Effect of pretreatment psoas muscle mass on survival for patients with unresectable pancreatic cancer undergoing systemic chemotherapy

  • Authors:
    • Noriko Ishii
    • Yoshinori Iwata
    • Hiroki Nishikawa
    • Hirayuki Enomoto
    • Nobuhiro Aizawa
    • Akio Ishii
    • Yuho Miyamoto
    • Yukihisa Yuri
    • Kunihiro Hasegawa
    • Chikage Nakano
    • Takashi Nishimura
    • Kazunori Yoh
    • Yoshiyuki Sakai
    • Naoto Ikeda
    • Tomoyuki Takashima
    • Ryo Takata
    • Hiroko Iijima
    • Shuhei Nishiguchi
  • View Affiliations

  • Published online on: September 15, 2017     https://doi.org/10.3892/ol.2017.6952
  • Pages: 6059-6065
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

To the best of our knowledge, there are few previous studies that have investigated the effect of decreased skeletal muscle mass (DSMM) on survival in patients with unresectable advanced pancreatic cancer (APC) who are undergoing systemic chemotherapy. Thus, the present study aimed to investigate the impact of DSMM, as determined by the psoas muscle index (PMI) following computed tomography and prior to systemic chemotherapy, on the outcomes of patients with unresectable APC (n=61). The primary endpoint used was the overall survival (OS) rate. The OS rates in the PMI‑High group (exceeds the median PMI value in each gender) were retrospectively compared with those in the PMI‑Low group (below the median PMI value in each gender), and factors associated with OS were investigated using univariate and multivariate analyses. The study cohort included 31 male and 30 female patients with a median age of 72 years, 13 of whom were stage IVA, and 48 were stage IVB. The median PMI in males was 4.3 cm2/m2 (range, 1.6‑8.2 cm2/m2), while that in females was 2.3 cm2/m2 (range, 0.7‑6.1 cm2/m2). The proportion of patients with performance status 0 in the PMI‑High group was significantly high, compared with that in the PMI‑Low group [83.3% (25/30) vs. 58.1% (18/31); P=0.0486]. Body mass index in the PMI‑High group was significantly higher compared with that in the PMI‑Low group (P=0.0154). The 1‑year cumulative survival rate was 43.3% in the PMI‑High group and 12.9% in the PMI‑Low group (P=0.0027). Following multivariate analysis, PMI (P=0.0036), prothrombin time (P=0.0044) and carbohydrate antigen 19‑9 (P=0.0451) were identified to be significant predictors of OS. In conclusion, DSMM, as determined by the PMI, could be a significant predictor of prognosis in patients with unresectable APC who are receiving systemic chemotherapy.

Introduction

The pancreas serves an essential role in the digestive system, including producing numerous digestive enzymes (13). In 2013, pancreatic cancer (PC) was reported to be the fourth leading cause of cancer-associated mortality worldwide (4). The majority of patients with PC present with locally advanced or metastatic disease at initial diagnosis and the proportion of patients who can proceed with curative intent surgery is <20% (1,3). Patients with advanced (A)PC have a poor prognosis (59). Among patients with metastatic PC, the 5-year survival rate is reported to be ~2% (10). Current Japanese guidelines for systemic chemotherapy in patients with APC recommend the use of gemcitabine monotherapy, S-1 monotherapy, gemcitabine and S-1 combination therapy, nab-paclitaxel and gemcitabine combination therapy, or a combination chemotherapy regimen consisting of oxaliplatin, irinotecan, fluorouracil and leucovorin, based on the baseline and tumor status of each patient (58).

Skeletal muscle is considered to be a large endocrine organ, which accounts for ~50% of an individual's body weight and possesses the capacity for high metabolic activity (11). In general, skeletal muscle mass is regulated depending on the balance between protein synthesis and protein catabolism (12). Sarcopenia, defined as decreased skeletal muscle mass (DSMM) and muscle strength, has become a relevant clinical feature for understanding the effects of aging on clinical outcomes (13). Sarcopenia is a commonly observed disorder in aged populations and is associated with disability, functional decline and frailty (13,14). Age-associated sarcopenia is defined as primary sarcopenia, whilst advanced malignancies, as well as chronic inflammatory diseases including renal, heart and liver diseases, can be the causes of secondary sarcopenia (1216). Severe underlying diseases can lead to sarcopenia and cachexia, which involves body weight loss and muscle wasting. Furthermore, substantial skeletal muscle wasting is an important predictor in patients with solid malignancies, although the precise mechanisms by which DSMM increases the risk of mortality remain unclear (17,18). Knowledge of the underlying mechanisms in advanced malignancies associated with skeletal muscle wasting may lead to the development of novel therapeutic drugs. Thus, in recent years, this clinical area has attracted much attention among oncologists.

A number of studies have demonstrated that DSMM could be an adverse predictor for patients with PC who were treated with surgical resection (16,1923). However, to the best of our knowledge, there are few reports regarding the impact of DSMM on survival in patients with unresectable APC undergoing systemic chemotherapy (24,25). Therefore, it is imperative to address these issues. Thus, the aims of the present study were to investigate the impact of DSMM prior to systemic chemotherapy on the clinical outcomes of patients with unresectable APC.

Patients and methods

Patients and indications for systemic chemotherapy

Between February 2008 and November 2015, 80 consecutive patients diagnosed with unresectable APC undergoing systemic chemotherapy were admitted to Hyogo College of Medicine (Nishinomiya, Hyogo, Japan). There were 31 male and 30 female patients with a median age of 72 years (range, 39–89). All patients were treatment naive for APC. Cases with distal common bile duct cancer, ampulla of Vater carcinoma or neuroendocrine carcinoma of the pancreas were excluded. Of these, 19 patients with unknown clinical outcomes (succumbed to disease or surviving) due to loss of follow-up were excluded from the current analysis. Thus, a total of 61 patients with APC who underwent systemic chemotherapy were analyzed in the present study. Clinical stage for APC was determined based on Union for International Cancer Control (UICC) classification system (26). In cases with local APC without distant metastases, indication for surgery was reviewed in each case through discussion with oncologists and surgeons (2731). In principal, systemic chemotherapy was recommended for patients with PC with the following characteristics, as determined by radiological findings: Dynamic computed tomography (CT), magnetic resonance imaging and endoscopic ultrasonography (EUS). This was following informed consent from each patient. The presence of distant metastases and/or the presence of tumor vascular invasion was judged as unresectable PC. Patients with poor performance status [PS; Eastern Cooperative Oncology Group (ECOG) classification 3] were not recommended for systemic chemotherapy (32). The presence of ascites was not contraindicated for systemic chemotherapy.

Definition of DSMM and the study protocol

Assessment of muscle mass was performed using CT scans obtained prior to systemic chemotherapy. The third lumber (L3) level was selected as a standard. Bilateral psoas muscles at the L3 level were identified on the CT images. Cross-sectional areas (cm2) of these muscles were measured by manual tracing on the CT images, and their sum was calculated. These sums were normalized for each patient to provide a psoas muscle index value (PMI; cm2/m2) (33,34). The median PMI value was calculated for males and females separately. Patients with a PMI of more than each median value were defined as the PMI-High (H) group and those with a PMI of less than each median value as the PMI-Low (L) group. This is due to the optimal cut-off point of PMI for DSMM in Japanese patients with PC having not yet been well established.

The primary endpoint was overall survival (OS) in the present study. Baseline characteristics and OS in the PMI-H and PMI-L groups were retrospectively compared, and factors associated with OS were investigated using univariate and multivariate analyses. The current study was performed in accordance with the Declaration of Helsinki and with approval from the Ethics Committee of Hyogo College of Medicine (approval no. 2117).

Diagnosis for pancreatic cancer and systemic chemotherapy

PC was diagnosed primarily based on the current guidelines (35). Briefly, abdominal US and dynamic CT of the pancreas was routinely performed prior to initiating systemic chemotherapy (33). In cases with atypical radiological findings for PC, tumor biopsy or EUS-guided fine needle aspiration was considered (36). In the present study, the pathological diagnosis was confirmed in 15 cases (24.6%).

The selection of chemotherapeutic agents was determined by each attending physician. For patients with no evident risk factors, the recommended initial dosage of each chemotherapeutic agent (gemcitabine, S-1, nab-paclitaxel, and 5-fluorouracil) was administered (5,37). The reduced initial dosage was administered to certain patients based on clinical features, including age, body weight, ECOG-PS and laboratory data. During systemic chemotherapy, each attending physician adjusted the dosage of chemotherapeutic agents according to the grade of adverse events. In patients with adverse events, systemic chemotherapy was discontinued until the clinical symptoms resolved to grade 1 or 2, and other alternative chemotherapeutic regimens were considered. In patients with poor response to initial chemotherapy, other alternative chemotherapeutic regimens were also considered.

In principle, the treatment efficacy for systemic chemotherapy was assessed every 2–4 months following the initiation of chemotherapy, according to the Response Evaluation Criteria in Solid Tumors (RECIST; version 1.1) using radiological findings and/or the levels of various tumor markers, including carcinoembryonic antigen (CEA) and carbohydrate antigen (CA) 19-9 (38). Patients continued systemic chemotherapy until the development of any of the following conditions: Unacceptable drug toxicity, tumor progression or the patient's request to stop treatment. Chemotherapy-associated adverse events were evaluated using the Common Terminology Criteria for Adverse Events (CTCAE; version 3.0) (39).

Evaluation of treatment response during chemotherapy

The most improved treatment response achieved during chemotherapy was determined according to the RECIST criteria (version 1.1), as previously described (7,38). The most improved treatment response was graded using the following four categories: (1) Complete response (CR); (2) partial response (PR); (3) stable disease (SD); (4) progressive disease (PD) (38). The objective response rate (ORR) was defined as the proportion of patients with the most improved treatment response rates when considering CR and PR. The disease control rate (DCR) was defined as the proportion of patients with the most improved treatment response rates when considering CR, PR and SD.

Statistical analysis

The categorical parameters in the PMI-H and PMI-L groups were analyzed using Fisher's exact test, while the numerical parameters were analyzed either with an unpaired Student's t-test or with a Mann-Whitney U test as appropriate. OS curves were created using the Kaplan-Meier estimator method and compared using the log-rank test. Variables that were considered significant following univariate analysis were entered into the multivariate analysis with Cox's proportional hazards model. For the purpose of analyzing the significance of predictors in multivariate analyses, analyzed variables were divided by the median values for all cases (n=61) and treated as dichotomous covariates. OS was defined as the time interval from the initiation of systemic chemotherapy until mortality (due to any cause) or to the final follow-up visit. Data are presented as the median values (range) unless otherwise stated. P<0.05 was considered to indicate a statistically significant difference. All statistical analyses were performed using JMP software (version 11.0; SAS Institute Inc., Cary, NC, USA).

Results

Baseline characteristics

The baseline characteristics of the analyzed patient cohort (n=61) are presented in Table I. Of these, 13 were stage IVA and 48 were stage IVB, as determined using the UICC classification system. Maximum tumor size in patients ranged between 1.4 and 9.4 cm (median, 3.6 cm). The median PMI in males was 4.3 cm2/m2 (range, 1.6–8.2 cm2/m2), whereas in females it was 2.3 cm2/m2 (range, 0.7–6.1 cm2/m2). Patients were predominantly PS-0 (70.5%; 43/61). As for initial chemotherapeutic regimens, gemcitabine monotherapy was performed in 44 patients, S-1 monotherapy in 10, gemcitabine and S-1 combination therapy in 5, nab-paclitaxel and gemcitabine combination therapy in 1, and 5-fluorouracil monotherapy in 1.

Table I.

Baseline characteristics of patients with unresectable advanced pancreatic cancer undergoing systemic chemotherapy (n=61).

Table I.

Baseline characteristics of patients with unresectable advanced pancreatic cancer undergoing systemic chemotherapy (n=61).

VariableValue (range)
Age, years72 (39–89)
Gender, male/female31/30
ECOG-performance status, 0/1/243/15/3
Psoas muscle index, cm2/m2, male4.3 (1.6–8.2)
Psoas muscle index, cm2/m2, female2.3 (0.7–6.1)
Body mass index, kg/m221.2 (15.1–31.6)
Pancreatic cancer stage, IVA/IVB13/48
Maximum tumor size, cm3.6 (1.4–9.4)
Primary site, uncus or head/body or tail33/28
Total bilirubin, mg/dl0.7 (0.3–6.6)
Serum albumin, g/dl3.5 (1.8–4.4)
Prothrombin time, %86.3 (47.5–127)
Platelet count, ×104/mm320.8 (7.1–45.9)
White blood cell, ×103/µl6.05 (2.54–29.76)
Hemoglobin, g/dl11.6 (7.5–15.7)
Serum creatinine, mg/dl0.65 (0.28–7.41)
C reactive protein, mg/dl0.6 (0–22.0)
AST, IU/l26 (11–265)
ALT, IU/l30 (8–289)
ALP, IU/l361 (139–1929)
GGT, IU/l98 (11–747)
Amylase, IU/l66 (7–357)
CEA, IU/la3.95 (1.4–286.1)
CA19-9, IU/l408 (0.6–42414)

{ label (or @symbol) needed for fn[@id='tfn1-ol-0-0-6952'] } Data are presented as the number of patients or the median (range).

a Missing data, n=3. ECOG, the Eastern Cooperative Oncology Group; AST, aspartate aminotransferase; ALT, alanine aminotransferase; ALP, alkaline phosphatase; GGT, gamma glutamyl transpeptidase; CEA, carcinoembryonic antigen; CA19-9, carbohydrate antigen 19-9.

Comparison of baseline characteristics between the PMI-H and the PMI-L group

The proportion of patients with PS-0 in the PMI-H group was significantly higher compared with that in the PMI-L group [83.3% (25/30) vs. 58.1% (18/31); P=0.0486]. Body mass index (BMI) in the PMI-H group was significantly higher compared with that in the PMI-L group (P=0.0154). As for other baseline characteristics, no significant differences were identified between the two groups (Table II).

Table II.

Comparison of baseline characteristics between the PMI-H and the PMI-L group.

Table II.

Comparison of baseline characteristics between the PMI-H and the PMI-L group.

Value (range)

VariablesPMI-HPMI-LP-value
Age, years70 (39–89)73 (48–88)0.1350
Sex, male/female15/1515/161.0000
ECOG-performance status, 0/1/225/518/130.0486
Body mass index, kg/m221.6 (17.3–31.6)19.9 (15.1–24.8)0.0154
Pancreatic cancer stage, IVA/IVB9/214/270.1271
Maximum tumor size, cm2.95 (1.4–9.4)3.8 (2.2–7.6)0.1635
Primary site, uncus or head/body or tail16/1417/141.0000
Best treatment response, CR/PR/SD/PD/NE0/4/12/11/30/3/8/13/7ORR, 0.7072/DCR, 0.2016
Total bilirubin, mg/dl0.7 (0.3–1.8)0.8 (0.3–6.6)0.7062
Serum albumin, g/dl3.6 (1.8–4.4)3.4 (2.5–4.4)0.2806
Prothrombin time, %86.15 (43.5–109.8)86.5 (64.7–127)0.5345
Platelet count, ×104/mm321.4 (9.1–45.9)19.5 (7.1–35.3)0.3124
White blood cell, ×103/µl5.92 (3.36–12.75)6.34 (2.54–29.76)0.5836
Hemoglobin, g/dl12.0 (7.6–15.1)11.5 (7.5–15.7)0.5342
Serum creatinine, mg/dl0.64 (0.33–1.28)0.65 (0.28–7.41)0.8062
C reactive protein, mg/dl0.4 (0–22.0)0.9 (0–6.9)0.3041
AST, IU/l31.5 (11–265)25 (12–124)0.7127
ALT, IU/l34 (8–289)29 (8–109)0.6084
ALP, IU/l387.5 (153–1358)338.5 (139–1929)0.7096
GGT, IU/l90.5 (14–747)98 (11–467)0.8140
Amylase, IU/l66 (7–201)67 (17–357)0.5255
CEA, IU/l3.2 (1.5–96.2)4.1 (1.4–286.1)0.1591
CA19-9, IU/l286.4 (0.6–42414)801.7 (0.6–31020)0.2741

[i] Data are presented as the number of patients or the median (range). PMI, psoas muscle index; H, high; L, low; CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; NE, not evaluated; ORR, objective response rate; DCR, disease control rate; ECOG, the Eastern Cooperative Oncology Group; AST, aspartate aminotransferase; ALT, alanine aminotransferase; ALP, alkaline phosphatase; GGT, gamma glutamyl transpeptidase; CEA, carcinoembryonic antigen; CA19-9, carbohydrate antigen 19-9.

Cumulative OS rates for all cases and comparison of OS rates between the PMI-H and the PMI-L group

The median follow-up period following initial systemic chemotherapy for all cases was 246 days (range, 25–1,304 days). For all cases, the 6 month, 1- and 2-year cumulative survival rates were 59.0, 27.9 and 9.1%, respectively (Fig. 1). The median follow-up period following initial systemic chemotherapy was 357 days (range, 25–1,304 days) in the PMI-H group and 155 days (range, 26–900 days) in the PMI-L group. The 6 month, 1- and 2-year cumulative survival rates were 73.3, 43.3 and 15.2%, respectively, in the PMI-H group, and 45.2, 12.9 and 3.2%, respectively, in the PMI-L group (P=0.0027; Fig. 2).

Comparison of serious adverse events (SAEs) of grade ≥3 between the PMI-H and the PMI-L groups

The prevalence of chemotherapy-associated SAEs of grade ≥3, as assessed using CTCAE (version 3.0), were 10.0% (3/30) in the PMI-H group and 25.8% (8/31) in the PMI-L group (P=0.1822; data not presented). In the PMI-H group, SAEs of grade ≥3 included severe vomiting (1 patient), severe neuropathy (1 patient) and neutropenia (1 patient). In the PMI-L group, SAEs of grade ≥3 included interstitial pneumonia (1 patient), severe anemia (1 patient), thrombocytopenia (2 patients), liver injury (1 patient), neutropenia (1 patient) and jaundice (2 patients). In the present study, chemotherapy-associated mortality was not observed (data not presented).

Most improved tumor treatment response during chemotherapy

With regard to the most improved treatment response during chemotherapy, out of all cases CR was achieved in 0, PR in 7, SD in 20, PD in 24 and not evaluated (NE) in 10 patients (Table II). The ORR and DCR were calculated to be 11.5% (7/61) and 44.3% (27/61), respectively. In the analysis of the most improved tumor response in the PMI-H group, CR was achieved in 0, PR in 4, SD in 12, PD in 11 and NE in 3 patients. The ORR and DCR were calculated to be 13.3% (4/30) and 53.3% (16/30), respectively. In the analysis of the most improved tumor response in the PMI-L group, CR was achieved in 0, PR in 3, SD in 8, PD in 13 and NE in 7 patients. The ORR and DCR were calculated to be 9.7% (3/31) and 35.5% (11/31), respectively. No significant differences in the most improved treatment response were identified between the PMI-H and PMI-L groups (ORR, P=0.7072; DCR, P=0.2016; Table II).

Causes of mortality

During the follow-up period, 60 patients (98.4%) succumbed to disease. In the PMI-H group, 29 (96.7%) patients succumbed during the follow-up period. All patients succumbed due to tumor progression. In the PMI-L group, 31 (100%) patients succumbed during the follow-up period. All patients succumbed due to tumor progression.

Univariate and multivariate analyses of parameters contributing to OS

The univariate analysis identified that the following factors significantly contributed to OS for all cases (n=61): PMI (H or L; P=0.0027); tumor stage (IVA or IVB; P=0.0026); maximum tumor size (>3.6 cm or ≤3.6 cm P=0.0262); prothrombin time (PT; >86.3% or ≤86.3%; P=0.0052); C reactive protein (CRP; >0.6 mg/dl or ≤0.6 mg/dl; P=0.0445); gamma glutamyl transpeptidase (>98 IU/l or ≤98 IU/l; P=0.0175); CA 19-9 >408 IU/l or ≤408 IU/l (P=0.0008) (Table III). The hazard ratios and 95% confidence intervals determined by multivariate analysis for the 7 variables (selected based on a P<0.05 in univariate analysis) are detailed in Table III. On multivariate analysis, PMI (H or L; P=0.0036), PT (>86.3% or ≤86.3%; P=0.0044) and CA19-9 (>408 IU/l or ≤408 IU/l; P=0.0451) were identified as significant predictors of OS.

Table III.

Univariate and multivariate analyses of factors associated with overall survival for patients with unresectable advanced pancreatic cancer (n=61).

Table III.

Univariate and multivariate analyses of factors associated with overall survival for patients with unresectable advanced pancreatic cancer (n=61).

Multivariate analysis

VariablesNo. of patientsUnivariate analysisHazard ratio95% CIP-value
Age, >72/≤72 years29/320.4593
Sex, male/female31/300.3967
ECOG-performance status, 0–1/243/180.6853
PMI, high/low30/310.00272.4461.340–4.5410.0036a
Body mass index, >21.2/≤21.2 kg/m230/310.5700
Pancreatic cancer stage, IVA/IVB13/480.00262.1470.934–5.2070.0725
Maximum tumor size, >3.6/≤3.6 cm29/320.02620.9010.489–1.6480.7351
Primary site, uncus or head/body or tail33/280.1758
Total bilirubin, >0.7/≤0.7 mg/dl29/320.3342
Serum albumin, >3.5/≤3.5 g/dl28/330.2986
Prothrombin time, >86.3/≤86.3%30/310.00522.2191.283–3.8740.0044a
Platelet count, >20.8/≤20.8 ×104/mm330/310.0951
WBC, >6.05/≤6.05×103/µl30/310.4832
Hemoglobin, >11.6/≤11.6 g/dl30//310.4077
Serum creatinine, >0.65/≤0.65 mg/dl28/330.6884
CRP, >0.6/≤0.6 mg/dl30/310.04450.6650.384–1.1560.1474
AST, >26/≤26 IU/l29/320.9588
ALT, >30/≤30 IU/l30/310.1678
ALP, >361/≤361 IU/l29/320.5993
GGT, >98/≤98 IU/l30/310.01750.8080.425–1.5090.5053
Amylase, >66/≤66 IU/l29/320.7084
CEA, >3.95/≤3.95 IU/l29/290.1372
CA19-9, >408/≤408 IU/l30/310.00080.5040.251–0.9850.0451a

a P<0.05. ECOG, the Eastern Cooperative Oncology Group; AST, aspartate aminotransferase; ALT, alanine aminotransferase; ALP, alkaline phosphatase; GGT, gamma glutamyl transpeptidase; CEA, carcinoembryonic antigen; CA19-9, carbohydrate antigen 19-9; CI, confidence interval; HR, hazard ratio; WBC, white blood cell; CRP, C reactive protein; PMI, psoas muscle index.

Discussion

The effect of muscle mass depletion on clinical outcomes in solid malignancies is a relevant topic among oncologists (17,18). However, as aforementioned, few reports have addressed this important clinical question in patients with unresectable APC who are receiving systemic chemotherapy (24,25). The present study was, therefore, conducted. The data of the present study revealed that subjects in the PMI-H group survived significantly longer compared with those in the PMI-L group (P=0.0027) and additionally, lower PMI was revealed to be an independent adverse predictor for survival. These results suggest that pretreatment PMI is useful for predicting outcomes for unresectable patients with APC undergoing systemic chemotherapy. Since the majority of previous reports have focused on the effect of skeletal muscle mass on survival for patients undergoing surgery, the results of the current study may be worth reporting (16,1923).

For the baseline PMI values, the median PMI in males was 4.3 cm2/m2 (range, 1.6–8.2 cm2/m2), whereas in females it was 2.3 cm2/m2 (range, 0.7–6.1 cm2/m2). However, Hamaguchi et al (34) demonstrated that the PMI figure below two standard deviations of the mean among 541 healthy living donors for liver transplantation were 6.36 cm2/m2 for males and 3.92 cm2/m2 for females. When these cut-off values are applied to the current cohort, 55 patients (90.2%) were determined to have muscle mass loss. The significant discrepancy between the present data and the results of Hamaguchi et al (34) for baseline PMI may be attributed to the presence of advanced malignancies. Advanced malignancies themselves can cause severe muscle mass loss (17,18). Regarding comparison of baseline characteristics between the PMI-H and PMI-L groups, ECOG-PS and BMI were identified to be significant factors. DSMM is associated with disability, functional decline, poorer nutritional status and frailty, which may lead to poorer PS and lower BMI (13,14). However, aging was not identified to be a significant factor in the present study. Advanced malignancies, rather than just aging, may also affect skeletal muscle loss (17,18). The proportion of SAEs of grade ≥3 in the PMI-H group was higher, as compared with in the PMI-L group, although the difference in the two groups did not reach significance in the present study. The majority of previous studies demonstrated that DSMM can increase the risk of development of surgery-associated complications for patients with PC (16,1923). Thus, caution for the development of SAEs during chemotherapy should be exercised, particularly in patients with PC with lower skeletal muscle mass.

CA19-9 was identified as an independent predictor for survival in the multivariate analysis. CA19-9 is the pancreatic cancer biomarker currently recommended for clinical use, and numerous reports have revealed that elevated CA 19-9 levels are associated with a worse survival rate, which are concordant with the results of the current study (35,40). In addition, CA19-9 levels in stage IVB patients were significantly higher compared with those in stage IVA patients (P=0.0060), as determined in the present study, which indicates that this biomarker effectively reflects tumor status. However, CRP is an inflammation marker and elevated CRP levels have been demonstrated to be adverse predictive factors in patients with solid malignancies (41). Although CRP was not identified to be significant in the present multivariate analysis, this marker may be important for predicting outcomes.

Clinical evidence that physical activity is beneficial for patients with solid malignancies in reducing chemotherapy-associated symptoms and improving quality of life, as well as drug tolerance and drug adherence for chemotherapy, has previously been reported (42). However, the effects of physical activity in patients with APC undergoing systemic chemotherapy remain unclear. Currently, a randomized controlled trial for investigating the impact of physical activity on outcomes in patients with APC is underway (43). If positive results are obtained in the study, treatment strategies for patients with APC receiving systemic chemotherapy may be altered in the future.

A number of limitations must be acknowledged with regard to the present study. Firstly, it was a retrospective observational study and biases inherent to retrospective analyses could not be completely removed. Secondly, the initial chemotherapeutic agents differed between the patients and these therapies could have potentially caused bias for clinical outcomes. Thirdly, the sample size was relatively small for analysis, potentially creating bias. Fourth, muscle quality as reflected by muscle strength was not evaluated in the current analysis. Finally, the present study population only included Japanese patients with PC with relatively low body weights compared with patients with PC in Western countries (18). Therefore, these results may not be directly applied to different ethnic populations. However, the results of the present study demonstrated that DSMM is associated with the clinical outcomes of patients with APC undergoing systemic chemotherapy.

In conclusion, DSMM as determined by PMI may be a significant predictor of prognosis in patients with APC receiving systemic chemotherapy. In such patients, appropriate interventions may be required for ameliorating the clinical outcome.

Acknowledgements

The authors would like to thank all medical staff at the Department of Internal Medicine, Hyogo College of Medicine (Nishinomiyashi, Japan) for their assistance.

Glossary

Abbreviations

Abbreviations:

APC

advanced pancreatic cancer

DSMM

decreased skeletal muscle mass

UICC

Union for International Cancer Control

CT

computed tomography

EUS

endoscopic ultrasonography

PS

performance status

ECOG

Eastern Cooperative Oncology Group

L3

the third lumber

PMI

psoas muscle index

H

high

L

low

OS

overall survival

RECIST

Response Evaluation Criteria in Solid Tumors

CEA

carcinoembryonic antigen

CA19-9

carbohydrate antigen 19-9

CTCAE

Common Terminology Criteria for Adverse Events

CR

complete response

PR

partial response

SD

stable disease

PD

progressive disease

ORR

objective response rate

DCR

disease control rate

SAE

serious adverse event

NE

not evaluated

PT

prothrombin time

CRP

C reactive protein

References

1 

Kamisawa T, Wood LD, Itoi T and Takaori K: Pancreatic cancer. Lancet. 388:73–85. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Sudo K, Nakamura K and Yamaguchi T: S-1 in the treatment of pancreatic cancer. World J Gastroenterol. 20:15110–15118. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Cid-Arregui A and Juarez V: Perspectives in the treatment of pancreatic adenocarcinoma. World J Gastroenterol. 21:9297–9316. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Malvezzi M, Bertuccio P, Levi F, La Vecchia C and Negri E: European cancer mortality predictions for the year 2013. Ann Oncol. 24:792–800. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Ueno H, Ioka T, Ikeda M, Ohkawa S, Yanagimoto H, Boku N, Fukutomi A, Sugimori K, Baba H, Yamao K, et al: Randomized phase III study of gemcitabine plus S-1, S-1 alone, or gemcitabine alone in patients with locally advanced and metastatic pancreatic cancer in Japan and Taiwan: GEST study. J Clin Oncol. 31:1640–1648. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Imaoka H, Kou T, Tanaka M, Egawa S, Mizuno N, Hijioka S, Hara K, Yazumi S, Shimizu Y and Yamao K: Clinical outcome of elderly patients with unresectable pancreatic cancer treated with gemcitabine plus S-1, S-1 alone, or gemcitabine alone: Subgroup analysis of a randomised phase III trial, GEST study. Eur J Cancer. 54:96–103. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, et al: Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 369:1691–1703. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Kaga Y, Sunakawa Y, Kubota Y, Tagawa T, Yamamoto T, Ikusue T, Uto Y, Miyashita K, Toshima H, Kobayashi K, et al: Early tumor shrinkage as a predictor of favorable outcomes in patients with advanced pancreatic cancer treated with FOLFIRINOX. Oncotarget. 7:67314–67320. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Boursi B, Finkelman B, Giantonio BJ, Haynes K, Rustgi AK, Rhim AD, Mamtani R and Yang YX: A clinical prediction model to assess risk for pancreatic cancer among patients with new-onset diabetes. Gastroenterology. 152:840–850.e3. 2017. View Article : Google Scholar : PubMed/NCBI

10 

American Cancer Society, . Cancer Facts & Figures 2013. American Cancer Society, Inc.; Atlanta, GA: 2013

11 

Muller MJ, Wang Z, Heymsfield SB, Schautz B and Bosy-Westphal A: Advances in the understanding of specific metabolic rates of major organs and tissues in humans. Curr Opin Clin Nutr Metab Care. 16:501–508. 2013.PubMed/NCBI

12 

Dasarathy S: Consilience in sarcopenia of cirrhosis. J Cachexia Sarcopenia Muscle. 3:225–237. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Rosenberg IH: Sarcopenia: Origins and clinical relevance. J Nutr. 127 5 Suppl:990S–991S. 1997.PubMed/NCBI

14 

Wang C and Bai L: Sarcopenia in the elderly: Basic and clinical issues. Geriatr Gerontol Int. 12:388–396. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Fukushima H, Nakanishi Y, Kataoka M, Tobisu K and Koga F: Prognostic significance of sarcopenia in patients with metastatic renal cell carcinoma. J Urol. 195:26–32. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Levolger S, van Vugt JL, de Bruin RW and IJzermans JN: Systematic review of sarcopenia in patients operated on for gastrointestinal and hepatopancreatobiliary malignancies. Br J Surg. 102:1448–1458. 2015. View Article : Google Scholar : PubMed/NCBI

17 

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 tumors of the respiratory and gastrointestinal tracts: A population based study. Lancet Oncol. 9:629–635. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Chindapasirt J: Sarcopenia in Cancer Patients. Asian Pac J Cancer Prev. 16:8075–8077. 2015. View Article : Google Scholar : PubMed/NCBI

19 

van Dijk DP, Bakens MJ, Coolsen MM, Rensen SS, van Dam RM, Bours MJ, Weijenberg MP, Dejong CH and Damink SW Olde: Low skeletal muscle radiation attenuation and visceral adiposity are associated with overall survival and surgical site infections in patients with pancreatic cancer. J Cachexia Sarcopenia Muscle. 8:317–326. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Carrara G, Pecorelli N, De Cobelli F, Cristel G, Damascelli A, Beretta L and Braga M: Preoperative sarcopenia determinants in pancreatic cancer patients. Clin Nutr. Oct 20–2016.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

21 

Onesti JK, Wright GP, Kenning SE, Tierney MT, Davis AT, Doherty MG and Chung MH: Sarcopenia and survival in patients undergoing pancreatic resection. Pancreatology. 16:284–289. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Amini N, Spolverato G, Gupta R, Margonis GA, Kim Y, Wagner D, Rezaee N, Weiss MJ, Wolfgang CL, Makary MM, et al: Impact total psoas volume on short- and long-term outcomes in patients undergoing curative resection for pancreatic adenocarcinoma: N new tool to assess sarcopenia. J Gastrointest Surg. 19:1593–1602. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Zalite I Ozola, Zykus R, Gonzalez M Francisco, Saygili F, Pukitis A, Gaujoux S, Charnley RM and Lyadov V: Influence of cachexia and sarcopenia on survival in pancreatic ductal adenocarcinoma: A systematic review. Pancreatology. 15:19–24. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Choi Y, Oh DY, Kim TY, Lee KH, Han SW, Im SA, Kim TY and Bang YJ: Skeletal muscle depletion predicts the prognosis of patients with advanced pancreatic cancer undergoing palliative chemotherapy, independent of body mass index. PLoS One. 10:e01397492015. View Article : Google Scholar : PubMed/NCBI

25 

Park I, Choi SJ, Kim YS, Ahn HK, Hong J, Sym SJ, Park J, Cho EK, Lee JH, Shin YJ and Shin DB: Prognostic factors for risk stratification of patients with recurrent or metastatic pancreatic adenocarcinoma who were treated with gemcitabine-based chemotherapy. Cancer Res Treat. 48:1264–1273. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Edge SB and Compton CC: The American Joint Committee on Cancer: The 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 17:1471–1474. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Trotti A, Colevas AD, Setser A, Rusch V, Jaques D, Budach V, Langer C, Murphy B, Cumberlin R, Coleman CN and Rubin P: CTCAE v3.0: Development of a comprehensive grading system for the adverse effects of cancer treatment. Semin Radiat Oncol. 13:176–181. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Nawaz H, Fan CY, Kloke J, Khalid A, McGrath K, Landsittel D and Papachristou GI: Performance characteristics of endoscopic ultrasound in the staging of pancreatic cancer: A meta-analysis. JOP. 14:484–497. 2013.PubMed/NCBI

29 

Zhao WY, Luo M, Sun YW, Xu Q, Chen W, Zhao G and Wu ZY: Computed tomography in diagnosing vascular invasion in pancreatic and periampullary cancers: A systematic review and meta-analysis. Hepatobiliary Pancreat Dis Int. 8:457–464. 2009.PubMed/NCBI

30 

Yang R, Lu M, Qian X, Chen J, Li L, Wang J and Zhang Y: Diagnostic accuracy of EUS and CT of vascular invasion in pancreatic cancer: A systematic review. J Cancer Res Clin Oncol. 140:2077–2086. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Puli SR, Singh S, Hagedorn CH, Reddy J and Olyaee M: Diagnostic accuracy of EUS for vascular invasion in pancreatic and periampullary cancers: A meta-analysis and systematic review. Gastrointest Endosc. 65:788–797. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Doi R, Imamura M, Hosotani R, Imaizumi T, Hatori T, Takasaki K, Funakoshi A, Wakasugi H, Asano T, Hishinuma S, et al: Surgery versus radiochemotherapy for resectable locally invasive pancreatic cancer: Final results of a randomized multi-institutional trial. Surg Today. 38:1021–1028. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Okumura S, Kaido T, Hamaguchi Y, Fujimoto Y, Masui T, Mizumoto M, Hammad A, Mori A, Takaori K and Uemoto S: Impact of preoperative quality as well as quantity of skeletal muscle on survival after resection of pancreatic cancer. Surgery. 157:1088–1098. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Hamaguchi Y, Kaido T, Okumura S, Kobayashi A, Hammad A, Tamai Y, Inagaki N and Uemoto S: Proposal for new diagnostic criteria for low skeletal muscle mass based on computed tomography imaging in Asian adults. Nutrition. 32:1200–1205. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Yamaguchi K, Okusaka T, Shimizu K, Furuse J, Ito Y, Hanada K and Shimosegawa T; Committee for revision of clinical guidelines for pancreatic cancer of Japan Pancreas Society, : EBM-based clinical guidelines for pancreatic cancer (2013) issued by the japan pancreas society: A synopsis. Jpn J Clin Oncol. 44:883–888. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Hewitt MJ, McPhail MJ, Possamai L, Dhar A, Vlavianos P and Monahan KJ: EUS-guided FNA for diagnosis of solid pancreatic neoplasms: A meta-analysis. Gastrointest Endosc. 75:319–331. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Ishii H, Furuse J, Boku N, Okusaka T, Ikeda M, Ohkawa S, Fukutomi A, Hamamoto Y, Nakamura K and Fukuda H; JCOG Gastrointestinal Oncology Study Group, : Phase II study of gemcitabine chemotherapy alone for locally advanced pancreatic carcinoma: JCOG0506. Jpn J Clin Oncol. 40:573–579. 2010. View Article : Google Scholar : PubMed/NCBI

38 

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). Eur J Cancer. 45:228–247. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET and Carbone PP: Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 5:649–655. 1982. View Article : Google Scholar : PubMed/NCBI

40 

Le N, Sund M and Vinci A; GEMS collaborating group of Pancreas, : 2000 Prognostic and predictive markers in pancreatic adenocarcinoma. Dig Liver Dis. 48:223–230. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Mahmoud FA and Rivera NI: The role of C-reactive protein as a prognostic indicator in advanced cancer. Curr Oncol Rep. 4:250–255. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Speck RM, Courneya KS, Mâsse LC, Duval S and Schmitz KH: An update of controlled physical activity trials in cancer survivors: A systematic review and meta-analysis. J Cancer Surviv. 4:87–100. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Neuzillet C, Vergnault M, Bonnetain F and Hammel P: Rationale and design of the Adapted Physical Activity in advanced Pancreatic Cancer patients (APACaP) GERCOR (Groupe Coopérateur Multidisciplinaire en Oncologie) trial: Study protocol for a randomized controlled trial. Trials. 16:4542015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 14 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ishii N, Iwata Y, Nishikawa H, Enomoto H, Aizawa N, Ishii A, Miyamoto Y, Yuri Y, Hasegawa K, Nakano C, Nakano C, et al: Effect of pretreatment psoas muscle mass on survival for patients with unresectable pancreatic cancer undergoing systemic chemotherapy. Oncol Lett 14: 6059-6065, 2017
APA
Ishii, N., Iwata, Y., Nishikawa, H., Enomoto, H., Aizawa, N., Ishii, A. ... Nishiguchi, S. (2017). Effect of pretreatment psoas muscle mass on survival for patients with unresectable pancreatic cancer undergoing systemic chemotherapy. Oncology Letters, 14, 6059-6065. https://doi.org/10.3892/ol.2017.6952
MLA
Ishii, N., Iwata, Y., Nishikawa, H., Enomoto, H., Aizawa, N., Ishii, A., Miyamoto, Y., Yuri, Y., Hasegawa, K., Nakano, C., Nishimura, T., Yoh, K., Sakai, Y., Ikeda, N., Takashima, T., Takata, R., Iijima, H., Nishiguchi, S."Effect of pretreatment psoas muscle mass on survival for patients with unresectable pancreatic cancer undergoing systemic chemotherapy". Oncology Letters 14.5 (2017): 6059-6065.
Chicago
Ishii, N., Iwata, Y., Nishikawa, H., Enomoto, H., Aizawa, N., Ishii, A., Miyamoto, Y., Yuri, Y., Hasegawa, K., Nakano, C., Nishimura, T., Yoh, K., Sakai, Y., Ikeda, N., Takashima, T., Takata, R., Iijima, H., Nishiguchi, S."Effect of pretreatment psoas muscle mass on survival for patients with unresectable pancreatic cancer undergoing systemic chemotherapy". Oncology Letters 14, no. 5 (2017): 6059-6065. https://doi.org/10.3892/ol.2017.6952