Clinical significance of skeletal muscle density and sarcopenia in patients with pancreatic cancer undergoing first-line chemotherapy: a retrospective observational study.

DOI: https://doi.org/10.21203/rs.3.rs-17124/v1

Abstract

Background To investigate the clinical impact of sarcopenia and skeletal muscle density among patients with metastatic pancreatic adenocarcinoma who underwent palliative first line gemcitabine-based chemotherapy. Methods A total of 330 patients with metastatic pancreatic adenocarcinoma who were treated with palliative first line gemcitabine-based chemotherapy between January 2010 and March 2017 were included in this study. Sarcopenia and skeletal muscle density status were identified by L3 vertebra level skeletal muscle index in cm 2 /m 2 and muscle attenuation in Hounsfield units using computed tomography. Results A skeletal muscle index to skeletal muscle density comparison revealed a positive correlation (R 2 = 0.058, P<0.001). Kaplan–Meier analysis showed that low skeletal muscle density was associated with poor overall survival. Multivariate analysis using Cox regression showed that low skeletal muscle index and low skeletal muscle density were poor prognostic factors for overall survival, respectively. Co-presence of low skeletal muscle index and low skeletal muscle density had more powerful prognostic implication for overall survival. Grade 3 or higher toxicity of chemotherapy was more frequently observed in patients with low skeletal muscle index and low skeletal muscle density. Overall survival was not associated with skeletal muscle density status among patients who were chemotherapy responders (complete or partial responses). However, among non-responders (stable or progressive disease), low skeletal muscle density groups had significantly poorer overall survival than did the high skeletal muscle density groups. Conclusions Sarcopenia and skeletal muscle density status can be considered a prognostic factor in patients with metastatic pancreatic adenocarcinoma who receive palliative first line gemcitabine-based chemotherapy. Severe chemotherapy toxicity occurred in the sarcopenia and low skeletal muscle density groups. Our data suggest that comprehensive assessment of skeletal muscle parameters may be more useful prognostic factors.

Background

Metastatic pancreatic adenocarcinoma (mPCa) is one of the most aggressive types of cancer [1]. Although systemic chemotherapy with agents such as gemcitabine plus nab-paclitaxel and FOLFIRINOX show clinical benefits, mPCa has a dismal prognosis with a median overall survival (OS) of < 1 year [2, 3]. Cancer cachexia, experienced by most patients with mPCa, is associated with poor prognosis which has warranted numerous studies on the factors that affect it. [46]. This is important for patients who receive chemotherapy because, although chemotherapy can give patients a survival benefit, it causes toxicity and can lead to physical inactivity. In this regard, sarcopenia is associated with morbidity, mortality, and to a decreased quality of life [79]. Sarcopenia is also associated with chemotherapy-induced toxicity [10].

Several studies have recently been conducted on the relationship between the cancer prognosis and a patient’s skeletal muscle density (SMD) [11, 12]. SMD is a radiological characteristic and a low SMD reflects intramuscular adipose tissue infiltration and poor ‘quality’ skeletal muscle, which is also associated with poor muscle strength [13].

The application of computed tomography (CT) in clinical practice has led to skeletal muscle parameter evaluation improvement and is considered the gold standard for studying such parameters [14]. In this study, we aimed to investigate the clinical impact of sarcopenia and SMD by CT scan among patients with mPCa who undergo palliative first line gemcitabine-based chemotherapy.

Methods

Study population and design

A total of 330 patients with mPCa treated with palliative first line gemcitabine-based chemotherapy between January 2010 and March 2017 were initially included in this study. Among them, 79 patients who either did not undergo baseline CT scans within two weeks of the initiation of chemotherapy, had Eastern Cooperative Oncology Group performance status scores of 3–4, or were experiencing other systemic medical problems such as infection were excluded, resulting in 251 patients in the study. We performed baseline CT scans before chemotherapy and after 8 ± 2 weeks to evaluate chemotherapy responses. All diagnoses were confirmed via biopsy or aspiration of the primary tumor or metastatic lesion. The chemotherapy regimens consisted of gemcitabine alone or in combination with other agents. Radiological changes were evaluated using the Response Evaluation Criteria in Solid Tumors version 1.1 [15]. Objective response was defined as complete response (CR) or partial response (PR), while disease control was defined as CR, PR, or stable disease (SD).

Body composition assessment

CT images acquired before the chemotherapy were retrieved for analysis. One axial portal phase image was selected at the level of the third lumbar vertebral body transverse process. Skeletal muscle area measurement was performed on the selected axial image by using a commercially available system (Advantage Windows workstation 4.6, GE Healthcare, Milwaukee, Wisconsin, USA). Skin, visceral organs, and the central spinal canal were excluded manually when drawing the area containing the abdominal wall and back muscles on the axial image. The areas of the abdominal wall and back muscles were calculated based on the areas of the pixels with attenuation between − 29 and 150 Hounsfield units (HU) in the demarcated areas.

The L3 skeletal muscle index (SMI) was used to detect sarcopenia and was calculated as the total area of the L3 skeletal muscle divided by the height-squared (cm2/m2). The cut-off points for SMI were defined as 43 and 53 cm2/m2 for non-overweight (body mass index [BMI] < 25 kg/m2) and overweight men (BMI≥25 kg/m2), respectively, and as 41 cm2/m2 for women [16]. SMD was quantified as the mean muscle radiation attenuation (in HU) of the muscle cross-sectional area across the L3 vertebral body level and was assessed between − 29 and + 150 HU [17]. The cut-off points for SMD were set at 41 and 33 cm2/m2 for non-overweight and overweight patients, respectively [16]. Total fat area was calculated by the sum of visceral and subcutaneous adipose tissue.

Statistical analysis

The correlation between clinicopathologic factors and both SMI and SMD were analyzed using the Pearson’s chi-square test and linear-by-linear association. The correlation between SMI and SMD was determined using the Pearson’s chi-square test and t-test. The correlation between chemotherapy response and both SMI and SMD were analyzed using the Pearson’s chi-square test. OS and progression-free survival (PFS) were calculated from the start date of first-line palliative chemotherapy until the date of death from any cause or of disease progression, respectively. For survival analyses, living patients or those with no disease progression were censored from the last follow-up date. Univariate analyses of OS and PFS were performed using the Kaplan-Meier method and log-rank test. Multivariate Cox regression forward models were used to verify the prognostic values of SMI and SMD, and were adjusted for age, sex, performance status, tumor site, histology, number of metastatic organs, CA19-9 level, and chemotherapy regimen. All analyses were performed using SPSS software (version 24; IBM Corp., Armonk, NY), and a two-sided P < 0.05 was considered statistically significant.

Ethics

The study was performed according to the Helsinki declaration and approved by the Institutional Review Board of Seoul St. Mary's Hospital.

Results

Baseline patient characteristics

A total of 251 patients were included in the analysis; their characteristics are listed in Table 1. Low SMD was associated with high total fat areas. There were no other significant associations between clinicopathologic factors and either low SMI or low SMD. The correlation between SMI and SMD was assessed (Fig. 1). A positive correlation between SMI and SMD was found (R2 = 0.058, P < 0.001). The low-SMD group exhibited a lower SMI than the high-SMD group (42.31 ± 10.18 vs. 48.77 ± 11.67 cm2/m2, P < 0.001). Among the 166 patients with high SMD before chemotherapy, 53 (31.9%) had low SMI, whereas among the 85 patients with low SMD before chemotherapy, 49 (57.6%) had low SMI. Moreover, 49 of all 251 patients (19.5%) had both low SMI and low SMD.

Table 1
Baseline characteristics
 
All patients
SMI
 
SMD
   
High
(n = 149, 59%)
Low
(n = 102, 41%)
P
 
High
(n = 166, 66%)
Low
(n = 85, 34%)
P
Age, median (range)
63.4 ± 9.4
64.5 ± 9.1
61.7 ± 9.7
0.019
 
63.8 ± 9.0
62.6 ± 10.2
0.336
< 65
137 (54.6%)
69 (46.3%)
68 (66.7%)
0.002
 
88 (53.0%)
49 (57.6%)
0.573
≥ 65
114 (45.4%)
80 (53.7%)
34 (33.3%)
   
78 (47.0%)
36 (42.4%)
 
Sex
     
< 0.001
     
0.582
Female
90 (35.9%)
70 (47.0%)
20 (19.6%)
   
62 (37.3%)
28 (32.9%)
 
Male
161 (64.1%)
79 (53.0%)
82 (80.4%)
   
104 (62.7%)
57 (67.1%)
 
ECOG
     
0.802
     
0.368
0
26 (10.4%)
17 (11.4%)
9 8.8%)
   
16 ( 9.6%)
10 (11.8%)
 
1
182 (72.5%)
107 (71.8%)
75 (73.5%)
   
125 (75.3%)
57 (67.1%)
 
2
43 (17.1%)
25 (16.8%)
18 (17.6%)
   
25 (15.1%)
18 (21.2%)
 
Location
     
0.524
     
0.980
Head
129 (51.4%)
81 (54.4%)
48 (47.1%)
   
85 (51.2%)
44 (51.8%)
 
Body
52 (20.7%)
29 (19.5%)
23 (22.5%)
   
35 (21.1%)
17 (20.0%)
 
Tail
70 (27.9%)
39 (26.2%)
31 (30.4%)
   
46 (27.7%)
24 (28.2%)
 
Histological type
     
0.319
     
0.300
Well diff.
24 (9.6%)
14 (9.4%)
10 (9.8%)
   
18 (10.8%)
6 (7.1%)
 
Moderate diff.
151 (60.2%)
95 (63.8%)
56 (54.9%)
   
104 (62.7%)
47 (55.3%)
 
Poor diff.
41 (16.3%)
24 (16.1%)
17 (16.7%)
   
24 (14.5%)
17 (20.0%)
 
Unknown
35 (13.9%)
16 (10.7%)
19 (18.6%)
   
20 (12.0%)
15 (17.6%)
 
Number of metastatic organs
     
0.991
     
0.615
Only one (1)
134 (53.4%)
79 (53.0%)
55 (53.9%)
   
91 (54.8%)
43 (50.6%)
 
More than one (≥ 2)
117 (46.6%)
70 (47.0%)
47 (46.1%)
   
75 (45.2%)
42 (49.4%)
 
CA19-9
7216.2 ± 22900.6
5097.8 ± 15422.6
10310.7 ± 30549.6
0.114
 
6390.7 ± 21320.2
8828.4 ± 25767.8
0.454
       
0.512
     
0.034
Normal
48 (19.1%)
31 (20.8%)
17 (16.7%)
   
25 (15.1%)
23 (27.1%)
 
Elevated
203 (80.9%)
118 (79.2%)
85 (83.3%)
   
141 (84.9%)
62 (72.9%)
 
First line chemotherapy
     
0.889
     
0.930
Gemcitabine single
91 (36.3%)
53 (35.6%)
38 (37.3%)
   
61 (36.7%)
30 (35.3%)
 
Gemcitabine based chemotherapy
160 (63.7%)
96 (64.4%)
64 (62.7%)
   
105 (63.3%)
55 (64.7%)
 
BMI
21.7 ± 3.1
21.8 ± 3.1
21.6 ± 3.0
0.666
 
21.8 ± 3.4
21.6 ± 2.5
0.514
Total fat area
43.3 ± 7.8
144.6 ± 96.3
157.0 ± 85.3
0.298
 
125.8 ± 76.7
196.3 ± 101.6
< 0.001
SMA
122.1 ± 26.9
137.9 ± 20.6
98.9 ± 15.8
< 0.001
 
127.2 ± 26.6
112.1 ± 24.6
< 0.001
SMI
46.6 ± 11.6
54.0 ± 8.5
35.7 ± 4.9
< 0.001
 
48.8 ± 11.7
42.3 ± 10.2
< 0.001
SMD
43.3 ± 7.8
45.1 ± 7.4
40.7 ± 7.8
< 0.001
 
47.7 ± 5.2
34.8 ± 4.4
< 0.001
BMI, body mass index; CA19-9, carbohydrate antigen 19 − 9; diff, differentiation; ECOG, Eastern Cooperative Oncology Group; SMA, skeletal muscle area; SMD, skeletal muscle density; SMI, skeletal muscle index

Survival according to SMI and SMD

OS and PFS were assessed according to SMI and SMD (Fig. 2). With the univariate analysis, low SMI was not associated with OS (median, 6.0 versus 8.0 months; P = 0.076) or PFS (P = 0.752). Patients with low SMD had poorer OS than those with high SMD (6.1 versus 7.9 months, P = 0.010). However, there were no differences in PFS (P = 0.116) with respect to SMD. Furthermore, patients with both low SMI and low SMD showed poorer OS than others (4.1 vs 7.8 months, P = 0.004) (Fig. 3). We also performed a multivariate Cox proportional hazard regression for SMI, SMD, and baseline characteristics (Table 2). Low SMI, low SMD, and co-presence of low SMI and low SMD were statistically significant prognostic factors for OS but not for PFS (Low SMI, hazard ratio [HR]: 1.35, 95% confidence interval [CI]: 1.03–1.78, P = 0.032; low SMD, HR: 1.45, 95% CI: 1.09–1.93, P = 0.011; and Co-presence of low SMI and low SMD, HR: 1.58, 95% CI: 1.12–2.23, P = 0.010). Moreover, Eastern Cooperative Oncology Group performance status and type of first line chemotherapy regimen were prognostic factors for OS, while the number of metastatic sites and baseline CA19-9 levels were associated with both OS and PFS (Supplementary Material 1).

Table 2
Multivariate analysis for SMI and SMD with Cox regression.
   
OS
   
HR
95% CI
P
SMIa
High
1
     
 
Low
1.352
1.03
1.78
0.032
SMDb
High
1
     
 
Low
1.451
1.09
1.93
0.011
Low SMI and low SMDc
No
1
     
 
Yes
1.579
1.12
2.23
0.010
CI, confidence interval; HR, hazard ratio; OS, overall survival; SMD, skeletal muscle density.
a SMI is adjusted for age, sex, ECOG, location, histological type, number of metastatic organs, CA19-9, first line chemotherapy.
b SMD is adjusted for age, sex, ECOG, location, histological type, number of metastatic organs, CA19-9, first line chemotherapy.
c Co-presence of low SMI and low SMD is adjusted for age, sex, ECOG, location, histological type, number of metastatic organs, CA19-9, first line chemotherapy.

Chemotherapy response and toxicities according to SMI and SMD

Chemotherapy response was assessed according to SMI, SMD, and their rate of change (Table 3). Objective responses were not associated with low SMI or low SMD (mean SMI: CR/PR vs. SD/PD, 46.58 vs. 46.62 cm2/m2, P = 0.981; mean SMD: CR/PR vs. SD/PD, 43.27 vs. 43.48, respectively, HU, P = 0.856). Disease control was also not associated with low SMI or low SMD (mean SMI: CR/PR/SD vs. PD, 47.21 vs. 45.44 cm2/m2, P = 0.249; mean SMD: CR/PR/SD vs. PD,43.58 vs. 42.85, respectively, HU, P = 0.484). We also assessed the correlation between chemotherapy response and the rate of change of SMI and SMD and found that objective response was not associated with change in SMI or SMD (SMI change: CR/PR vs. SD/PD, -1.99 vs. -4.15%, respectively, P = 0.157; SMD change: CR/PR vs. SD/PD, -3.95 vs. -3.05%, respectively, P = 0.501). Disease control was also not associated with change (SMI change: CR/PR/SD vs. PD, -1.10 vs. -2.85%, respectively, P = 0.298; SMD change (%), CR/PR/SD vs, PD, -1.67 vs. -1.26, respectively, P = 0.784).

Table 3
Correlation between chemotherapy response and SMI, SMD, and their rate of change
   
CR/PR
(n = 60)
SD/PD
(n = 191)
P
 
CR/PR/SD
(n = 163)
PD
(n = 88)
P
SMI
 
46.58 ± 11.46
46.62 ± 12.07
0.981
 
47.21 ± 11.94
45.44 ± 10.88
0.249
SMI change (%)
 
-1.99 ± 11.46
-4.15 ± 9.74
0.157
 
-3.95 ± 10.59
-3.05 ± 9.77
0.501
SMD
 
43.27 ± 7.95
43.48 ± 7.52
0.856
 
43.58 ± 8.22
42.85 ± 7.09
0.484
SMD change (%)
 
-2.88 ± 11.45
-1.10 ± 11.48
0.298
 
-1.67 ± 11.11
-1.26 ± 12.17
0.784
CR, complete response; PD, progressive disease; PR, partial response; SD, stable disease; SMD, skeletal muscle density; SMI, skeletal muscle index. Values are in Hounsfield units.

We also investigated the relationship between chemotherapy-related toxicities and SMI or SMD (Table 4). Low SMI and low SMD were not associated with grade 3 or higher neutropenia, anaemia, thrombocytopenia, fatigue, or diarrhea, separately. However, all grade 3 or higher adverse events were more frequently reported by patients with low SMI (43% by high-SMI patients vs. 59% by low-SMI patients, P = 0.019) as well as by patients with low SMD (44% by high-SMD patients vs. 60% by low-SMD patients, P = 0.023).

Table 4
Correlation between toxicity and skeletal muscle parameters
 
SMI
 
SMD
 
High
(n = 149, 59%)
Low
(n = 102, 41%)
P
 
High
(n = 166, 66%)
Low
(n = 85, 34%)
P
all grade ≥ 3 toxicity
64 (43%)
60 (59%)
0.019
 
73 (44%)
51 (60%)
0.023
neutropenia
32 (22%)
28 (28%)
0.348
 
35 (21%)
25 (29%)
0.191
anemia
21 (14%)
21 (21%)
0.237
 
24 (15%)
18 (21%)
0.242
thrombocytopenia
14 (9%)
15 (15%)
0.275
 
18 (11%)
11 (13%)
0.777
fatigue
18 (12%)
21 (21%)
0.100
 
24 (15%)
15 (18%)
0.634
diarrhea
2 (1%)
1 (1%)
0.999
 
2 (1%)
1 (1%)
0.999
SMD, skeletal muscle density; SMI, skeletal muscle index.

Survival rates among chemotherapy responders and non-responders

We assessed survival according to the presence of low SMI and low SMD in both chemotherapy responders and non-responders (Fig. 4). OS was not associated with SMD status among responders (CR/PR); however, among non-responder patients (SD/PD), the low SMD group showed poorer OS than the high SMD group (median: 5.6 vs 7.4 months, P = 0.006). Thus, we analyzed survival after progression at 8 weeks of initiation of chemotherapy according to SMD status among non-responders. In this case, the low SMD group showed poorer survival after progression at 8 weeks than the high SMD group (median: 2.2 vs. 3.4 months, P = 0.004). Moreover, OS was not associated with SMI in either responder (P = 0.489) or non-responder patients (P = 0.061).

Discussion

In this study, we assessed the clinical impact of sarcopenia and SMD status in patients with mPCa who received palliative first line gemcitabine-based chemotherapy. To our knowledge, our study on the prognostic value of both sarcopenia and SMD status is the first study to evaluate comprehensively the association between skeletal muscle parameters, SMI and SMD with the largest cohort of patients with metastatic pancreatic cancer.

Our results showed that SMD is positively correlated with SMI. Several previous studies showed that SMI and SMD are positively correlated, in accordance with our research [18, 19]. However, other studies showed no significant correlation between SMI and SMD [20, 21]. It is likely that the cutoff values differed from one study to another and there might be differences among the study populations in terms of cancer type, stage of disease, sex, and age. Our study in particular consisted of those at advanced disease stages (stage 4) and, therefore, with very poor prognoses. In general, decreases in SMD are detected earlier than decreases in SMI. Furthermore, CT-based calculations allow for early detection of decreases in HUs (SMD), even when the muscle area remains unchanged [22]. Because our patients were at an advanced stage with poor prognoses, it is likely that they experienced decreases in both SMD and skeletal muscle area (SMA), which could result in the significant correlation observed between SMI and SMD. This suggests that, when assessing a patient using skeletal muscle parameters as prognostic factors, it may be useful to simultaneously evaluate SMI and SMD, rather than just one of these parameters.

Our results showed that SMD was a better prognostic factor than SMI in terms of statistical significance. Some previous studies also showed that low SMD is a better prognostic factor than SMI [12, 2225]. Similar to our findings, three previous studies [12, 22, 25] found that a low SMD was significantly associated with poor OS while sarcopenia was not, suggesting that SMD is a more reliable prognostic factor than sarcopenia status. This may also explain findings from a previous study that showed low SMD leads to muscle weakness independently of muscle area, resulting in higher prognostic value [17]. Additionally, intermuscular fat development reflects the level of physical activity [26, 27] and has also been associated with severe inflammation [28], suggesting that these patients are more likely to encounter a higher number of severe adverse events during chemotherapy. It is also possible that SMD could be a more accurate measurement of muscle function and, therefore, precedes the development of sarcopenia development [23]. Furthermore, if reduced muscle density was a manifestation of muscle wasting, SMD can become a useful tool for assessing the patient’s performance status. In fact, performance status is an important prognostic factor among patients receiving chemotherapy [29].

Several previous studies have shown how sarcopenia is a good prognostic factor for patients with cancer, which overlaps with our finding [4, 30]. Further verification is required using a better-defined cutoff on a larger patient number. Also, our results showed that analysis using both SMI and SMD is of better prognostic value than SMI or SMD alone in terms of statistical significance, which has clinical applicability. Our data suggest that comprehensive assessment of skeletal muscle parameters may be more useful prognostic factors.

Based on these results, we also determined whether the prognostic role of the skeletal muscle parameters is associated with the chemotherapy effects. Neither sarcopenia status nor SMD had any association with chemotherapy response; furthermore, changes in these parameters were not related to chemotherapy responses. These results are inconsistent with the results from previous studies. For instance, Chu, M et al. showed that high SMD was strongly associated with radiographic complete responses [18]; however, Daly et al. showed no correlation between skeletal muscle parameters and chemotherapy response [31]. In fact, comparisons with previous studies may not be feasible because variables such as the type of cancer, purpose of chemotherapy, and chemotherapy regimens differed among studies. If no association between chemotherapy response and skeletal muscle parameters is determined, it may be due to the relatively low baseline level of the skeletal muscle parameter, as well as and the rate of change for which no statistical significance could be found. Further studies are needed to clarify this issue.

Our data revealed that severe chemotherapy toxicity was associated with low SMI and low SMD, which was consistent with previous studies [7, 21]. This could be due to the link between body composition and drug pharmacokinetics and has important clinical implications. Patients with sarcopenia or low SMD should be considered for prevention and aggressive management of chemotherapy toxicity.

Although no survival rate differences were observed according to SMD in patients who responded to chemotherapy, non-responder patients with low SMD showed poor survival time after disease progression at 8 weeks. In other words, the worse the chemotherapy response, the better SMD works as a prognostic factor. This should be taken into account when deciding whether to perform second line chemotherapy or best supportive care only after disease progression. As clinicians consider several factors, such as performance status, when deciding whether to provide additional chemotherapy, weakness of skeletal muscle may also be helpful in the decision-making process.

There are limitations to our study owing to its retrospective nature. The first is that the skeletal muscle parameters were evaluated using CT, which represents a single aspect of the muscle functional status. It would be optimal to assess muscle depletion from the perspectives of both function and strength; which should be considered in future studies. The second limitation is that other parameters that reflect nutrition and health status such as food intake, and albumin and C-reactive protein levels were not investigated.

In conclusion, our results showed that SMD and sarcopenia could be considered as prognostic factors in patients with mPCa who received palliative first line gemcitabine-based chemotherapy. Severe toxicity of chemotherapy occurred in the sarcopenia and low SMD groups. Our data suggest that comprehensive assessment of skeletal muscle parameters may be useful prognostic factors.

Abbreviations

BMI: body mass index; CI: confidence interval; CR: complete response; CT: computed tomography; HR: hazard ratio; HU: Hounsfield units; mPCa: metastatic pancreatic adenocarcinoma; OS: overall survival; PFS: progression-free survival; PR: partial response; SD: stable disease; SMA: skeletal muscle area; SMD: skeletal muscle density

Declarations

Ethics approval and consent to participate: This study was approved by the Institutional Review Board of Seoul St. Mary’s Hospital.

Consent for publication: Written informed consent was obtained from all patients [32].

Availability of data and materials:

The data that support the findings of this study are available from the corresponding author but restrictions apply to the availability of these data, which were used under license for the current study, and so are not publicly available. Data are however available from the corresponding author upon reasonable request and with permission of Institutional Review Board of the Seoul St. Mary’s Hospital.

Conflict of interest: In-Ho Kim, Moon Hyung Choi, In Seok Lee, Tae Ho Hong, and Myung Ah Lee declare that they have no conflict of interest.

Funding: This research did not receive any specific grant.

Authors’ contributions:

IHK, MHC and MAL were involved in the conception and design of the study, IHK, MHC, MAL, ISL, THH acquired the data, and were involved in the analysis and interpretation of the data. IHK, MHC and MAL drafted the manuscript. MAL is the guarantor of the information.

Acknowledgments: Not applicable

References

  1. American Cancer Society. Cancer Facts & Figures 2014. Atlanta: American Cancer Society. 2014.
  2. Goldstein D, El-Maraghi RH, Hammel P, Heinemann V, Kunzmann V, Sastre J, Scheithauer W, Siena S, Tabernero J, Teixeira L et al: nab-Paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. Journal of the National Cancer Institute 2015, 107(2).
  3. Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardiere C et al: FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. The New England journal of medicine 2011, 364(19):1817-1825.
  4. Choi Y, Oh DY, Kim TY, Lee KH, Han SW, Im SA, Kim TY, Bang YJ: Skeletal Muscle Depletion Predicts the Prognosis of Patients with Advanced Pancreatic Cancer Undergoing Palliative Chemotherapy, Independent of Body Mass Index. PloS one 2015, 10(10):e0139749.
  5. Tan BH, Birdsell LA, Martin L, Baracos VE, Fearon KC: Sarcopenia in an overweight or obese patient is an adverse prognostic factor in pancreatic cancer. Clinical cancer research : an official journal of the American Association for Cancer Research 2009, 15(22):6973-6979.
  6. Aoyagi T, Terracina KP, Raza A, Matsubara H, Takabe K: Cancer cachexia, mechanism and treatment. World journal of gastrointestinal oncology 2015, 7(4):17-29.
  7. Prado CM, Baracos VE, McCargar LJ, Reiman T, Mourtzakis M, Tonkin K, Mackey JR, Koski S, Pituskin E, Sawyer MB: Sarcopenia as a determinant of chemotherapy toxicity and time to tumor progression in metastatic breast cancer patients receiving capecitabine treatment. Clinical cancer research : an official journal of the American Association for Cancer Research 2009, 15(8):2920-2926.
  8. Lieffers JR, Bathe OF, Fassbender K, Winget M, Baracos VE: Sarcopenia is associated with postoperative infection and delayed recovery from colorectal cancer resection surgery. British journal of cancer 2012, 107(6):931-936.
  9. Ozola Zalite I, Zykus R, Francisco Gonzalez M, Saygili F, Pukitis A, Gaujoux S, Charnley RM, Lyadov V: Influence of cachexia and sarcopenia on survival in pancreatic ductal adenocarcinoma: a systematic review. Pancreatology : official journal of the International Association of Pancreatology (IAP) [et al] 2015, 15(1):19-24.
  10. Anandavadivelan P, Brismar TB, Nilsson M, Johar AM, Martin L: Sarcopenic obesity: A probable risk factor for dose limiting toxicity during neo-adjuvant chemotherapy in oesophageal cancer patients. Clinical nutrition (Edinburgh, Scotland) 2016, 35(3):724-730.
  11. Shachar SS, Deal AM, Weinberg M, Nyrop KA, Williams GR, Nishijima TF, Benbow JM, Muss HB: Skeletal Muscle Measures as Predictors of Toxicity, Hospitalization, and Survival in Patients with Metastatic Breast Cancer Receiving Taxane-Based Chemotherapy. Clinical cancer research : an official journal of the American Association for Cancer Research 2017, 23(3):658-665.
  12. Antoun S, Lanoy E, Iacovelli R, Albiges-Sauvin L, Loriot Y, Merad-Taoufik M, Fizazi K, di Palma M, Baracos VE, Escudier B: Skeletal muscle density predicts prognosis in patients with metastatic renal cell carcinoma treated with targeted therapies. Cancer 2013, 119(18):3377-3384.
  13. Miljkovic I, Zmuda JM: Epidemiology of myosteatosis. Current opinion in clinical nutrition and metabolic care 2010, 13(3):260-264.
  14. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, Jatoi A, Loprinzi C, MacDonald N, Mantovani G et al: Definition and classification of cancer cachexia: an international consensus. The Lancet Oncology 2011, 12(5):489-495.
  15. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M et al: New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). European journal of cancer (Oxford, England : 1990) 2009, 45(2):228-247.
  16. Martin L, Birdsell L, Macdonald N, Reiman T, Clandinin MT, McCargar LJ, Murphy R, Ghosh S, Sawyer MB, Baracos VE: Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2013, 31(12):1539-1547.
  17. Goodpaster BH, Kelley DE, Thaete FL, He J, Ross R: Skeletal muscle attenuation determined by computed tomography is associated with skeletal muscle lipid content. Journal of applied physiology (Bethesda, Md : 1985) 2000, 89(1):104-110.
  18. Chu MP, Lieffers J, Ghosh S, Belch A, Chua NS, Fontaine A, Sangha R, Turner RA, Baracos VE, Sawyer MB: Skeletal muscle density is an independent predictor of diffuse large B-cell lymphoma outcomes treated with rituximab-based chemoimmunotherapy. Journal of cachexia, sarcopenia and muscle 2017, 8(2):298-304.
  19. Caan BJ, Cespedes Feliciano EM, Prado CM, Alexeeff S, Kroenke CH, Bradshaw P, Quesenberry CP, Weltzien EK, Castillo AL, Olobatuyi TA et al: Association of Muscle and Adiposity Measured by Computed Tomography With Survival in Patients With Nonmetastatic Breast Cancer. JAMA oncology 2018, 4(6):798-804.
  20. Weinberg MS, Shachar SS, Muss HB, Deal AM, Popuri K, Yu H, Nyrop KA, Alston SM, Williams GR: Beyond sarcopenia: Characterization and integration of skeletal muscle quantity and radiodensity in a curable breast cancer population. The breast journal 2018, 24(3):278-284.
  21. Miyata H, Sugimura K, Motoori M, Fujiwara Y, Omori T, Yanagimoto Y, Ohue M, Yasui M, Miyoshi N, Tomokuni A et al: Clinical Assessment of Sarcopenia and Changes in Body Composition During Neoadjuvant Chemotherapy for Esophageal Cancer. Anticancer research 2017, 37(6):3053-3059.
  22. Hayashi N, Ando Y, Gyawali B, Shimokata T, Maeda O, Fukaya M, Goto H, Nagino M, Kodera Y: Low skeletal muscle density is associated with poor survival in patients who receive chemotherapy for metastatic gastric cancer. Oncology reports 2016, 35(3):1727-1731.
  23. Chu MP, Lieffers J, Ghosh S, Belch AR, Chua NS, Fontaine A, Sangha R, Turner AR, Baracos VE, Sawyer MB: Skeletal muscle radio-density is an independent predictor of response and outcomes in follicular lymphoma treated with chemoimmunotherapy. PloS one 2015, 10(6):e0127589.
  24. Sabel MS, Lee J, Cai S, Englesbe MJ, Holcombe S, Wang S: Sarcopenia as a prognostic factor among patients with stage III melanoma. Annals of surgical oncology 2011, 18(13):3579-3585.
  25. Rier HN, Jager A, Sleijfer S, van Rosmalen J, Kock M, Levin MD: Low muscle attenuation is a prognostic factor for survival in metastatic breast cancer patients treated with first line palliative chemotherapy. Breast (Edinburgh, Scotland) 2017, 31:9-15.
  26. Goodpaster BH, Chomentowski P, Ward BK, Rossi A, Glynn NW, Delmonico MJ, Kritchevsky SB, Pahor M, Newman AB: Effects of physical activity on strength and skeletal muscle fat infiltration in older adults: a randomized controlled trial. Journal of applied physiology (Bethesda, Md : 1985) 2008, 105(5):1498-1503.
  27. Tuttle LJ, Sinacore DR, Cade WT, Mueller MJ: Lower physical activity is associated with higher intermuscular adipose tissue in people with type 2 diabetes and peripheral neuropathy. Physical therapy 2011, 91(6):923-930.
  28. Miljkovic I, Kuipers AL, Kammerer CM, Wang X, Bunker CH, Patrick AL, Wheeler VW, Kuller LH, Evans RW, Zmuda JM: Markers of inflammation are heritable and associated with subcutaneous and ectopic skeletal muscle adiposity in African ancestry families. Metabolic syndrome and related disorders 2011, 9(4):319-326.
  29. Hamada T, Nakai Y, Yasunaga H, Isayama H, Matsui H, Takahara N, Sasaki T, Takagi K, Watanabe T, Yagioka H et al: Prognostic nomogram for nonresectable pancreatic cancer treated with gemcitabine-based chemotherapy. British journal of cancer 2014, 110(8):1943-1949.
  30. Park I, Choi SJ, Kim YS, Ahn HK, Hong J, Sym SJ, Park J, Cho EK, Lee JH, Shin YJ et al: Prognostic Factors for Risk Stratification of Patients with Recurrent or Metastatic Pancreatic Adenocarcinoma Who Were Treated with Gemcitabine-Based Chemotherapy. Cancer research and treatment : official journal of Korean Cancer Association 2016, 48(4):1264-1273.
  31. Daly LE, Ni Bhuachalla EB, Power DG, Cushen SJ, James K, Ryan AM: Loss of skeletal muscle during systemic chemotherapy is prognostic of poor survival in patients with foregut cancer. Journal of cachexia, sarcopenia and muscle 2018, 9(2):315-325.
  32. von Haehling S, Morley JE, Coats AJS, Anker SD: Ethical guidelines for publishing in the journal of cachexia, sarcopenia and muscle: update 2017. Journal of cachexia, sarcopenia and muscle 2017, 8(6):1081-1083.