 (79)
96 (83)
0.669
216 (85)
59 (74)
0.026
 ≥7.5
30 (27)
15 (13)
15 (14)
18 (16)
22 (21)
20 (17)
39 (15)
21 (26)
Lymphocyte count (×109/L)
 <1.0
31 (27)
23 (20)
22 (20)
0.334
28 (25)
29 (27)
19 (16)
0.125
57 (22)
19 (24)
0.795
 ≥1.0
82 (73)
91 (80)
86 (80)
84 (75)
78 (73)
97 (84)
198 (78)
61 (76)
Albumin (g/L)
 ≥35
97 (86)
101 (89)
100 (93)
0.275
98 (88)
95 (89)
105 (91)
0.766
229 (90)
69 (86)
0.376
 <35
16 (14)
13 (11)
8 (7)
14 (13)
12 (11)
11 (9)
26 (10)
11 (14)
TNM, tumor-node-metastasis
a p values represent X 2 tests for a linear trend in categorical variables and Mann–Whitney U tests for continuous variables
bAnemia is defined as <13 g/dL in males, <11.5 g/dL in females
Finally, logistic regression analyses were used to examine whether survival relationships were independent of established clinicopathologic risk factors. During the follow-up period, 213 patients died, leaving 234 were alive at the date of censor (31 March 2015). The median follow-up period for the survivors was 62 months (range 3–105 months).
In the multivariate analysis, the only independent predictor of long-term survival for the patients with esophagogastric cancer was tumor stage [hazard ratio (HR) 2.78; p < 0.001] (Table 4). For the patients with colorectal cancer, advanced tumor stage (HR 1.67; p < 0.001), lymphovascular invasion (HR 2.61; p < 0.001), and elevated neutrophil count (HR 1.76; p = 0.005) were independently associated with reduced overall survival (Table 5). No single measure of body composition was an independent predictor of reduced survival for patients with primary operable GI cancer.
Table 4
Multivariate analysis of the relationships between body composition parameters and overall survival for patients with esophagogastric cancer
Variables
No. of patients
No. of deaths n (%)
Univariate analysis
Multivariate analysis
HR
(95% CI)
p value
HR
(95% CI)
p value
Age (years)
 ≤65
46
23 (33)
1.148
0.832–1.584
0.402
1.578
1.03–2.417
NS
 65–74
40
27 (40)
 ≥75
22
12 (35)
Sex
 Female
33
18 (35)
1.026
0.593–1.777
0.926
1.145
0.605–2.167
0.667
 Male
75
44 (37)
TNM stage
 1
30
5 (14)
2.390
1.681–3.398
<0.001
2.782
1.766–4.382
<0.001
 2
43
30 (41)
 3
35
27 (44)
Neoadjuvant therapy
 Yes
43
20 (32)
1.579
0.926–2.691
0.093
2.111
1.015–4.388
NS
 No
65
42 (39)
Adjuvant therapy
 Yes
42
23 (35)
0.719
0.429–1.206
0.719
0.403
0.22–0.737
NS
 No
66
39 (37)
Lymphovascular invasion
 Yes
51
34 (40)
1.722
1.037–2.859
0.036
0.814
0.425–1.560
NS
 No
57
28 (33)
Neutrophil count (×109/L)
 <7.5
87
48 (36)
1.033
0.549–1.946
0.919
1.048
0.517–2.124
NS
 ≥7.5
19
12 (39)
Subcutaneous fat index (cm2/m2)
 High
38
24 (39)
0.912
0.678–1.228
0.545
0.934
0.627–1.39
NS
 Medium
33
19 (37)
 Low
37
19 (34)
Visceral fat index (cm2/m2)
 High
38
26 (41)
0.786
0.571–1.083
0.141
0.738
0.473–1.152
NS
 Medium
38
21 (36)
 Low
32
15 (32)
Skeletal muscle index (cm2/m2)
 Normal
85
48 (36)
1.165
0.642–2.114
0.616
0.761
0.351–1.649
NS
 Sarcopenia
23
14 (38)
HR hazard ratio, CI confidence interval, TNM tumor-node-metastasis
Table 5
Multivariate analysis of the relationships between body composition parameters and overall survival for patients with colorectal cancer
Variable
No. of patients
No. of deaths n (%)
Univariate analysis
Multivariate analysis
HR
(95% CI)
p value
HR
(95% CI)
p Value
Age (years)
 ≤65
87
36 (29)
1.197
0.976–1.467
0.084
1.099
0.871–1.386
NS
 65–74
108
41 (28)
 ≥75
144
74 (34)
Sex
 Female
158
74 (32)
0.856
0.622–1.176
0.339
0.994
0.703–1.405
NS
 Male
181
77 (30)
TNM stage
 1
58
12 (17)
1.921
1.503–2.455
<0.001
1.667
1.263–2.2
<0.001
 2
153
64 (29)
 3
128
75 (37)
Neoadjuvant therapy
 Yes
273
120 (31)
1.095
0.738–1.626
0.651
1.444
0.946–2.203
NS
 No
66
31 (32)
Adjuvant therapy
 Yes
62
28 (31)
0.979
0.649–1.476
0.976
0.764
0.479–1.218
NS
 No
277
123 (31)
Lymphovascular invasion
 Yes
60
48 (44)
3.663
2.585–5.190
<0.001
2.606
1.764–3.851
<0.001
 No
279
103 (27)
Neutrophil count (× 109/L)
 <7.5
275
108 (28)
2.556
1.780–3.669
<0.001
1.760
1.182–2.62
0.005
 ≥7.5
60
41 (41)
Subcutaneous fat index (cm2/m2)
 High
114
62 (35)
0.720
0.589–0.880
0.001
0.846
0.662–1.08
NS
 Medium
115
52 (31)
 Low
110
37 (25)
Visceral fat index (cm2/m2)
 High
114
56 (33)
0.873
0.718–1/061
0.172
1.00
0.796–1.256
NS
 Medium
108
48 (31)
 Low
117
47 (29)
Skeletal muscle index (cm2/m2)
 Normal
258
107 (29)
1.527
1.075–2.170
0.018
1.211
0.818–1.795
NS
 Sarcopenia
81
44 (35)
HR hazard ratio, CI confidence interval, TNM tumor-node-metastasis

Discussion

The results of the current study show that CT measures of body composition, particularly sarcopenia and reduced levels of subcutaneous fat, are associated with shorter survival for patients with primary operable colorectal cancer, but not for patients with esophagogastric cancer. Furthermore, strong associations exist between these parameters and other indicators of poor outcome such as advanced age and elevated systemic inflammatory response. However, when body composition parameters were analyzed in a multivariate model, no single measure was found to have independent predictive value for patients with either esophagogastric or colorectal cancer.
To our knowledge, this is one of the largest studies to investigate the impact of body composition on long-term survival of patients with operable GI cancers. Although associations between sarcopenia and colorectal cancer outcomes have been reported previously,3,4,6,7,17,18 the results have been inconsistent. Most previous studies have included a high proportion of patients with advanced disease, whereas the current study focused specifically on patients with operable disease.
A systematic review by Malietzis et al.2 evaluated the role of body composition in predicting outcomes for patients with colorectal cancer and concluded that whereas evidence was consistent that sarcopenia is associated with poorer short-term outcomes, including excess chemotherapy toxicity1719 and an increased risk of surgical complications,20,21 the evidence for a relationship with long-term survival was less robust. Indeed, the reviewers identified only one study of 196 patients, all of whom had metastatic disease,7 in which sarcopenia had a detrimental effect on survival.
Not included in the aforementioned review but widely referenced as demonstrating the prognostic value of skeletal muscle depletion for cancer patients, a study by Martin et al.6 analyzed the body composition parameters of 1473 patients with respiratory and GI cancers. The authors reported that a predictive model composed entirely of body composition variables (weight loss, skeletal muscle depletion, and muscle attenuation) was superior to conventional prognostic markers, including cancer stage. However, more than 50% of the patients studied had metastatic disease, and our results suggest that their findings may not be applicable to patients with primary operable cancers.
It is clear from our own appraisal of the literature and the conclusions of recent reviews3,4 that the question whether sarcopenia has prognostic value for patients with GI malignancies is being hampered by study heterogeneity. Despite the volume of published work, there still is no standard definition of CT-based assessments of skeletal muscle mass.
Although a number of different cutoff values have been proposed,7,17,22 we chose to use a skeletal muscle index lower than 43 cm2/m2 for men and lower than 41 cm2/m2 for women to define sarcopenia. These values were proposed by the largest published dataset to document the body composition of patients with cancer6 and have been validated in at least one external cohort.7
It must be emphasised that discrepancies in the thresholds used to define sarcopenia have led to considerable variation in the proportion of patients reported to be “sarcopenic” in the aforementioned studies. For example, the study by van Vledder et al.,7 using one threshold, reported that 19% of patients with colorectal liver metastases have sarcopenia, whereas Martin et al.,6 using different definitions, reported that 53% of women and 31% of men are sarcopenic. Using the latter definitions, our levels of sarcopenia were considerably lower (23%), but all the patients in our cohort were undergoing curative surgery, whereas their study contained a large number of patients with metastatic disease. Similarly, the assessment of subcutaneous and visceral adiposity has been undertaken using a variety of methods including dichotomous cutoff values,23,24 continuous parameters,25 and visceral-to-subcutaneous ratios.26
Given this variability and with no single method yet validated, we chose to use sex-specific tertiles to assess adiposity. It may be that using an alternative technique would have yielded different results, but we believe our approach was a rational way of demonstrating any survival effect.
One noteworthy finding from the current study was the association between depleted levels of skeletal muscle and subcutaneous fat and an elevation of the systemic inflammatory response in patients with colorectal cancer. The neutrophil count was used as a marker of systemic inflammation because findings previously showed it to be the most reliable prognostic component of the white cell count.27
In experimental models, pro-inflammatory cytokines such as interleukin-1 (IL-1), IL-6, and tumor necrosis factor-α (TNF) have been shown to play a key role in both anorexia and skeletal muscle proteolysis,28 but the relationships between systemic inflammation and changes in body composition in cancer patients are less well understood. Good evidence currently shows that systemic inflammation is universally associated with poor short- and long-term outcomes in a variety of solid organ tumor types,2931 and an association with skeletal muscle wasting may offer one explanation for the unfavorable outcomes observed in sarcopenic patients.14,32,33 In the current study, despite no significant difference in the prevalence of sarcopenia between cancer types, a clear relationship was demonstrated between sarcopenia and survival in colorectal cancer but not in upper GI cancers. Further work is needed to clarify the relationships between tumor biology, inflammatory mediators, and parameters of body composition.
The current study had a number of limitations. The retrospective nature of the data collection meant that contemporary records of patients’ height were missing in a number of cases. As a result, body composition indices could not be normalized for stature, thereby limiting the size of the cohort. Similarly, preoperative weight was poorly documented in the medical notes, so conventional parameters of body composition such as body mass index (BMI) could not be calculated. However, preoperative CT images were available for almost all the patients, and we believe that both the size and maturity of the cohort mean our results are likely to be reliable.
In summary, the current study showed that sarcopenia and reduced subcutaneous adiposity are associated with shorter overall survival for patients with primary operable colorectal cancer. However, no parameter of body composition was an independent prognostic marker when considered with age, tumor stage, and systemic inflammatory response. No relationships between body composition and overall survival were observed in patients with esophagogastric cancers.

Acknowledgment

Professor Graeme Murray, Department of Pathology, University of Aberdeen provided us access to the colorectal cancer pathology databases from which the colorectal component of the research was based.

Conflict of interest

There are no conflicts of interest.
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.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Yip C, Dinkel C, Mahajan A, Siddique M, Cook GJR, Goh V. Imaging body composition in cancer patients: visceral obesity, sarcopenia and sarcopenic obesity may impact on clinical outcome. Insights Imaging. 2015;6:489–97.CrossRefPubMedPubMedCentral Yip C, Dinkel C, Mahajan A, Siddique M, Cook GJR, Goh V. Imaging body composition in cancer patients: visceral obesity, sarcopenia and sarcopenic obesity may impact on clinical outcome. Insights Imaging. 2015;6:489–97.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Malietzis G, Aziz O, Bagnall NM, Johns N, Fearon KC, Jenkins JT. The role of body composition evaluation by computerized tomography in determining colorectal cancer treatment outcomes: a systematic review. Eur J Surg Oncol. 2015;41:186–96.CrossRefPubMed Malietzis G, Aziz O, Bagnall NM, Johns N, Fearon KC, Jenkins JT. The role of body composition evaluation by computerized tomography in determining colorectal cancer treatment outcomes: a systematic review. Eur J Surg Oncol. 2015;41:186–96.CrossRefPubMed
3.
Zurück zum Zitat Wagner D, DeMarco MM, Amini N, et al. Role of frailty and sarcopenia in predicting outcomes among patients undergoing gastrointestinal surgery. World J Gastrointest Surg. 2016;8:27–40.CrossRefPubMedPubMedCentral Wagner D, DeMarco MM, Amini N, et al. Role of frailty and sarcopenia in predicting outcomes among patients undergoing gastrointestinal surgery. World J Gastrointest Surg. 2016;8:27–40.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Levolger S, van Vugt JLA, de Bruin RWF, et al. Systematic review of sarcopenia in patients operated on for gastrointestinal and hepatopancreatobiliary malignancies. Br J Surg. 2015;102:1448–58.CrossRefPubMed Levolger S, van Vugt JLA, de Bruin RWF, et al. Systematic review of sarcopenia in patients operated on for gastrointestinal and hepatopancreatobiliary malignancies. Br J Surg. 2015;102:1448–58.CrossRefPubMed
5.
Zurück zum Zitat Veasey Rodrigues H, Baracos VE, Wheler JJ, et al. Body composition and survival in the early clinical trials setting. Eur J Cancer. 2013;49:3068–75.CrossRefPubMed Veasey Rodrigues H, Baracos VE, Wheler JJ, et al. Body composition and survival in the early clinical trials setting. Eur J Cancer. 2013;49:3068–75.CrossRefPubMed
6.
Zurück zum Zitat Martin L, Birdsell L, Macdonald N, et al. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol. 2013;31:1539–47.CrossRefPubMed Martin L, Birdsell L, Macdonald N, et al. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol. 2013;31:1539–47.CrossRefPubMed
7.
Zurück zum Zitat van Vledder MG, Levolger S, Ayez N, Verhoef C, Tran TCK, Ijzermans JNM. Body composition and outcome in patients undergoing resection of colorectal liver metastases. Br J Surg. 2012;99:550–7.CrossRefPubMed van Vledder MG, Levolger S, Ayez N, Verhoef C, Tran TCK, Ijzermans JNM. Body composition and outcome in patients undergoing resection of colorectal liver metastases. Br J Surg. 2012;99:550–7.CrossRefPubMed
8.
Zurück zum Zitat Harada K, Baba Y, Ishimoto T, et al. Low visceral fat content is associated with poor prognosis in a database of 507 upper gastrointestinal cancers. Ann Surg Oncol. 2015;22:3946–53.CrossRefPubMed Harada K, Baba Y, Ishimoto T, et al. Low visceral fat content is associated with poor prognosis in a database of 507 upper gastrointestinal cancers. Ann Surg Oncol. 2015;22:3946–53.CrossRefPubMed
9.
Zurück zum Zitat Balentine CJ, Enriquez J, Fisher W, et al. Intra-abdominal fat predicts survival in pancreatic cancer. J Gastrointest Surg. 2010;14:1832–7.CrossRefPubMed Balentine CJ, Enriquez J, Fisher W, et al. Intra-abdominal fat predicts survival in pancreatic cancer. J Gastrointest Surg. 2010;14:1832–7.CrossRefPubMed
10.
Zurück zum Zitat Moon HG, Ju YT, Jeong CY, et al. Visceral obesity may affect oncologic outcome in patients with colorectal cancer. Ann Surg Oncol. 2008;15:1918–22.CrossRefPubMed Moon HG, Ju YT, Jeong CY, et al. Visceral obesity may affect oncologic outcome in patients with colorectal cancer. Ann Surg Oncol. 2008;15:1918–22.CrossRefPubMed
11.
Zurück zum Zitat Lee CS, Murphy DJ, McMahon C, et al. Visceral adiposity is a risk factor for poor prognosis in colorectal cancer patients receiving adjuvant chemotherapy. J Gastrointest Cancer. 2015;46:243–50.CrossRefPubMed Lee CS, Murphy DJ, McMahon C, et al. Visceral adiposity is a risk factor for poor prognosis in colorectal cancer patients receiving adjuvant chemotherapy. J Gastrointest Cancer. 2015;46:243–50.CrossRefPubMed
12.
Zurück zum Zitat Mourtzakis M, Prado CMM, Lieffers JR, et al. A practical and precise approach to quantification of body composition in cancer patients using computed tomography images acquired during routine care. Appl Physiol Nutr Metab. 2008;33:997–1006.CrossRefPubMed Mourtzakis M, Prado CMM, Lieffers JR, et al. A practical and precise approach to quantification of body composition in cancer patients using computed tomography images acquired during routine care. Appl Physiol Nutr Metab. 2008;33:997–1006.CrossRefPubMed
13.
Zurück zum Zitat Maltoni M, Caraceni A, Brunelli C, et al. Prognostic factors in advanced cancer patients: evidence-based clinical recommendations: a study by the Steering Committee of the European Association for Palliative Care. J Clin Oncol. 2005;23:6240–8.CrossRefPubMed Maltoni M, Caraceni A, Brunelli C, et al. Prognostic factors in advanced cancer patients: evidence-based clinical recommendations: a study by the Steering Committee of the European Association for Palliative Care. J Clin Oncol. 2005;23:6240–8.CrossRefPubMed
14.
Zurück zum Zitat Proctor MJ, Morrison DS, Talwar D, et al. An inflammation-based prognostic score (mGPS) predicts cancer survival independent of tumour site: a Glasgow Inflammation Outcome Study. Br J Cancer. 2011;104:726–34.CrossRefPubMedPubMedCentral Proctor MJ, Morrison DS, Talwar D, et al. An inflammation-based prognostic score (mGPS) predicts cancer survival independent of tumour site: a Glasgow Inflammation Outcome Study. Br J Cancer. 2011;104:726–34.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Lieffers JR, Bathe OF, Fassbender K, et al. Sarcopenia is associated with postoperative infection and delayed recovery from colorectal cancer resection surgery. Br J Cancer. 2012;107:931–6.CrossRefPubMedPubMedCentral Lieffers JR, Bathe OF, Fassbender K, et al. Sarcopenia is associated with postoperative infection and delayed recovery from colorectal cancer resection surgery. Br J Cancer. 2012;107:931–6.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Reisinger KW, van Vugt JLA, Tegels JJW, et al. Functional compromise reflected by sarcopenia, frailty, and nutritional depletion predicts adverse postoperative outcome after colorectal cancer surgery. Ann Surg. 2015;261:345–52.CrossRefPubMed Reisinger KW, van Vugt JLA, Tegels JJW, et al. Functional compromise reflected by sarcopenia, frailty, and nutritional depletion predicts adverse postoperative outcome after colorectal cancer surgery. Ann Surg. 2015;261:345–52.CrossRefPubMed
17.
Zurück zum Zitat Prado CM, Lieffers JR, McCargar LJ, 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;9:629–35.CrossRefPubMed Prado CM, Lieffers JR, McCargar LJ, 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;9:629–35.CrossRefPubMed
18.
Zurück zum Zitat Prado CMM, Baracos VE, McCargar LJ, et al. Body composition as an independent determinant of 5-fluorouracil-based chemotherapy toxicity. Clin Cancer Res. 2007;13:3264–8.CrossRefPubMed Prado CMM, Baracos VE, McCargar LJ, et al. Body composition as an independent determinant of 5-fluorouracil-based chemotherapy toxicity. Clin Cancer Res. 2007;13:3264–8.CrossRefPubMed
19.
Zurück zum Zitat Mir O, Coriat R, Blanchet B, et al. Sarcopenia predicts early dose-limiting toxicities and pharmacokinetics of sorafenib in patients with hepatocellular carcinoma. PLoS One. 2012;7:e37563.CrossRefPubMedPubMedCentral Mir O, Coriat R, Blanchet B, et al. Sarcopenia predicts early dose-limiting toxicities and pharmacokinetics of sorafenib in patients with hepatocellular carcinoma. PLoS One. 2012;7:e37563.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Cruz-Jentoft AJ, Baeyens JP, Bauer JM, et al. Sarcopenia: European consensus on definition and diagnosis. Report of the European Working Group on Sarcopenia in Older People. Age Ageing. 2010;39:412–23.CrossRefPubMedPubMedCentral Cruz-Jentoft AJ, Baeyens JP, Bauer JM, et al. Sarcopenia: European consensus on definition and diagnosis. Report of the European Working Group on Sarcopenia in Older People. Age Ageing. 2010;39:412–23.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Jung H-W, Kim JW, Kim J-Y, et al. Effect of muscle mass on toxicity and survival in patients with colon cancer undergoing adjuvant chemotherapy. Support Care Cancer. 2015;23:687–94.CrossRefPubMed Jung H-W, Kim JW, Kim J-Y, et al. Effect of muscle mass on toxicity and survival in patients with colon cancer undergoing adjuvant chemotherapy. Support Care Cancer. 2015;23:687–94.CrossRefPubMed
22.
Zurück zum Zitat Fujiwara N, Nakagawa H, Kudo Y, et al. Sarcopenia, intramuscular fat deposition, and visceral adiposity predict the outcomes of hepatocellular carcinoma. J Hepatol. 2015;63:131–140.CrossRefPubMed Fujiwara N, Nakagawa H, Kudo Y, et al. Sarcopenia, intramuscular fat deposition, and visceral adiposity predict the outcomes of hepatocellular carcinoma. J Hepatol. 2015;63:131–140.CrossRefPubMed
23.
Zurück zum Zitat Watanabe J, Tatsumi K, Mitsuyoshi O, et al. The impact of visceral obesity on surgical outcomes of laparoscopic surgery for colon cancer. Int J Colorectal Dis. 2014;29:343–51.CrossRefPubMed Watanabe J, Tatsumi K, Mitsuyoshi O, et al. The impact of visceral obesity on surgical outcomes of laparoscopic surgery for colon cancer. Int J Colorectal Dis. 2014;29:343–51.CrossRefPubMed
24.
Zurück zum Zitat Rickles AS, Iannuzzi JC, Mironov O, et al. Visceral obesity and colorectal cancer: are we missing the boat with BMI? J Gastrointest Surg. 2013;17:133–43.CrossRefPubMed Rickles AS, Iannuzzi JC, Mironov O, et al. Visceral obesity and colorectal cancer: are we missing the boat with BMI? J Gastrointest Surg. 2013;17:133–43.CrossRefPubMed
25.
Zurück zum Zitat Ballian N, Lubner MG, Munoz A, et al. Visceral obesity is associated with outcomes of total mesorectal excision for rectal adenocarcinoma. J Surg Oncol. 2012;105:365–70.CrossRefPubMed Ballian N, Lubner MG, Munoz A, et al. Visceral obesity is associated with outcomes of total mesorectal excision for rectal adenocarcinoma. J Surg Oncol. 2012;105:365–70.CrossRefPubMed
26.
Zurück zum Zitat Clark W, Siegel EM, Chen YA, et al. Quantitative measures of visceral adiposity and body mass index in predicting rectal cancer outcomes after neoadjuvant chemoradiation. J Am Coll Surg. 2013;216;1070–81.CrossRefPubMedPubMedCentral Clark W, Siegel EM, Chen YA, et al. Quantitative measures of visceral adiposity and body mass index in predicting rectal cancer outcomes after neoadjuvant chemoradiation. J Am Coll Surg. 2013;216;1070–81.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Watt DG, Martin JC, Park JH, et al. Neutrophil count is the most important prognostic component of the differential white cell count in patients undergoing elective surgery for colorectal cancer. Am J Surg. 2015;210:24–30.CrossRefPubMed Watt DG, Martin JC, Park JH, et al. Neutrophil count is the most important prognostic component of the differential white cell count in patients undergoing elective surgery for colorectal cancer. Am J Surg. 2015;210:24–30.CrossRefPubMed
28.
Zurück zum Zitat Argilés JM, Busquets S, López-Soriano FJ. The pivotal role of cytokines in muscle wasting during cancer. Int J Biochem Cell Biol. 2005;37:2036–46.CrossRefPubMed Argilés JM, Busquets S, López-Soriano FJ. The pivotal role of cytokines in muscle wasting during cancer. Int J Biochem Cell Biol. 2005;37:2036–46.CrossRefPubMed
29.
30.
Zurück zum Zitat Cohen S, Nathan JA, Goldberg AL. Muscle wasting in disease: molecular mechanisms and promising therapies. Nat Rev Drug Discov. 2015;14:58–74.CrossRefPubMed Cohen S, Nathan JA, Goldberg AL. Muscle wasting in disease: molecular mechanisms and promising therapies. Nat Rev Drug Discov. 2015;14:58–74.CrossRefPubMed
31.
Zurück zum Zitat Vaughan VC, Martin P, Lewandowski PA. Cancer cachexia: impact, mechanisms, and emerging treatments. J Cachexia Sarcopenia Muscle. 2013;4:95–109.CrossRefPubMed Vaughan VC, Martin P, Lewandowski PA. Cancer cachexia: impact, mechanisms, and emerging treatments. J Cachexia Sarcopenia Muscle. 2013;4:95–109.CrossRefPubMed
32.
Zurück zum Zitat Roxburgh CSD, McMillan DC. Role of systemic inflammatory response in predicting survival in patients with primary operable cancer. Future Oncol. 2010;6:149–63.CrossRefPubMed Roxburgh CSD, McMillan DC. Role of systemic inflammatory response in predicting survival in patients with primary operable cancer. Future Oncol. 2010;6:149–63.CrossRefPubMed
33.
Zurück zum Zitat Richards CH, Roxburgh CSD, MacMillan MT, et al. The relationships between body composition and the systemic inflammatory response in patients with primary operable colorectal cancer. PLoS One. 2012;7:e41883.CrossRefPubMedPubMedCentral Richards CH, Roxburgh CSD, MacMillan MT, et al. The relationships between body composition and the systemic inflammatory response in patients with primary operable colorectal cancer. PLoS One. 2012;7:e41883.CrossRefPubMedPubMedCentral
Metadaten
Titel
Prognostic Value of Computed Tomography: Measured Parameters of Body Composition in Primary Operable Gastrointestinal Cancers
verfasst von
Douglas Black, MBChB, MSc, MRCS
Craig Mackay, MBChB, MRCS, PhD
George Ramsay, MBChB, MRCS, PhD
Zaid Hamoodi, MBChB
Shayanthan Nanthakumaran, MBChB, FRCS, MD
Kenneth G. M. Park, MBChB, FRCS, MD
Malcolm A. Loudon, MBChB, FRCS, MD
Colin H. Richards, MBChB, FRCS, MD
Publikationsdatum
21.03.2017
Verlag
Springer International Publishing
Erschienen in
Annals of Surgical Oncology / Ausgabe 8/2017
Print ISSN: 1068-9265
Elektronische ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-017-5829-z

Weitere Artikel der Ausgabe 8/2017

Annals of Surgical Oncology 8/2017 Zur Ausgabe

Update Chirurgie

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

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

CME: 2 Punkte

Prof. Dr. med. Gregor Antoniadis 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“

CME: 2 Punkte

Dr. med. Benjamin Meyknecht, PD Dr. med. Oliver Pieske Das Webinar S2e-Leitlinie „Distale Radiusfraktur“ 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“

CME: 2 Punkte

Dr. med. Mihailo Andric
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.