Low skeletal muscle density is associated with poor survival in patients who receive chemotherapy for metastatic gastric cancer

  • Authors:
    • Naomi Hayashi
    • Yuichi Ando
    • Bishal Gyawali
    • Tomoya Shimokata
    • Osamu Maeda
    • Masahide Fukaya
    • Hidemi Goto
    • Masato Nagino
    • Yasuhiro Kodera
  • View Affiliations

  • Published online on: December 8, 2015     https://doi.org/10.3892/or.2015.4475
  • Pages: 1727-1731
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Low skeletal muscle density (SMD) and low skeletal muscle index (SMI) are associated with poor overall survival (OS) in patients with various types of cancer. We retrospectively studied SMD and SMI using computed tomographic (CT) scans in patients with gastric cancer receiving chemotherapy to evaluate its prognostic significance. SMD and SMI were obtained from CT-based analysis using Slice-O-Matic® medical imaging software in patients who received S-1 plus cisplatin chemotherapy for metastatic gastric cancer. The CT images taken within 1 month before starting chemotherapy were used. The cut-off values for determining low SMD [<33 Hounsfield units (HU) in obese and <41 HU in non-obese patients] and low SMI (<41 cm2/m2 in females, <43 cm2/m2 in non-obese males and <53 cm2/m2 in obese males) were referenced from a large population based study. The CT images of 53 patients were reviewed. The median SMD was 36.8 HU (range, 19.5-59.3 HU), and the median SMI was 39.8 cm2/m2 (range, 23.7-60.0 cm2/m2). Patients with low SMD had significantly shorter OS compared with patients having normal SMD (8.9 vs. 12.8 months, P=0.03). However, OS did not differ significantly between patients with low and normal SMI (11.1 and 14.3 months, P=0.18). Multivariate analyses confirmed that low SMD was an independent predictor of poor outcomes (P<0.01). SMD is an important prognosticator of survival in patients with metastatic gastric cancer receiving chemotherapy.

Introduction

The role of skeletal muscle as a prognostic and predictive factor in cancer management has been a topic of interest lately (16). In particular, two skeletal muscle parameters have been studied in detail: skeletal muscle density (SMD) and skeletal muscle index (SMI). SMD reflects the lipid content of the muscle: the higher the lipid content, the lower the SMD and the weaker the muscle strength (79). SMI reflects the skeletal muscle volume or mass and is measured as skeletal muscle area divided by the square of the body height.

Both SMD and SMI have been shown to predict survival in patients with various types of cancer (10,11). Low SMI, also known as sarcopenia, is also a predictor of severe toxicities associated with anticancer drugs such as 5-fluorouracil and sorafenib (12,13). However, this association of SMD and/or SMI with prognosis or treatment-toxicities among gastric cancer patients has not been studied yet.

The clinical significance of SMD and SMI in patients with gastric cancer may differ from that in patients with other types of cancer owing to differences in dietary status caused by gastrectomy, gastrointestinal disorders, or both. Because chemotherapy is the only treatment for metastatic gastric cancer and has only limited survival benefit (14), it is important to have factors that could effectively predict the prognosis or adverse effects for proper assessment of risk-benefit ratio to the patients. Hence, we undertook this study to evaluate the prognostic implications of SMD and SMI in patients with metastatic gastric cancer receiving chemotherapy.

Materials and methods

Patients

Patients with recurrent or metastatic gastric cancer who received S-1 plus cisplatin as first-line chemotherapy in Nagoya University Hospital (Nagoya, Japan) from January 2009 through June 2014 were studied retrospectively. All but 2 patients received S-1 (80 mg/m2 on days 1–21) orally and cisplatin (60 mg/m2 on day 8) intravenously every 5 weeks. The other 2 patients received S-1 (80 mg/m2 on days 1–14) orally and cisplatin (60 mg/m2 on day 1) intravenously every 3 weeks. All patients had histologically proven adenocarcinoma of stomach with at least one metastatic lesion as confirmed by diagnostic imaging. CT scanning was performed every 2 months in most patients to evaluate treatment response. Patients with double primary cancers were excluded. This retrospective study was conducted after receiving formal approval from our Institutional Review Board.

Data collection

Clinical data were extracted from the hospital electronic medical database and included the following variables: age, gender, ECOG performance status (PS), height (m), body weight (kg), at baseline (before starting chemotherapy), history of gastrectomy, histopathological characteristics of primary tumor (well or poorly differentiated), and subsequent treatments, including conversion surgery. Relative dose intensity (RDI) was calculated by dividing the actual total dose intensity (actual total dose divided by the duration of therapy) by the planned total dose intensity (planned total dose divided by the planned duration of therapy).

Skeletal muscle assessment

Baseline CT images obtained within 1 month before starting chemotherapy were used to evaluate SMD and SMI. Skeletal muscle area at the level of the third lumber vertebra (l3) scan was quantified by using Slice-O-Matic® medical imaging software (version 5.0; TomoVision, Magog, Quebec, Canada) using a Hounsfield unit (HU) threshold of −29 to +150 for identification as has been described previously (15) (Fig. 1). The skeletal muscle area thus obtained was divided by the square of body height to get the skeletal muscle index (SMI) in the units of cm2/m2. The value for the skeletal muscle density (SMD) in HU at the same level was also obtained from the software. It is estimated that SMD decreases by 1 HU for each additional 1 g/100 ml lipid in muscle (8). The cut-off values for determining low SMD and low SMI were based on a recently published population-based study of 1,473 patients (10) (Table I). Although various cut-offs for diagnosing sarcopenia have been proposed, this criteria takes into account both gender and BMI, and therefore is more reliable.

Table I

Cut-off values of SMD and SMI.

Table I

Cut-off values of SMD and SMI.

BMI (kg/m2)SMD (HU)
SMI (cm2/m2)
MaleFemaleMaleFemale
<20.0<41<41<43<41
20.0 to 24.9<41<41<43<41
≥25.0<33<33<53<41

[i] SMD, skeletal muscle density; BMI, body mass index; SMI, skeletal mass index; HU, Hounsfield units. Reproduced from Martin et al with permission (10).

Statistical analysis

Overall survival (OS), progression-free survival (PFS), and tumor response (evaluated according to the Response Evaluation Criteria in Solid Tumors, version 1.0) were compared between patients with low SMD and those with normal SMD, as well as between patients with low SMI and those with normal SMI. Briefly, OS was defined as the time from the day of starting chemotherapy to the day of death or the last contact, and PFS was defined as the time from the day of starting chemotherapy to the day on which the first event of disease progression was diagnosed or the day of death from any cause. PFS and OS with 95% confidence intervals (95% CI) were estimated by the Kaplan-Meier method and were compared between groups by the log-rank test. Data on patients who were alive or lost to follow-up were censored in the calculation of OS. Data on patients who discontinued chemotherapy because of adverse events or who could not be followed up until disease progression were censored in the calculation of PFS. Associations of the following variables with OS were analyzed with the use of multivariate Cox hazard models: SMD (normal vs. low), SMI (normal vs. low), gender (male vs. female), PS (0–1 vs. 2), age (less than or equal to the median vs. higher than the median), number of metastatic sites (less than 2 vs. 2 or more), and tumor response to first-line chemotherapy (yes vs. no). P-values <0.05 were considered to indicate statistical significance. P-values are estimated from one-sided tests. Statistical analysis was performed using JMP software (version 9; SAS Institute Inc., Cary, NC, USA).

Results

Patient characteristics

A total of 53 patients were included in this study (Table II). The median SMD was 36.8 HU (range, 19.5–59.3 HU), and the median SMI was 39.8 cm2/m2 (range, 23.7–60.0 cm2/m2). Thirty-one patients (58.5%) had low SMD (median, 30.8 HU) and 37 patients (69.8%) had low SMI (median, 36.1 cm2/m2). The distribution of the 34 patients (64.1%) who had undergone gastrectomy before starting chemotherapy was balanced between low and normal groups of both SMD or SMI. There was no significant difference in RDI between the patients with low SMD and those with normal SMD (cisplatin, P=0.77; S-1, P=0.83) or between patients with low SMI and those with normal SMI (cisplatin, P=0.89; S-1, P=0.86). There was a numerically higher incidence of grade 3 or higher neutropenia in the patients with low SMI than in those with normal SMI (35.1 vs. 18.8%, P=0.22).

Table II

Patient characteristics.

Table II

Patient characteristics.

VariableSMD (HU)
P-valueSMI (cm2/m2)
P-value
Low (n=31) n (%)Normal (n=22) n (%)Low (n=37) n (%)Normal (n=16) n (%)
Age (years)
 Median6860.5<0.016663.50.71
 Range44–8033–8033–8044–80
Gender
 Male17 (54.8)16 (72.7)0.9019 (51.1)14 (87.5)0.01a
 Female14 (45.2)6 (27.2)18 (48.6)2 (12.5)
PS
 0 to 127 (87.1)19 (86.3)0.9432 (86.4)14 (87.5)0.23
 24 (12.9)3 (13.6)5 (13.5)2 (12.5)
Gastrectomy
 Yes22 (70.9)12 (54.5)0.2126 (70.2)8 (50.0)0.23
 No9 (29.1)10 (45.4)11 (29.7)8 (50.0)
Differentiation
 Well10 (32.2)4 (18.2)0.2511 (29.7)3 (18.8)0.40
 Moderate to poor21 (67.7)18 (81.8)26 (70.2)13 (81.3)
No. of metastatic site
 <225 (80.6)17 (77.3)0.7728 (75.7)14 (87.5)0.33
 ≥26 (19.3)5 (22.7)9 (24.3)2 (12.5)
Conversion surgery
 Yes6 (19.3)2 (9.1)0.303 (8.1)5 (31.5)0.03a
 No25 (80.6)20 (90.9)34 (91.9)11 (68.8)
SMD
 Median30.844.8<0.01a36.837.50.59
SMI
 Median39.841.70.4536.148.2<0.01a

a Statistically significant difference (P<0.05). PS, performance status; SMD, skeletal muscle density; BMI, body mass index; SMI, skeletal mass index; HU, Hounsfield units.

Overall survival

The median OS of the 53 patients was 11.7 months (95% CI, 8.2 to 13.2 months). Patients with low SMD had significantly shorter OS (8.9 months; 95% CI, 7.7 to 12.8 months) compared with patients with normal SMD (12.8 months; 95% CI, 10.1 to 37.0 months, P= 0.03) (Fig. 2A). On the other hand, median OS was similar between patients with low SMI and normal SMI (11.1 vs. 14.3 months, P=0.18) (Fig. 2B).

Progression-free survival

The median PFS of the 53 patients was 4.8 months (95% CI, 3.2 to 6.7 months). Median PFS did not differ significantly between patients with low SMD and those with normal SMD (4.4 vs. 4.9 months, P=0.16) or between patients with low SMI and those with normal SMI (4.4 vs. 5.1 months, P=0.31) (Fig. 3).

Tumor response

Among the 53 patients, 2 patients had a complete response, 12 had a partial response, and 18 had stable disease. The response rate was 26.4%, and the disease control rate was 60.4%. The response rate was similar in patients with low SMD (29.0%) and those with normal SMD (22.7%). On the other hand, there was a trend toward a lower response rate in the patients with low SMI (18.9%) than in those with normal SMI (43.5%, P=0.06).

Multivariate analysis

Multivariate analysis confirmed that low SMD was an independent predictor of poor outcomes; the hazard ratio (HR) for death in patients with low SMD versus those with normal SMD was 2.72 (95% CI, 1.32 to 5.82; P<0.01; Table III). The other independent predictors of poor OS included a PS of 2 (HR, 3.25; 95% CI, 1.21 to 7.88; P=0.02), two or more metastatic sites of disease (HR, 2.48; 95% CI, 1.13 to 5.55; P=0.03), and no response to first-line therapy (HR, 4.18; 95% CI, 1.65 to 11.6; P<0.01).

Table III

Multivariate Cox proportional-hazards model for survival.

Table III

Multivariate Cox proportional-hazards model for survival.

VariableHR95% CIP-value
SMD<0.01a
 NormalRef0.17–0.76
 Low2.721.32–5.82
SMI0.98
 NormalRef0.42–2.57
 Low1.010.39–2.38
PS0.02a
 0 to 1Ref0.13–0.83
 23.251.21–7.88
Age (years)0.10
 ≤65Ref0.24–1.14
 >651.910.87–4.21
Gender0.13
 MaleRef0.81–4.97
 Female0.510.20–1.23
No. of metastatic site0.03a
 <2Ref0.18–0.89
 ≥22.481.13–5.55
Response to first-line therapy<0.01a
 YesRef0.08–0.60
 No4.251.65–11.6

a Statistically significant difference (P<0.05). SMD, skeletal muscle density; SMI, skeletal mass index; PS, performance status; HR, hazard ratio.

Discussion

This is the first study to demonstrate that low SMD is an independent poor prognostic factor in metastatic gastric cancer patients receiving chemotherapy. Also, in contrast to reports of other cancers, our study showed that sarcopenia or low SMI is not associated with prognosis in metastatic gastric cancer. On the other hand, PFS and tumor response were not associated with either SMD or SMI in this study. These findings imply that low SMD, is an important prognostic indicator in patients with gastric cancer who are considered eligible for systemic chemotherapy, but is not a predictor of the response to chemotherapy.

The association of SMD, but not SMI, with survival outcomes in our present study is an intriguing finding. One plausible explanation for this could be that the increase in lipid content of muscle occurs before the decline in muscle mass and therefore, the decrease in SMD is detected earlier than the decrease in SMI. Also, CT based calculation allows for early detection of fall in HU (SMD) while the muscle area remains unchanged; thus decrease in SMD is detected earlier than corresponding decrease in SMI. Because most of the previous studies have examined either SMD or SMI only, this discrepancy has not yet been revealed in those studies. Second, many gastric cancer patients will have undergone gastrectomy as a part of their treatment. Gastrectomy, in itself, has been known to decrease muscle mass (16). Abdiev et al reported that after gastrectomy the skeletal muscle mass decreased to around 85% of preoperative level (17). Therefore, the low SMI found among patients in this study could have partly been an effect of gastrectomy. These results suggest that SMD would better facilitate assessment of the risk-benefit ratio of systemic chemotherapy in metastatic gastric cancer patients compared to SMI.

PFS, tumor response and toxicities were not related to SMD or SMI in this study. Nevertheless, both tumor response and toxicities were numerically better in normal SMI group versus low SMI group.

Some other studies in patients with diseases such as breast cancer or renal cell carcinoma have also reported that lower SMI may be linked to severe toxicities, leading to dose reduction and then to a shorter time to progression (12,13). The clinical value of skeletal muscle assessment is thus considered to warrant further investigation in gastric cancer patients.

Our study had several important limitations. It was retrospective and had a small sample size. In addition, the cut-off values that we used for SMD and SMI were based on the results of a study done in a Western population (10), because there is no clear consensus about the Asian-specific cut-off values of SMD and SMI.

In conclusion, our results demonstrated that low skeletal muscle density, rather than low skeletal muscle mass, is independently associated with poor survival in patients who receive chemotherapy for metastatic gastric cancer.

Acknowledgments

We are grateful to Dr Kenta Murotani, Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, for his expert assistance in statistical analyses. This study was partly supported by JSPS KAKENHI grant number 2646026.

References

1 

Prado CM, Lieffers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L and Baracos VE: 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. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Peng P, Hyder O, Firoozmand A, Kneuertz P, Schulick RD, Huang D, Makary M, Hirose K, Edil B, Choti MA, et al: Impact of sarcopenia on outcomes following resection of pancreatic adenocarcinoma. J Gastrointest Surg. 16:1478–1486. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Harimoto N, Shirabe K, Yamashita YI, Ikegami T, Yoshizumi T, Soejima Y, Ikeda T, Maehara Y, Nishie A and Yamanaka T: Sarcopenia as a predictor of prognosis in patients following hepatectomy for hepatocellular carcinoma. Br J Surg. 100:1523–1530. 2013. View Article : Google Scholar : PubMed/NCBI

4 

van Vledder MG, Levolger S, Ayez N, Verhoef C, Tran TC and Ijzermans JN: Body composition and outcome in patients undergoing resection of colorectal liver metastases. Br J Surg. 99:550–557. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Lanic H, Kraut-Tauzia J, Modzelewski R, Clatot F, Mareschal S, Picquenot JM, Stamatoullas A, Leprêtre S, Tilly H and Jardin F: Sarcopenia is an independent prognostic factor in elderly patients with diffuse large B-cell lymphoma treated with immunochemotherapy. Leuk Lymphoma. 55:817–823. 2014. View Article : Google Scholar

6 

Tan BH, Birdsell LA, Martin L, Baracos VE and Fearon KC: Sarcopenia in an overweight or obese patient is an adverse prognostic factor in pancreatic cancer. Clin Cancer Res. 15:6973–6979. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Goodpaster BH, Park SW, Harris TB, Kritchevsky SB, Nevitt M, Schwartz AV, Simonsick EM, Tylavsky FA, Visser M and Newman AB: The loss of skeletal muscle strength, mass, and quality in older adults: The health, aging and body composition study. J Gerontol A Biol Sci Med Sci. 61:1059–1064. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Goodpaster BH, Kelley DE, Thaete FL, He J and Ross R: Skeletal muscle attenuation determined by computed tomography is associated with skeletal muscle lipid content. J Appl Physiol (1985). 89:104–110. 2000.

9 

Delmonico MJ, Harris TB, Visser M, Park SW, Conroy MB, Velasquez-Mieyer P, Boudreau R, Manini TM, Nevitt M, Newman AB, et al: Health, Aging, and Body: Longitudinal study of muscle strength, quality, and adipose tissue infiltration. Am J Clin Nutr. 90:1579–1585. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Martin L, Birdsell L, Macdonald N, Reiman T, Clandinin MT, McCargar LJ, Murphy R, Ghosh S, Sawyer MB and Baracos VE: 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. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Antoun S, Lanoy E, Iacovelli R, Albiges-Sauvin L, Loriot Y, Merad-Taoufik M, Fizazi K, di Palma M, Baracos VE and Escudier B: Skeletal muscle density predicts prognosis in patients with metastatic renal cell carcinoma treated with targeted therapies. Cancer. 119:3377–3384. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Prado CM, Baracos VE, McCargar LJ, Reiman T, Mourtzakis M, Tonkin K, Mackey JR, Koski S, Pituskin E and Sawyer MB: 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. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Antoun S, Baracos VE, Birdsell L, Escudier B and Sawyer MB: Low body mass index and sarcopenia associated with dose-limiting toxicity of sorafenib in patients with renal cell carcinoma. Ann Oncol. 21:1594–1598. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Koizumi W, Narahara H, Hara T, Takagane A, Akiya T, Takagi M, Miyashita K, Nishizaki T, Kobayashi O, Takiyama W, et al: S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer (SPIRITS trial): A phase III trial. Lancet Oncol. 9:215–221. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Mitsiopoulos N, Baumgartner RN, Heymsfield SB, Lyons W, Gallagher D and Ross R: Cadaver validation of skeletal muscle measurement by magnetic resonance imaging and computerized tomography. J Appl Physiol (1985). 85:115–122. 1998.

16 

Yamaoka Y, Fujitani K, Tsujinaka T, Yamamoto K, Hirao M and Sekimoto M: Skeletal muscle loss after total gastrectomy, exacerbated by adjuvant chemotherapy. Gastric Cancer. 18:382–389. 2015. View Article : Google Scholar

17 

Abdiev S, Kodera Y, Fujiwara M, Koike M, Nakayama G, Ohashi N, Tanaka C, Sakamoto J and Nakao A: Nutritional recovery after open and laparoscopic gastrectomies. Gastric Cancer. 14:144–149. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 35 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hayashi N, Ando Y, Gyawali B, Shimokata T, Maeda O, Fukaya M, Goto H, Nagino M and Kodera Y: Low skeletal muscle density is associated with poor survival in patients who receive chemotherapy for metastatic gastric cancer. Oncol Rep 35: 1727-1731, 2016
APA
Hayashi, N., Ando, Y., Gyawali, B., Shimokata, T., Maeda, O., Fukaya, M. ... Kodera, Y. (2016). Low skeletal muscle density is associated with poor survival in patients who receive chemotherapy for metastatic gastric cancer. Oncology Reports, 35, 1727-1731. https://doi.org/10.3892/or.2015.4475
MLA
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 35.3 (2016): 1727-1731.
Chicago
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 35, no. 3 (2016): 1727-1731. https://doi.org/10.3892/or.2015.4475