Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Skeletal Muscle Depletion Predicts the Prognosis of Patients with Advanced Pancreatic Cancer Undergoing Palliative Chemotherapy, Independent of Body Mass Index

  • Younak Choi,

    Current address: Department of Internal Medicine, Dongguk University Gyeongju Hospital, Gyeongsangbuk-do, Korea

    Affiliation Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea

  • Do-Youn Oh ,

    ohdoyoun@snu.ac.kr

    Affiliations Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea

  • Tae-Yong Kim ,

    Contributed equally to this work with: Tae-Yong Kim, Kyung-Hun Lee, Sae-Won Han, Seock-Ah Im, Tae-You Kim

    Affiliations Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea

  • Kyung-Hun Lee ,

    Contributed equally to this work with: Tae-Yong Kim, Kyung-Hun Lee, Sae-Won Han, Seock-Ah Im, Tae-You Kim

    Affiliations Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea

  • Sae-Won Han ,

    Contributed equally to this work with: Tae-Yong Kim, Kyung-Hun Lee, Sae-Won Han, Seock-Ah Im, Tae-You Kim

    Affiliations Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea

  • Seock-Ah Im ,

    Contributed equally to this work with: Tae-Yong Kim, Kyung-Hun Lee, Sae-Won Han, Seock-Ah Im, Tae-You Kim

    Affiliations Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea

  • Tae-You Kim ,

    Contributed equally to this work with: Tae-Yong Kim, Kyung-Hun Lee, Sae-Won Han, Seock-Ah Im, Tae-You Kim

    Affiliations Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea

  • Yung-Jue Bang

    Affiliations Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea

Abstract

Introduction

Body composition has emerged as a prognostic factor in cancer patients. We investigated whether sarcopenia at diagnosis and loss of skeletal muscle during palliative chemotherapy were associated with survival in patients with pancreatic cancer.

Methods

We retrospectively reviewed the clinical outcomes of pancreatic cancer patients receiving palliative chemotherapy between 2003 and 2010. The cross-sectional area of skeletal muscle at L3 by computed tomography was analyzed with Rapidia 3D software. We defined sarcopenia as a skeletal muscle index (SMI)< 42.2 cm2/m2 (male) and < 33.9 cm2/m2 (female) using ROC curve.

Results

Among 484 patients, 103 (21.3%) patients were sarcopenic at diagnosis. Decrease in SMI during chemotherapy was observed in 156 (60.9%) male and 65 (40.6%) female patients. Decrease in body mass index (BMI) was observed in 149 patients (37.3%), with no gender difference. By multivariate analysis, sarcopenia (P< 0.001), decreasedBMI and SMI during chemotherapy (P = 0.002, P = 0.004, respectively) were poor prognostic factors for overall survival (OS). While the OS of male patients was affected with sarcopenia (P< 0.001) and decreased SMI (P = 0.001), the OS of female patients was influenced with overweight at diagnosis (P = 0.006), decreased BMI (P = 0.032) and decreased SMI (P = 0.014). Particularly, while the change of BMI during chemotherapy did not have impact on OS within the patients with maintained SMI (P = 0.750), decrease in SMI was associated with poor OS within the patients with maintained BMI (HR 1.502; P = 0.002).

Conclusions

Sarcopenia at diagnosis and depletion of skeletal muscle, independent of BMI change, during chemotherapy were poor prognostic factors in advanced pancreatic cancer.

Introduction

The pancreas, which plays a major role in digestion, produces various digestive enzymes. Therefore, most patients with pancreatic cancer have a history of unintentional weight loss at presentation. Numerous reports over several decades have shown that weight loss in cancer patients predicts poor survival [1]. We recently reported that weight loss during palliative chemotherapy predicts poor survival in advanced pancreatic cancer (APC) patients [2]. However, recent studies have reported that the change in body composition rather than body weight is important for predicting all-cause mortality of patients with cancer [38].

Cachexia can be diagnosed in patients with more than 5% of involuntary loss of body weight over the previous 6 months or in already depleted patients (body-mass index [BMI] <20 kg/m2) or sarcopenic patients with more than 2% of ongoing weight loss [9]. Sarcopenia is defined as the state of depleted muscle mass independent of fat mass. The current diagnostic criterion for sarcopenia is muscle mass over 2 standard deviations (SD) below that for typical healthy adults (men, 7.26 kg/m2; women, 5.45 kg/m2; by dual-energy x-ray absorptiometry [DEXA] in the western population) [9, 10]. Loss of skeletal muscle and gain of adipose tissue can occur simultaneously, which results in sarcopenic obesity. Sarcopenic obesity was recently shown to be an independent poor prognostic factor for survival in patients with cancer [36]. However, sarcopenia defined by the cutoffs from previous Western studies could not show the relationship with survival among the Asian population [11]. Furthermore, no study has investigated the dynamics of body weight and body muscle mass in cancer patients who receive chemotherapy and its clinical implication.

In this study, we investigated the prevalence of sarcopeniaby the cutoffs from our ownpopulation and changes in skeletal muscle mass during palliative chemotherapy, and their impact on clinical outcomes in patients with APC.

Materials and Methods

Study patients

We assessed all the patients with histologically confirmed pancreatic cancer (PC) who received palliative chemotherapy at Seoul National University Hospital between January 2003 and December 2012. Among the assessed patients, those without computed tomography (CT) scans that included the third lumbar (L3) level within 30 days before starting the first cycle of chemotherapy were excluded.

The patients’ medical records were reviewed retrospectively. Body weight and height were collected at the time of diagnosis of APC. Change in body weight was determined asthe difference in the weight between atdiagnosis and at the cessation of first-line chemotherapy because of progression. Patients were categorized according to initial BMI into 3 groups, as follows: <20.0 kg/m2 (underweight), 20.0–24.9 kg/m2 (normal weight), and ≥25.0 kg/m2 (overweight or obese).

Skeletal muscle mass measurement

Cross-sectional imaging of CT scans was used to analyze skeletal muscle mass as in previous studies [3, 4, 12]. CT scans performed at the date of diagnosis wereused to quantify the initial area of L3 skeletal muscle. The CT scans that provided diagnostic evidence of disease progression were used for the final measurement. Two adjacent axial images within the same serieswere used to determine the cross-sectional area of L3 skeletal muscle, and the mean of the 2 measurements was calculated for each patient. Muscles were quantified within a Hounsfield unit (HU) range of -29 to 150 HU [12], and analyzed with Rapidia 3D software (v2.8; INFINITT Healthcare, Seoul, Korea). A single trained staff person corrected the boundary of the entire area of L3 skeletal muscle (Fig 1). Muscle area was normalized for height in meters squared (m2) and reported as the lumbar skeletal muscle index (SMI) (cm2/m2) [4, 5, 9]. We used the area under the receiver operating characteristic (ROC) curve to determine the cutoff values ofsarcopenia for both genders.

thumbnail
Fig 1. Computed tomography images of the region of the third lumbar vertebra, with skeletal muscle highlighted in blue (-29 to 150 Hounsfield units).

The blue line on the three-dimensionally reconstructed image on the left indicates the level of the third lumbar vertebra, which is shown in the axial view on the right.

https://doi.org/10.1371/journal.pone.0139749.g001

Statistical analysis

Data on baseline characteristics and body composition are shown as mean and SD. The median overall survival (OS) and progression free survival (PFS) were determined using the Kaplan-Meier method. Between-group differences in demographic and clinical data were evaluated using the Fisher exact test for categorical variables and independent t-test for continuous variables. Relationships between continuous variables were assessed by determining Pearson correlations. Logistic regression was used to identify factors associated with decrease in SMI during chemotherapy. Comparison of survival among subgroups was done using the log-rank test. Cox proportional hazards regression model was used to determine the relationship of explanatory variables to survival as hazard ratios (HR) and 95% confidence intervals (CI). All tests were 2-sided, and P≤0.05 was considered statistically significant. Statistical analyses were performed using SPSS, ver. 19.0 (IBM Corp. Armonk, NY, USA).

Ethics.

This study was reviewed and approved by the Institutional Review Board of Seoul National University Hospital (IRB No: H-1307-146-507). All studies were conducted according to ethical guidelines (Declaration of Helsinki) for biomedical research. Requirement for informed consent was waived for this retrospective analysis of clinical data. All patients’records were anonymized and de-identified prior to analysis.

Results

Patient characteristics

Of 514 initially identified consecutive patients, this study included 484 patients with adequate CT images. Their clinical characteristics at initial assessment are shown according to gender in Table 1. The median age was 60.4 years (range, 20–85 years) and 80.2% of patients had metastatic disease. Most patients (90.7%) underwent gemcitabine-based chemotherapy, and the remainders received fluoropyrimidine-based chemotherapy including 8 patients treated with FOLFIRINOX regimen. The cutoff values of sarcopenia were determined through ROC curve were< 42.2 cm2/m2 for men and < 33.9 cm2/m2 for women, respectively (Fig 2).

Body composition

There were no significant differences in the mean BMI values and distribution of patients in BMI subgroups based on genders. The number of overweight or obese (BMI≥25 kg/m2) men and women were 10.6% and 13.3%, respectively. The mean SMI values (±SD) of men and women were 47.7±7.1 cm2/m2 and 38.6±5.4 cm2/m2, respectively (P<0.001). However, there were no significant differences in the distribution of men and women in SMI subgroups categorized with means and SDs.

Sarcopenia was found in 103 (21.3%) patients. The distribution of sarcopeniawas not significantly different between men and women(22.7% vs19.0%; P = 0.364,Table 1). There were 3 (0.6%) patients with sarcopenic obesity (overweight or obese patients with sarcopenia) and 48 (10.0%) patients with obvious cachexia (BMI<20 kg/m2 with sarcopenia; S1 Table), including 18 female patients. SMI at diagnosis was significantly correlated with BMI at diagnosis in both genders (males, r = 0.619, P<0.001; females, r = 0.506, P<0.001; S1A Fig).

The mean BMI loss during chemotherapy for each gender was 0.6 kg/m2, and one-third of each gender lost more than 1 kg/m2 (<-1 kg/m2). The mean SMI loss was significantly greater in men than in women (3.8 cm2/m2 vs 1.1 cm2/m2; P<0.001). More male patients lost greater than 2cm2/m2 (<-2 cm2/m2) than female patients (60.9% vs 40.6%; P<0.001; Table 2).

thumbnail
Table 2. Changes in body composition during first line chemotherapy by gender.

https://doi.org/10.1371/journal.pone.0139749.t002

Factors related to decrease in skeletal muscle

Logistic regression analysis revealed that male gender, sarcopenia, decreased BMI during chemotherapy, and progressed tumor response were significantly associated with decreased SMI during first-line chemotherapy (S2 Table).

Treatment outcomes

The median duration of follow-up was 11.0 months (95% CI 10.1–11.9 months). The median OS and PFS of first line chemotherapy were 8.4 months (95% CI 7.6–9.2 months) and 3.7 months (95% CI 3.1–4.2 months), respectively. The objective response rate and disease control rate were 21.9% and 60.5%, respectively.

Factors related to overall survival.

Table 3 shows the effect of multiple clinical factors on OS. By multivariate analysis, sarcopenia (HR 1.721; 95% CI 1.298–2.284; P < 0.001; Fig 3A), decreased BMI (HR 1.452; 95% CI 1.148–1.836; P = 0.002; Fig 4A), and decreased SMI (HR 1.390; 95% CI 1.109–1.742; P = 0.004; Fig 4D) were associated with poor survival. For male patients, sarcopenia (median OS of 6.1 vs 9.4 months; HR 1.736, 95% CI 1.310–2.300; P < 0.001; Fig 3B) and decreasedSMI (median OS of 8.2 vs 10.7 months; HR 1.560, 95% CI 1.199–2.030; P = 0.001; Fig 4E) was strongly related to worse prognosis. However,decreased BMI(median OS 8.9 vs 9.3 months; HR 1.218, 95% CI 0.936–1.585; P = 0.142; Fig 4B) showed a trend to worse prognosisand BMI at diagnosis did not influence on OS (P = 0.357). For female patients, both overweight at diagnosis (HR2.069, 95% CI 1.232–3.475; P = 0.006) and decreased BMI (median OS 8.2 vs 10.2 months; HR 1.464, 95% CI 1.034–2.071; P = 0.032; Fig 4C) were significantly associated with worse prognosis. However, while decreased SMI(medianOS 8.8 vs 9.9 months; HR 1.521, 95% CI 1.090–2.124; P = 0.014; Fig 4F) waspoor prognostic, sarcopenia at diagnosis had no relation with OS (P = 0.299; Fig 3C) (Table 4).

thumbnail
Fig 3. Survival according to skeletal muscle index.

Initial SMI was related to worse prognosis (A) for the entire patients (HR 1.721; P < 0.001); (B) for the male patients (HR 1.736; P < 0.001); (C) not for female patients (P = 0.299).

https://doi.org/10.1371/journal.pone.0139749.g003

thumbnail
Fig 4. Survival according to change in body mass indexand skeletal muscle index.

BMI decreased by more than 1 kg/m2 and SMI decreased by more than 2 cm2/m2 were strongly related to worseoutcome. Decrease of BMI (A) for the entire patients (HR 1.452; P = 0.002); (B) for the male patients (HR 1.218; P = 0.142); (C) for the female patients (HR 1.464; P = 0.032). Decrease of SMI (D) for the entire patients (HR 1.390;P = 0.004); (E) for the male patients (HR 1.560; P = 0.001); (F) for the female patients (HR 1.521; P = 0.014).

https://doi.org/10.1371/journal.pone.0139749.g004

thumbnail
Table 4. Body compositionanalyses for predictors of overall survivalby gender groups.

https://doi.org/10.1371/journal.pone.0139749.t004

Correlation of BMI and SMI at diagnosis.

Further analysis categorizing BMI at diagnosis into overweight or obese (≥25 kg/m2) or not (<25 kg/m2) and SMI at diagnosis into nonsarcopenia or sarcopenia revealed that median OS of sarcopenic male patients was reduced by 3.8 months against nonsarcopenic male patients eventhough they are not overweight or obse (median OS 6.0 months vs 9.8 months; HR1.746; P< 0.001). On the other hands, median OS of overweight or obese female patients was reduced by 3.0 months against not overweight female patients eventhough they are notinsarcopenic condition (median OS 6.0 months vs 9.0 months; HR 1.964; P = 0.003) (Table 5).

Correlation of changes in BMI and SMI.

Although decreased BMI was an important factor associated with decrease in SMI (HR 2.520; P<0.001; S2 Table), the distribution of change in BMI was not well correlated with the distribution of change in SMI in either men (r = 0.165) or women (r = 0.347; S1B Fig).

Analysis of patients in SMI and BMI subgroups (SMI and BMI both maintained during chemotherapy, SMI maintained and BMI decreased, SMI decreased and BMI maintained, SMI and BMI both decreased) revealed that the patients with decreased SMI had shorter survival regardless of changes in BMI(P = 0.002; Table 6).

Compared with a median OS of 10.7 months for male patients with both SMI and BMI maintained during chemotherapy, a decreased SMI was a poor prognostic indicator although BMI was maintained (median OS 7.8 months, HR 1.589; P = 0.004). For female patients,cachectic patients(BMI and SMI both decreased)showedthe worst OS (median OS 7.6 months, HR 1.849; P = 0.010). For the patients with maintained SMI, decreased BMI had no effect on survival (P = 0.750). Analyses according to each gender showed similar results.

Discussion

In this study, we found that sarcopenia at diagnosis and skeletal muscle depletion during chemotherapy predicted worse survival for advanced pancreatic cancer patients.

The influence of weight at diagnosis and change of weight (BMI) during chemotherapy is still under debate issue. In our study, the meaning of BMI was different based on gender; forfemale patients, a BMI at diagnosis ≥25 kg/m2 conferred poor prognostic significance (HR 2.069; P = 0.006), and decreased BMI during chemotherapy had significantly worse effect on the survival in female patients (median OS 8.2 vs 10.2 months; P = 0.032), not male patients (Table 4).

Sarcopenia at diagnosis was observed in one-fifth of patients (22.7% of male patients and 19.0% of female patients; Table 1). The association of sarcopeniawith survival was particularly prominent in male patients (Fig 4; Table 4). Sarcopenic obesity, usually focused on Western studies, was worse than obesity without sarcopenia and did not result in better outcomes than cachexia [46]. In our study, there was a very few sarcopenic obese patients (0.6%), mainly because the prevalence of obesity is lower in Asian populations than in Western populations. We showed that the decreased SMI during chemotherapy was associated with poor survival (P = 0.004; Table 3) and cachectic change in body composition resulted in the worst outcome (P<0.001; Table 6). However, the most important finding of our study was that sarcopenic male patients at diagnosis had significantly shorter survival, independently of initial BMI (HR 1.746; P<0.001) and overweight or obese female patients at diagnosis had significantly shorter survival, independently of initial SMI (HR 1.964; P = 0.003; Table 5). Furthermore, while the change of BMI during chemotherapy did not have the statistically significant meaning in survival within the patients of maintained SMI (P = 0.750), decrease of SMI was associated with reduced survival within the patients of maintained BMI (HR 1.502; P = 0.002; Table 6). Several reports showed that not only cross-sectional area but also radiation attenuation value of muscle has potential prognostic meaning [5].

Inflammation, inactivity, low-protein intake and age-related factors are suggested as causes of sarcopenia. However, the mechanisms by which sarcopenia increases the risk of mortality remain unclear. Recently, in addition to the fact that glutamine is released from muscle and involved in production of rapid dividing immune cells [13], many signaling molecules, called as myokines, such as IL-6, IL-8, IL-15, brain-derived neurotrophic factor, leukemia inhibitory factor, follistatin-like 1, fibroblast growth factor-21, irisin, etc. have been identified to be produced by contracting myofibers, mediating the metabolic and endocrine function [14, 15]. Skeletal muscle is the largest organ accounting 40–50% of body weight and has high metabolic capacity using 20% of resting energy expenditure [16]. However, in the condition of muscle atrophy, energy expenditure is decreased and the rate of fat deposition tends to increase [17]. Reduced muscle mass and contractile dysfunction causes aberrant energy homeostasis, impaired cell growth, insulin resistance, and immune dysfunction by means of inadequate myokines and impaired amino acid metabolismin patients with sarcopenicobesty [15, 18, 19].

Clinically, increased susceptibility to infection, musculoskeletal injuries and other morbidities are known as a possible contributor to the shortened survival of sarcopenic patients [15, 20, 21]. Another obvious disadvantage of sarcopenia is the premature termination of treatment due to increased toxicities of chemotherapy. Because the dosage of chemotherapeutic agents is conventionally determined by body surface area or body weight, patients with sarcopenia masked by obesity, who have a lean body mass comparable to underweight patients, are prone to manifest chemotherapy toxicities [2225]. Considering the vicious cycle of sarcopenia and increased morbidity and mortality, timely interventions to increase physical activity and muscle loading would be helpful for survival of cancer patients [15].

The main limitations of our study were the retrospective design and the threshold values used to identify sarcopenia. The most frequently used cutoffs of sarcopenia in the western population are 7.26 kg/m2 in men and 5.45 kg/m2 in women, which were obtained from the Gallagher et al’s study, which had beenconducted for young healthy adults aged 18–40 years by dual-energy x-ray absorptiometry (DEXA)[10, 26]. These were converted to CT measurements of 52.4 cm2/m2 for men and 38.5 cm2/m2 for women by a regression equationto correlate the area of L3 skeletal muscle as follows: appendicular skeletal muscle mass/height2 (kg/m2) = 0.11 × CT-derived SMI (cm2/m2) + 1.17; r = 0.89; P< 0.001) [3]. Sarcopeniadefined by these cutoffs were proved the potential as a powerful predictor of poor prognosis in Western studies [46]. However, we did not apply these values because recent Asian studies reported that the cutoffs of sarcopenia from the previous Western studieswere inappropriate for Asian patients [11, 27]. Furthermore, the mean SMIs of our study population were lower than those of Western cancer populations [4, 5]. At first, we have tried the cutoffs froma previous study of healthy young Korean adults of which age distribution was similar to Gallagher et al.’s study andcomposed of 1,245 men and 1,268 women [28]. In that study, thesex-specific cutoff values defining sarcopeniahave beensuggested as 6.58 kg/m2 for men and 4.59 kg/m2 for women, by DEXA, which was identified as <49.2cm2/m2 for men and <31.1cm2/m2 forwomen with CT scan by regression equations. However, these values allowed as many as 59.0% of male patients and only 6.9% of female patients to be sarcopenic state and these cutoffs could not obtain general consensus yet. To the best of our knowledge, there are no published cutoffs for sarcopeniain Asians. Therefore, we used the thresholds obtained from our own population by ROC curve, convincing that our study contains sufficiently large Asian population to try finding Asian cutoffs of sarcopenia. In our study, we obtained new cutoffs of < 42.2 cm2/m2 for men and < 33.9 cm2/m2 for women categorizing about one-fifth of both sex as sarcopenia. Although our study did not contain enough patients with sarcopenic obesity to perform statistically valid impact of sarcopenic obesity on survival, we could demonstrate the independent impact of SMI and BMI on survival. Our cutoffs for sarcopenia need to be valid through the additional analyseswith other Asian cancer population containing more sarcopenic obese patients.

Conclusions

Sarcopenia at the diagnosis and the change in body composition during chemotherapy were independent predictors of poor survival in APC patients. Skeletal muscle depletion during chemotherapy was especially associated with poor prognosis, regardless of change in BMI. Assessment of body composition should be considered when treating patients with advanced pancreatic cancer.

Supporting Information

S1 Fig. Scatter plot for L3 lumbar skeletal muscle index and body mass index.

(A) Relationship between L3 lumbar skeletal muscle index and body mass index (Pearson r = 0.455, P < 0.001 for all the patients; r = 0.619, P < 0.001 for men; r = 0.506, P < 0.001 for women). (B) Relationship between the changes in L3 lumbar skeletal muscle index and body mass index (r = 0.214, P <0.001 for all the patients; r = 0.165, P = 0.012 for men; r = 0.347, P < 0.001 for women).

https://doi.org/10.1371/journal.pone.0139749.s001

(TIF)

S1 Table. Prevalence of sarcopenia for each body mass index subgroup.

https://doi.org/10.1371/journal.pone.0139749.s002

(DOCX)

S2 Table. Risk factors for decrease in skeletal muscle index.

https://doi.org/10.1371/journal.pone.0139749.s003

(DOCX)

Author Contributions

Conceived and designed the experiments: YC DYO. Performed the experiments: YC DYO TK KHL SWH SAI TYK. Analyzed the data: YC DYO TK KHL SWH SAI TYK. Contributed reagents/materials/analysis tools: YC DYO TK KHL SWH SAI TYK YJB. Wrote the paper: YC DYO.

References

  1. 1. Andreyev HJ, Norman AR, Oates J, Cunningham D (1998) Why do patients with weight loss have a worse outcome when undergoing chemotherapy for gastrointestinal malignancies? Eur J Cancer 34:503–509. pmid:9713300
  2. 2. Choi Y, Kim TY, Lee KH, Han SW, Oh DY, Im SA, et al. (2014) The impact of body mass index dynamics on survival of patients with advanced pancreatic cancer receiving chemotherapy. J Pain Symptom Manage 48:13–25. pmid:24321510
  3. 3. Mourtzakis M, Prado CM, Lieffers JR, Reiman T, McCargar LJ, Baracos VE (2008) 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 33:997–1006. pmid:18923576
  4. 4. Prado CM, Lieffers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L, et al. (2008) Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study. Lancet Oncol 9:629–635. pmid:18539529
  5. 5. Martin L, Birdsell L, Macdonald N, Reiman T, Clandinin MT, McCargar LJ, et al. (2013) Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol 31:1539–1547 pmid:23530101
  6. 6. Tan BH, Birdsell LA, Martin L, Baracos VE, Fearon KC. (2009) Sarcopenia in an overweight or obese patient is an adverse prognostic factor in pancreatic cancer. Clin Cancer Res 15:6973–6979. pmid:19887488
  7. 7. Veasey Rodrigues H, Baracos VE, Wheler JJ, Parsons HA, Hong DS, Naing A, et al. (2013) Body composition and survival in the early clinical trials setting. Eur J Cancer 49:3068–3075. pmid:23867127
  8. 8. Lieffers JR, Bathe OF, Fassbender K, Winget M, Baracos VE. (2012) Sarcopenia is associated with postoperative infection and delayed recovery from colorectal cancer resection surgery. Br J Cancer 107:931–936. pmid:22871883
  9. 9. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, et al. (2011) Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 12:489–495. pmid:21296615
  10. 10. Baumgartner RN, Koehler KM, Gallagher D, Romero L, Heymsfield SB, Ross RR, et al. (1998) Epidemiology of sarcopenia among the elderly in New Mexico. Am J Epidemiol 147:755–763. pmid:9554417
  11. 11. Wen X, Wang M, Jiang CM, Zhang YM. (2011) Are current definitions of sarcopenia applicable for older Chinese adults? J Nutr Health Aging. 15:847–851. pmid:22159771
  12. 12. Mitsiopoulos N, Baumgartner RN, Heymsfield SB, Lyons W, Gallagher D, Ross R. (1998) Cadaver validation of skeletal muscle measurement by magnetic resonance imaging and computerized tomography. J Appl Physiol (1985) 85:115–122.
  13. 13. Biolo G, Zorat F, Antonione R, Ciocchi B. (2005) Muscle glutamine depletion in the intensive care unit. Int J Biochem Cell Biol 37:2169–2179. pmid:16084750
  14. 14. Washington TA, White JP, Davis JM, Wilson LB, Lowe LL, Sato S, et al. (2011) Skeletal muscle mass recovery from atrophy in IL-6 knockout mice. Acta Physiol (Oxf). 202:657–669.
  15. 15. Biolo G, Cederholm T, Muscaritoli M. (2014) Muscle contractile and metabolic dysfunction is a common feature of sarcopenia of aging and chronic diseases: from sarcopenic obesity to cachexia. Clin Nutr 33:737–748. pmid:24785098
  16. 16. Muller MJ, Wang Z, Heymsfield SB, Schautz B, Bosy-Westphal A. (2013) Advances in the understanding of specific metabolic rates of major organs and tissues in humans. Curr Opin Clin Nutr Metab Care 16:501–508. pmid:23924948
  17. 17. Olsen RH, Krogh-Madsen R, Thomsen C, Booth FW, Pedersen BK. (2008) Metabolic responses to reduced daily steps in healthy nonexercising men. JAMA 299:1261–1263. pmid:18349087
  18. 18. Demontis F, Piccirillo R, Goldberg AL, Perrimon N. (2013) The influence of skeletal muscle on systemic aging and lifespan. Aging Cell 12:943–949. pmid:23802635
  19. 19. Aversa Z, Bonetto A, Penna F, Costelli P, Di Rienzo G, Lacitignola A, et al. (2012) Changes in myostatin signaling in non-weight-losing cancer patients. Ann Surg Oncol 19:1350–1356. pmid:21519918
  20. 20. Cosqueric G, Sebag A, Ducolombier C, Thomas C, Piette F, Weill-Engerer S. (2006) Sarcopenia is predictive of nosocomial infection in care of the elderly. Br J Nutr 96:895–901. pmid:17092379
  21. 21. Kyle UG, Pirlich M, Lochs H, Schuetz T, Pichard C. (2005) Increased length of hospital stay in underweight and overweight patients at hospital admission: a controlled population study. Clin Nutr 24:133–142. pmid:15681111
  22. 22. Huillard O, Mir O, Peyromaure M, Tlemsani C, Giroux J, Boudou-Rouquette P, et al. (2013) Sarcopenia and body mass index predict sunitinib-induced early dose-limiting toxicities in renal cancer patients. Br J Cancer 108:1034–1041. pmid:23462722
  23. 23. Aslani A, Smith RC, Allen BJ, Pavlakis N, Levi JA (2000) The predictive value of body protein for chemotherapy-induced toxicity. Cancer 88:796–803. pmid:10679649
  24. 24. Prado CM, Baracos VE, McCargar LJ, Reiman T, Mourtzakis M, Tonkin K, et al. (2009) Sarcopenia as a determinant of chemotherapy toxicity and time to tumor progression in metastatic breast cancer patients receiving capecitabine treatment. Clin Cancer Res 15:2920–2926. pmid:19351764
  25. 25. Antoun S, Baracos VE, Birdsell L, Escudier B, Sawyer MB (2010) Low body mass index and sarcopenia associated with dose-limiting toxicity of sorafenib in patients with renal cell carcinoma. Ann Oncol 21:1594–1598. pmid:20089558
  26. 26. Gallagher D, Visser M, De Meersman RE, Sepulveda D, Baumgartner RN, Pierson RN, et al. (1997) Appendicular skeletal muscle mass: effects of age, gender, and ethnicity. J Appl Physiol (1985) 83:229–239.
  27. 27. Lau EM, Lynn HS, Woo JW, Kwok TC, Melton LJ 3rd (2005) Prevalence of and risk factors for sarcopenia in elderly Chinese men and women. J Gerontol A Biol Sci Med Sci. 60:213–216. pmid:15814865
  28. 28. Kim YS, Lee Y, Chung YS, Lee DJ, Joo NS, Hong D, et al. (2012) Prevalence of sarcopenia and sarcopenic obesity in the Korean population based on the Fourth Korean National Health and Nutritional Examination Surveys. J Gerontol A Biol Sci Med Sci 67:1107–0013. pmid:22431554