Minireviews Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Dec 15, 2022; 14(12): 2302-2312
Published online Dec 15, 2022. doi: 10.4251/wjgo.v14.i12.2302
Sarcopenia in pancreatic cancer: Effect on patient outcomes
Moon Hyung Choi, Department of Radiology, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, South Korea
Seung Bae Yoon, Division of Gastroenterology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 03312, South Korea
ORCID number: Moon Hyung Choi (0000-0001-5962-4772); Seung Bae Yoon (0000-0002-6119-7236).
Author contributions: Choi MH and Yoon SB contributed equally to the conception, design, and literature search; Choi MH drafted the manuscript and prepared the tables; Yoon SB modified and revised the manuscript.
Supported by the National Research Foundation of Korea, No. NRF-2021 R1F1A1062255.
Conflict-of-interest statement: All the authors report having no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Seung Bae Yoon, MD, PhD, Associate Professor, Division of Gastroenterology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, 1021, Tongil Ro, Eunpyeong-gu, Seoul 03312, South Korea. sbyoon@catholic.ac.kr
Received: September 17, 2022
Peer-review started: September 17, 2022
First decision: October 19, 2022
Revised: October 29, 2022
Accepted: November 28, 2022
Article in press: November 28, 2022
Published online: December 15, 2022
Processing time: 85 Days and 19.4 Hours

Abstract

Pancreatic cancer is a challenging disease with an increasing incidence and extremely poor prognosis. The clinical outcomes of pancreatic cancer depend on tumor biology, responses to treatments, and malnutrition or cachexia. Sarcopenia represents a severe catabolic condition defined by the age-related loss of muscle mass and strength and affects as much as 70% of malnourished pancreatic cancer patients. The lumbar skeletal muscle index, defined as the total abdominal muscle area at the L3 vertebral level adjusted by the square of the height, is widely used for assessing sarcopenia in patients with pancreatic cancer. Several studies have suggested that sarcopenia may be a risk factor for perioperative complications and decreased recurrence-free or overall survival in patients with pancreatic cancer undergoing surgery. Sarcopenia could also intensify chemotherapy-induced toxicities and worsen the quality of life and survival in the neoadjuvant or palliative chemotherapy setting. Sarcopenia, not only at the time of diagnosis but also during treatment, decreases survival in patients with pancreatic cancer. Theoretically, multimodal interventions may improve sarcopenia and clinical outcomes; however, no study has reported positive results. Further prospective studies are needed to confirm the prognostic role of sarcopenia and the effects of multimodal interventions in patients with pancreatic cancer.

Key Words: Sarcopenia; Pancreatic cancer; Skeletal muscle; Computed tomography; Outcomes; Survival

Core Tip: Despite advances in diagnosing and treating pancreatic cancer, the prognosis remains poor. More than half of patients with pancreatic cancer develop cachexia and sarcopenia, resulting in poor adherence to intensive treatments. Here, we introduced computed tomography-based body composition analysis, which has been used for analyzing sarcopenia in cancer patients, and covered controversial issues regarding the lack of consensus and diagnostic cutoff points. Recent studies analyzed the effect of sarcopenia on pancreatic cancer on surgery, neoadjuvant therapy, and palliative chemotherapy. Finally, we suggested recommendations for multimodal interventions for the management of sarcopenia and the design of future studies.



INTRODUCTION

Pancreatic cancer is the fourth leading cause of cancer-related deaths in both men and women worldwide[1]. Although overall cancer mortality continues to decrease in both sexes, the mortality rate of pancreatic cancer is still increasing[2]. Further, despite advances in cancer treatment, the 5-year survival rate remains poor at approximately 8%. Less than 20% of patients are in a resectable state and can be treated with curative surgery, and approximately 80% of patients have locally advanced or metastatic disease at the time of diagnosis. As such, efforts have been recently made to improve pancreatic cancer treatment, including advanced surgical techniques, adjuvant chemotherapy, neoadjuvant therapy (NAT), and combination chemotherapy regimens [e.g., folinic acid, fluorouracil, irinotecan hydrochloride, and oxaliplatin (FOLFIRINOX), and gemcitabine plus nab-paclitaxel][3,4].

The clinical outcomes of pancreatic cancer not only depend on tumor biology and treatment responses but are also strongly influenced by the nutrition and performance status of the patients. Before or during treatment, many patients experience early alteration of the metabolic state with rapid weight loss or treatment-related performance deterioration. Therefore, the assessment of nutritional status and performance status is crucial to determine the best treatment modality for extending survival with adequate quality of life.

The assessment of body composition typically refers to the measurement of fat and muscle mass. Sarcopenia is a term used to describe the age-related loss of muscle mass and strength. Beyond the quantification of the muscle mass, the importance of the muscle quality assessed for fat infiltration within the muscle is also emerging. A number of parameters have been analyzed for sarcopenic obesity, such as subcutaneous adipose tissue, visceral adipose tissue, and visceral fat-to-skeletal muscle ratio. Sarcopenia has been proven to be related to the prognosis of various diseases, especially in several types of cancer. A wide range of techniques such as body imaging modalities, including computed tomography (CT) and magnetic resonance imaging, bioimpedance analysis, or anthropometric measures, have been used to assess muscle mass; however, no gold standard diagnostic method for sarcopenia has been established yet[5]. Despite its high cost and radiation exposure, CT is the most accessible way to measure the fat and muscle area separately because of the regular follow-up CT examinations for cancer patients[6].

This study aimed to describe a method to assess body composition using CT images and the role of sarcopenia in the management and prognosis of pancreatic cancer.

CT-BASED BODY COMPOSITION ANALYSIS

Various methods have been introduced for CT-based body composition analysis. The total abdominal muscle area, including the entire abdominal wall and back muscle, is commonly measured on CT images. Muscle area can be measured on one axial slice, or muscle volume can be measured on several consecutive slices. Among the many different landmarks, the level of the transverse processes of the L3 vertebra is generally used. Measurement of the psoas muscle area is a simple method, and the psoas muscle area has been proven to be highly correlated with the total abdominal muscle area.

Thresholds of CT attenuation can affect the muscle area, as they determine the pixels that contain muscles and other tissues. If the threshold range is wider, more pixels are selected as the muscle area, leading to a larger muscle area. The use of intravenous contrast or slice thickness can affect body composition data[7]. The phase of CT acquisition (e.g., arterial or portal) also affects the assessment of the skeletal muscle area because the contrast agent increases tissue attenuation. Therefore, the consistent use of certain thresholds and a particular phase of CT is important to obtain reliable results. In addition, CT acquisition parameters should be reported together with body composition data using CT. As body habitus affects muscle mass, several methods are used to adjust the body habitus using the square of height and body weight. The most commonly used index is the skeletal muscle index, which is calculated as muscle area/height squared (cm2/m2). In pancreatic cancer, the lumbar skeletal muscle index (cm2/m2), defined as the total abdominal muscle area at the L3 vertebral level adjusted by the height square, is commonly used. Additionally, the mean density of the muscle reflecting the amount of intervening fat in the muscle may be related to muscle quality.

PANCREATIC CANCER AND SARCOPENIA

There is a lack of consensus regarding the definition of sarcopenia in patients with pancreatic cancer. Among the many definitions of sarcopenia, the cutoff values for sex-specific lumbar skeletal muscle index suggested by Prado et al[8] (52.4 cm2/m2 for males and 38.5 cm2/m2 for females) have been widely used in early Western studies[9-11]. These sex-specific cutoffs were obtained from the most significant P value by optimal stratification of mortality in obese cancer patients. In addition, Martin et al[12] reported a new sex- and body mass index-specific threshold value of sarcopenia applicable to both obese and non-obese cancer patients as follows: < 43 cm2/m2 for males with body mass index < 25 kg/m2 or < 53 cm2/m2 for males with body mass index > 25 kg/m2 and < 41 cm2/m2 for females. This definition of sarcopenia has also been widely used in studies on pancreatic cancer[13-17]. However, if the cutoff values based on Western studies are applied to Eastern cancer patients, the prevalence of sarcopenia is increased, with more than two-thirds of males classified as having sarcopenia, and a maldistribution between sexes occurs[18,19]. Therefore, many Eastern studies on pancreatic cancer have applied the following criteria based on a consensus report of the Asian Working Group for Sarcopenia[20]: 42 cm2/m2 for males and 38 cm2/m2 for females[21-23]. Because body composition can vary among ethnicities and tumor stages, a few studies have set their own cutoff values based on the lowest sex-specific tertile or quartile of the individual cohorts[18,24,25].

Pretreatment sarcopenia is present in 40%-73% of patients with pancreatic cancer. The incidence of sarcopenia and cancer cachexia is particularly higher in pancreatic cancer than in other malignancies[26], possibly owing to the high activation of host inflammatory response and its catabolic pathways in patients with pancreatic cancer. Pancreatic exocrine insufficiency also contributes to malnutrition and weight loss. Pancreatic enzymes are essential for the degradation and absorption of fat and liposoluble vitamins; thus, deficiency of pancreatic enzymes results in steatorrhea and severe maldigestion[27]. Finally, patients with pancreatic cancer can exhibit endocrine insufficiency, usually resulting in pancreatogenic diabetes.

SURGICAL TREATMENT

Surgical resection is the only curative treatment option for localized pancreatic cancer. However, pancreatic cancer surgery carries a high risk of perioperative morbidity and recurrence. Therefore, the role of sarcopenia in patients undergoing surgery is a major topic of interest in the field of pancreatic cancer. The main studies that analyzed the effect of sarcopenia on the surgical treatment of pancreatic cancer are summarized in Table 1[7,9,10,18,19,24,25,28-30]. In 2012, Peng et al[24] evaluated 557 patients with pancreatic cancer who underwent curative resection at Johns Hopkins University. Sarcopenia stratified by total psoas muscle area increased the 3-year mortality by 63%. A few years later, a study by Amini et al[7] showed that assessing the psoas muscle volume might be a better method than assessing the psoas muscle area to define sarcopenia. Most subsequent studies have evaluated the total abdominal muscle area instead of the psoas muscle area or volume.

Table 1 Studies analyzing the effect of sarcopenia on surgical outcomes of pancreatic cancer.
Ref.CountryNo. of patientsImaging modalityLevelTimeDefinition and cutoffSarcopenia prevalence before surgeryTypes of surgeryPerioperative complicationsSurvivalAdditional meaningful findings or comments
Peng et al[24], 2012United States557CTL3Before surgeryTPAI (mm2/m2), lowest quartile: < 564.2 (M), < 414.5 (F)25%PD, DP(-)(+) OSSarcopenia was an independent predictor of survival in multivariable analysis
Amini et al[7], 2015United States763CTL3Before surgeryTPAI (mm2/m2), < 564.2 (M), 414.5 (F); TPVI (cm³/m2), < 17.2 (M), < 12.0 (F)25% by TPAI, 20% by TPVIPD, DP. TP(+) Overall Cx. by TPVI(+) OS by TPVITPVI was a better measure for defining sarcopenia rather than TPAI
Pecorelli et al[9], 2016Italy202CTL3Before surgeryLSMI, < 52.4 cm2/m2 (M), < 38.5 cm2/m2 (F)165%PD(-)NEThe combination of visceral obesity and sarcopenia was a predictor of perioperative Cx
Ninomiya et al[28], 2017Japan265CTL3Before surgeryLSMI, < 43.75 cm2/m2 (M), < 38.5 cm2/m2 (F)64%PD, DP. TP(-)(-)Sarcopenia was an independent prognostic factor only in patients with BMI ≥ 22 kg/m2
Okumura et al[29], 2017 Japan301CTL3Before surgeryLSMI, clinically relevant cutoff: < 47.1 cm2/m2 (M), < 36.6 cm2/m2 (F)40%PD, DP. TP(-)(+) OS and RFSLow muscle attenuation, as well as low muscle mass, was associated with worse OS and RFS
Choi et al[18], 2018 South Korea180CTL3Before and after 60 d of surgeryLSMI, the lowest tertile; < 45.3 cm2/m2 (M), < 39.3 cm2/m2 (F)33%PD, DP(-)(+) OSAccelerated muscle loss after surgery negatively impacts OS
Sugimoto et al[19], 2018 United States323CTL3Before surgeryLSMI, < 55.4 cm2/m2 (M), < 38.9 cm2/m2 (F)62%PD, DP. TPNE(-)Smaller sex-standardized LSMI as a continuous variable is associated with a shorter OS
Gruber et al[10], 2019 Austria133CTL3Before surgeryLSMI, < 52.4 cm2/m2 (M), < 38.5 cm2/m2 (F)159%PD, DP(-)(+) OSObese patients (BMI ≥ 25) with sarcopenia have higher incidence of major post-operative Cx
Ryu et al[30], 2020 South Korea548CTL3Before surgeryLSMI, < 50.18 cm2/m2 (M), < 38.63 cm2/m2 (F)46%PD(-)(+) OS Sarcopenic obesity is a predictive factor for post-operative pancreatic fistula after PD
Rom et al[25], 2022 Israel111CTL3Before surgeryLSMI, the lowest quartile: < 44; 35 cm2/m2 (M), < 34.82 cm2/m2 (F)25%PD, DP(+) Overall Cx.(+) OS, DSS, and RFSHigh intramuscular adipose tissue content correlates with poor OS and DSS

The effect of sarcopenia in the surgical setting has been well-summarized in a recent meta-analysis[31]. Bundred et al[31] analyzed 43 studies assessing body composition in patients with pancreatic cancer before surgery, of which 30 studies assessed body composition using CT. Among these, 10 studies reported the impact of preoperative sarcopenia on postoperative outcomes. Sarcopenia was associated with perioperative mortality (odds ratio: 2.40; 95% confidence interval: 1.19-4.85) and overall survival (hazard ratio: 1.95; 95% confidence interval, 1.54-2.05) but not with overall complications (odds ratio: 0.96; 95% confidence interval, 0.78-1.19). This meta-analysis was limited by the heterogeneity in the methods and cutoff values for assessing sarcopenia in individual studies.

Patients with overweight or obesity and sarcopenia exhibit worse clinical outcomes than those with sarcopenia alone. In many studies, the combination of obesity and sarcopenia was associated with a higher incidence of perioperative complications and lower survival[9,10,28,30]. Sarcopenic obesity is a complex syndrome associated with aging and lifestyle changes. Reduced physical activity may result in accelerated muscle loss, decreased energy consumption, and adverse health effects such as hypertension, dyslipidemia, and insulin resistance. Sarcopenia and obesity should be comprehensively considered to stratify patients undergoing pancreatic cancer surgery into risk categories for predicting clinical outcomes.

The amount of skeletal muscle mass has been traditionally used as a criterion to determine sarcopenia. However, some studies reported that a decrease in muscle quality, represented by low skeletal muscle attenuation also negatively impacts prognosis after pancreatic cancer surgery[25,29]. Although the muscle mass remains normal, muscle strength and function may be reduced. In such cases, the deposition of intramuscular adipose tissue causes reduced muscle density, resulting in a decline in muscle quality. A previous study reported that skeletal muscle density decreased before the reduction in skeletal muscle mass in patients with cancer[32]. Thus, efforts should be made to evaluate and monitor muscle quantity and quality closely.

Choi et al[18] demonstrated that preoperative sarcopenia and post-operative accelerated muscle loss were associated with poor overall survival in pancreatic cancer patients undergoing surgery. Postoperative skeletal muscle changes were assessed based on the difference between the initial and follow-up CT scans at an approximately 60-d interval. Approximately 30% of their patients showed significant muscle loss of more than 10% over 60 d. Given that most patients undergoing pancreatic cancer surgery receive adjuvant chemotherapy, it may be necessary to maintain muscle mass through active nutritional support and rehabilitation exercise after surgery.

NAT

In recent years, NAT, including neoadjuvant chemotherapy and chemoradiation, has become the standard of care for borderline resectable or locally advanced pancreatic cancers. NAT may increase the rate of margin-negative resections and help clinicians screen patients with progressive disease during NAT who might not benefit from surgery[33]. In addition, NAT may be able to treat micrometastases at the time of diagnosis, which can reduce early lymph node or hepatic recurrence after surgery[34]. However, because not all patients receiving NAT are eligible for curative surgery and have increased survival, it is imperative to develop biomarkers that can predict responses to NAT. Recent studies that assessed the correlation of body composition with the response to and outcome of NAT in patients with pancreatic cancer are summarized in Table 2[13-15,35-37].

Table 2 Studies analyzing the effect of sarcopenia on neoadjuvant therapy outcomes of pancreatic cancer.
Ref.
Country
No. of patients
Inclusion
Imaging modality
Level
Time
Definition and cutoff
Sarcopenia prevalence before NAT
Resectability
Survival
Additional meaningful findings or comments
Cooper et al[35], 2015 United States89RPCCTL3Before and after NATLSMI, < 55.4 cm2/m2 (M), < 38.9 cm2/m2 (F)55%(-)(-)SKM loss during NAT was correlated with DFS
Cloyd et al[36], 2018 United States127RPC, BRPC, LAPCCTL3Before and after NAT, 3 mo and 12 mo after surgery (PD)LSMI, < 55.4 cm2/m2 (M), < 38.9 cm2/m2 (F)63%NE(-)SKM gain between the postoperative period and 1-yr follow-up was correlated with improved OS
Sandini et al[13], 2018 United States and Italy193BRPC, LAPCCTL3Before and after NATLSMI, < 43 cm2/m2 (M) where BMI < 25 kg/m2, < 53 cm2/m2 (M) where BMI > 25 kg/m2, < 41 cm2/m2 (F)144%(-)NESKM gain during NAT is correlated with better resectability
Griffin et al[14], 2019 Ireland78BRPCCTL3Before and after NATLSMI, < 43 cm2/m2 (M) where BMI < 25 kg/m2, < 53 cm2/m2 (M) where BMI > 25 kg/m2, < 41 cm2/m2 (F)150%(-)(-)Low muscle attenuation before NAT and SKM loss during NAT was correlated with decreased OS
Takeda et al[15], 2021 Japan62RPCCTL3Before NATLSMI, < 43 cm2/m2 (M) where BMI < 25 kg/m2, < 53 cm2/m2 (M) where BMI > 25 kg/m2, < 41 cm2/m2 (F)140%(-)NESarcopenia before NAT did not correlate with antitumor response and toxicity of therapy
Jin et al[37], 2022 China119RPCCTL3Before and after NATLSMI, < 41 cm2/m2 (M), < 38.5 cm2/m2 (F) 48%NE(+) OS, DFSSKM and fat wasting during NAT was correlated with decreased OS and DFS

The prevalence of sarcopenia before NAT ranges from 40% to 63%. However, no studies have shown that sarcopenia at the time of diagnosis affects resectability after NAT. Meanwhile, in a recent study by Jin et al[37] in 2022, sarcopenia before NAT was associated with decreased overall survival and disease-free survival. Among 119 patients, 57 (47.9%) had sarcopenia before NAT. The median overall survival and disease-free survival for sarcopenia patients were 16.6 mo and 10.9 mo, respectively, which were significantly lower than those for non-sarcopenia patients (21.4 mo and 14.0 mo, respectively; all P < 0.001). However, because of the retrospective nature of this study, unavoidable biases were associated with variations in the NAT regimens and treatment durations.

Several studies have evaluated changes in body composition during NAT and their effect on clinical outcomes[13,14,35-37]. In these studies, most patients experienced further depletion of skeletal muscle during NAT and the degree of skeletal muscle loss correlated with resectability or survival. Sandini et al[13] reported that patients who underwent resection after NAT had skeletal muscle gain, whereas unresectable patients experienced muscle wasting during NAT. Therefore, skeletal muscle changes must be considered in the setting of NAT, and further efforts should focus on maintaining muscle mass during treatment.

PALLIATIVE CHEMOTHERAPY

Approximately 80% of pancreatic cancer patients are diagnosed at an advanced stage, including locally advanced or metastatic disease. Combination chemotherapy with FOLFIRINOX or gemcitabine plus nab-paclitaxel is associated with more prolonged overall survival than gemcitabine monotherapy, with acceptable adverse events[3,4]. Currently, these two combination regimens are considered the standard first-line treatments for advanced pancreatic cancer. Therefore, selecting appropriate patients who can tolerate aggressive palliative chemotherapy is crucial. In palliative chemotherapy settings, the occurrence of sarcopenia can be related to exacerbated chemotherapy toxicity, reduced adherence to treatment, or worsened survival.

Several recent studies evaluated the effect of sarcopenia on various clinical outcomes in patients with advanced pancreatic cancer receiving palliative chemotherapy (Table 3)[11,16,17,21-23,38-40]. Kim et al[17] investigated the clinical impact of sarcopenia in 330 patients with metastatic pancreatic cancer who were treated with first-line gemcitabine-based chemotherapy. All grade ≥ 3 toxicities developed at a significantly higher frequency in sarcopenia patients than in non-sarcopenia patients. This result might be explained by the link between body composition and the pharmacokinetics of chemotherapy drugs. In addition, a recent study by Emori et al[23] in 2022 showed that major adverse events, including hematologic toxicity, occurred more frequently in sarcopenia patients. Remarkably, the grade ≥ 3 neutropenia rate was significantly higher in sarcopenia patients than in non-sarcopenia patients (64% vs 40%, P = 0.028). Therefore, patients with sarcopenia should be considered for dose modification or aggressive preventive interventions to reduce chemotherapy-related toxicity.

Table 3 Studies analyzing the effect of sarcopenia on palliative chemotherapy outcomes of pancreatic cancer.
Ref.
Country
No. of patients
Inclusion (%)
Imaging modality
Level
Time
Definition and cutoff
Sarcopenia prevalence before CTX
CTX regimen
CTX toxicity
PFS
OS
Additional meaningful findings or comments
Kays et al[11], 2018 United States53LAPC (49), MPC (51)CTL3Before and during CTX (median 5.6 times)LSMI, < 52.4 cm2/m2 (M), < 38.5 cm2/m2 (F)149%1st line FOLFIRINOXNENE(-)No muscle wasting during CTX improved OS
Basile et al[16], 2019 Italy94LAPC (50), MPC (50)CTL3Before and after 12 wk of CTXLSMI, < 43 cm2/m2 (M) where BMI < 25 kg/m2, < 53 cm2/m2 (M) where BMI > 25 kg/m2, < 41 cm2/m2 (F)273%VariousNE(-)(-)Loss of skeletal muscle mass (≥ 10%) was associated with worse OS and PFS
Kurita et al[38], 2019 Japan82LAPC (35), MPC (65)CTL3Before CTXLSMI, clinically relevant cut-off: < 45.3 cm2/m2 (M), < 37.1 cm2/m2 (F)51%1st line FOLFIRINOX(-)(+)(+)Sarcopenic obesity was associated with hematologic toxicity
Lee et al[39], 2019South Korea57LAPC (5), MPC (95)CTL3Before and after 8 wk of CTXLSMI, median level: unknown50%2nd line FOLFIRINOXNE(+)(+)Baseline LSMI was an independent predictor of survival in multivariable analysis
Kim et al[17], 2021 South Korea251MPC (100)CTL3Before and after 8 wk of CTXLSMI, < 43 cm2/m2 (M) where BMI < 25 kg/m2, < 53 cm2/m2 (M) where BMI > 25 kg/m2, < 41 cm2/m2 (F)241%1st line gemcitabine-based CTX(+) Overall grade ≥ 3 toxicity(-)(-)Sarcopenia was a prognostic factor for OS but not for PFS in multivariable analysis
Uemura et al[21], 2021 Japan69LAPC (29), MPC (71)CTL3Before and after 8 wk of CTXLSMI, < 42 cm2/m2 (M), < 38 cm2/m2 (F)3 48%1st line FOLFIRINOX(-)(-)(-)Loss of skeletal muscle mass (≥ 7.9%) is associated with worse OS
Williet et al[40], 2021 France79MPC (100)CTL3Before CTXTPAI, clinically relevant cutoff: 5.73 cm2/m2 (M), 4.37 cm2/m2 (F)38%Various(-)(+)(+)Measuring TPAI was less time-consuming than measuring LSMI
Asama et al[22], 2022 Japan124LAPC (29), MPC (60), RePC (15)CTL3Before CTXLSMI, < 42 cm2/m2 (M), < 38 cm2/m2 (F)349%1st line Gem-Nab(-)(-)(-)In elderly patients (> 70 yr), sarcopenia was associated with worse OS
Emori et al[23], 2022 Japan176LAPC (14), MPC (86)CTL3Before CTXLSMI, < 42 cm2/m2 (M), < 38 cm2/m2 (F)353%1st line Gem-Nab(+) Overall grade ≥ 3 toxicity(+)(+)Propensity score matching analysis was performed

A study by Kurita et al[38] conducted on 82 pancreatic cancer patients treated with FOLFIRINOX showed that compared with non-sarcopenia patients, sarcopenia patients had a significantly lower median overall survival (11.3 mo vs 17.0 mo) and progression-free survival (3.0 mo vs 6.1 mo). In another study that evaluated 84 patients treated with gemcitabine plus nab-paclitaxel, the median overall and progression-free survival were also lower in sarcopenia patients than in non-sarcopenia patients (10.3 mo vs 18.1 mo and 5.0 mo vs 8.0 mo, respectively)[23]. Skeletal muscle mass can also be used as a critical prognostic factor in patients receiving second-line FOLFIFIRNOX chemotherapy for advanced pancreatic cancer[39]. In addition, body composition-based patient selection and dose determination may be clinically useful for patients receiving palliative chemotherapy to minimize toxicity and maximize therapeutic benefits.

Some studies have reported the negative impact of accelerated muscle loss during palliative chemotherapy on the clinical outcomes of advanced pancreatic cancer[16,21]. Basile et al[16] reported that early loss of skeletal muscle by more than 10% during the first 3 mo of chemotherapy was significantly associated with poor overall and progression-free survival. In a study by Uemura et al[21], patients with a greater decrease in skeletal muscle index (≥ 7.9%) 2 mo after the start of FOLFIRINOX therapy had a shorter survival (10.9 mo) than those who did not (21.0 mo, P < 0.01). The management of sarcopenia, not only at the time of diagnosis but also during palliative chemotherapy, is important in patients with advanced pancreatic cancer.

LIMITATIONS

There has been heterogeneity among studies regarding the threshold for sarcopenia based on low skeletal muscle index. The races of study participants, clinical stages, and treatment methods could affect skeletal muscle index. Therefore, caution is needed when synthesizing or comparing each study. Another limitation of the studies based on CT-assessed sarcopenia relates to the failure to include any functional measurement or patient-reported quality of life. Although the decrease and change of skeletal muscle mass is a major concern for supportive care in pancreatic cancer patients, physical functional assessments and quality of life measures have been highlighted as meaningful outcomes for cancer cachexia research.

FUTURE DIRECTIONS

Since sarcopenia adversely affects the outcomes of patients with pancreatic cancer in surgical or chemotherapy settings, interventions to improve sarcopenia may help increase survival rates. However, studies investigating the impact of nutritional or exercise interventions on survival are immature, and the results are still far from demonstrating their clinical efficacy. A phase II trial on inoperable pancreatic or lung cancer patients reported that multimodal intervention, including polyunsaturated fatty acid nutritional supplements, exercise, and anti-inflammatory medication is feasible and safe[41]. In the IMPACT study by Basile et al[16], more than half of the patients undergoing FOLFIRINOX chemotherapy were evaluated by a nutritionist and received dietary supplementation. Body weight loss during chemotherapy was the only factor associated with early dietary supplementation; however, nutritional support or intervention did not affect prognosis with respect to overall survival. A “Nutritional Oncology Board” has recently emerged as a good clinical practice tool of routine care for cancer patients[26]. Based on the adoption of this system, early nutritional assessment before or during oncological treatment can provide patient-tailored management for preventing or treating sarcopenia.

Although there has been increasing interest in the assessment of sarcopenia using CT-based methods, there are some areas to be improved in future studies[42]. It is recommended to use validated techniques and appropriate diagnostic criteria based on the study populations[43]. For sequential measurements, CT protocols should be controlled, including the timing of image acquisition and amount of contrast agent. It is also recommended to measure various physical performance measures (e.g., gait speed or handgrip strength) as indicators of muscle quality along with skeletal muscle mass, which reflects muscle quantity. Through the application of artificial intelligence, CT-based body composition analysis, which is a time-consuming process, can be applied to routine clinical practice[44].

CONCLUSION

Sarcopenia has been recognized as a prognostic biomarker in patients with pancreatic cancer receiving surgical or chemotherapy treatments. The CT-based analysis is an objective and useful tool to assess sarcopenia and skeletal muscle changes during treatment. It may be helpful to consider sarcopenia when predicting patient outcomes and to minimize complications. However, whether early nutritional support or exercise improves sarcopenia and clinical outcomes remains unclear. Further prospective studies are necessary to confirm the prognostic role of sarcopenia and the effects of multimodal interventions in patients with pancreatic cancer.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Corresponding Author’s Membership in Professional Societies: The Korean Society of Gastroenterology, No. 1-16-2632.

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: South Korea

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C, C

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Marquardt JP, United States; Pan Y, China; Shariati MBH, Iran S-Editor: Gong ZM L-Editor: Filipodia P-Editor: Gong ZM

References
1.  Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4235]  [Cited by in F6Publishing: 9179]  [Article Influence: 4589.5]  [Reference Citation Analysis (2)]
2.  Carioli G, Malvezzi M, Bertuccio P, Boffetta P, Levi F, La Vecchia C, Negri E. European cancer mortality predictions for the year 2021 with focus on pancreatic and female lung cancer. Ann Oncol. 2021;32:478-487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 79]  [Article Influence: 26.3]  [Reference Citation Analysis (0)]
3.  Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C, Bennouna J, Bachet JB, Khemissa-Akouz F, Péré-Vergé D, Delbaldo C, Assenat E, Chauffert B, Michel P, Montoto-Grillot C, Ducreux M; Groupe Tumeurs Digestives of Unicancer;  PRODIGE Intergroup. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817-1825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4838]  [Cited by in F6Publishing: 5248]  [Article Influence: 403.7]  [Reference Citation Analysis (1)]
4.  Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, Harris M, Reni M, Dowden S, Laheru D, Bahary N, Ramanathan RK, Tabernero J, Hidalgo M, Goldstein D, Van Cutsem E, Wei X, Iglesias J, Renschler MF. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691-1703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4035]  [Cited by in F6Publishing: 4472]  [Article Influence: 406.5]  [Reference Citation Analysis (0)]
5.  Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, Martin FC, Michel JP, Rolland Y, Schneider SM, Topinková E, Vandewoude M, Zamboni M; European Working Group on Sarcopenia in Older People. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing. 2010;39:412-423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6987]  [Cited by in F6Publishing: 7820]  [Article Influence: 558.6]  [Reference Citation Analysis (0)]
6.  Lee K, Shin Y, Huh J, Sung YS, Lee IS, Yoon KH, Kim KW. Recent Issues on Body Composition Imaging for Sarcopenia Evaluation. Korean J Radiol. 2019;20:205-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 184]  [Article Influence: 36.8]  [Reference Citation Analysis (0)]
7.  Amini N, Spolverato G, Gupta R, Margonis GA, Kim Y, Wagner D, Rezaee N, Weiss MJ, Wolfgang CL, Makary MM, Kamel IR, Pawlik TM. Impact Total Psoas Volume on Short- and Long-Term Outcomes in Patients Undergoing Curative Resection for Pancreatic Adenocarcinoma: a New Tool to Assess Sarcopenia. J Gastrointest Surg. 2015;19:1593-1602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 182]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
8.  Prado CM, Lieffers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L, 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. 2008;9:629-635.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1822]  [Cited by in F6Publishing: 2171]  [Article Influence: 135.7]  [Reference Citation Analysis (0)]
9.  Pecorelli N, Carrara G, De Cobelli F, Cristel G, Damascelli A, Balzano G, Beretta L, Braga M. Effect of sarcopenia and visceral obesity on mortality and pancreatic fistula following pancreatic cancer surgery. Br J Surg. 2016;103:434-442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 175]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
10.  Gruber ES, Jomrich G, Tamandl D, Gnant M, Schindl M, Sahora K. Sarcopenia and sarcopenic obesity are independent adverse prognostic factors in resectable pancreatic ductal adenocarcinoma. PLoS One. 2019;14:e0215915.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 51]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
11.  Kays JK, Shahda S, Stanley M, Bell TM, O'Neill BH, Kohli MD, Couch ME, Koniaris LG, Zimmers TA. Three cachexia phenotypes and the impact of fat-only loss on survival in FOLFIRINOX therapy for pancreatic cancer. J Cachexia Sarcopenia Muscle. 2018;9:673-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 94]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
12.  Martin L, Birdsell L, Macdonald N, Reiman T, Clandinin MT, McCargar LJ, Murphy R, Ghosh S, Sawyer MB, Baracos VE. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol. 2013;31:1539-1547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1436]  [Cited by in F6Publishing: 1512]  [Article Influence: 137.5]  [Reference Citation Analysis (0)]
13.  Sandini M, Patino M, Ferrone CR, Alvarez-Pérez CA, Honselmann KC, Paiella S, Catania M, Riva L, Tedesco G, Casolino R, Auriemma A, Salandini MC, Carrara G, Cristel G, Damascelli A, Ippolito D, D'Onofrio M, Lillemoe KD, Bassi C, Braga M, Gianotti L, Sahani D, Fernández-Del Castillo C. Association Between Changes in Body Composition and Neoadjuvant Treatment for Pancreatic Cancer. JAMA Surg. 2018;153:809-815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 92]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
14.  Griffin OM, Duggan SN, Ryan R, McDermott R, Geoghegan J, Conlon KC. Characterising the impact of body composition change during neoadjuvant chemotherapy for pancreatic cancer. Pancreatology. 2019;19:850-857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 44]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
15.  Takeda T, Sasaki T, Mie T, Furukawa T, Yamada Y, Kasuga A, Matsuyama M, Ozaka M, Sasahira N. The impact of body composition on short-term outcomes of neoadjuvant chemotherapy with gemcitabine plus S-1 in patients with resectable pancreatic cancer. Jpn J Clin Oncol. 2021;51:604-611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
16.  Basile D, Parnofiello A, Vitale MG, Cortiula F, Gerratana L, Fanotto V, Lisanti C, Pelizzari G, Ongaro E, Bartoletti M, Garattini SK, Andreotti VJ, Bacco A, Iacono D, Bonotto M, Casagrande M, Ermacora P, Puglisi F, Pella N, Fasola G, Aprile G, Cardellino GG. The IMPACT study: early loss of skeletal muscle mass in advanced pancreatic cancer patients. J Cachexia Sarcopenia Muscle. 2019;10:368-377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 52]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
17.  Kim IH, Choi MH, Lee IS, Hong TH, Lee MA. Clinical significance of skeletal muscle density and sarcopenia in patients with pancreatic cancer undergoing first-line chemotherapy: a retrospective observational study. BMC Cancer. 2021;21:77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 30]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
18.  Choi MH, Yoon SB, Lee K, Song M, Lee IS, Lee MA, Hong TH, Choi MG. Preoperative sarcopenia and post-operative accelerated muscle loss negatively impact survival after resection of pancreatic cancer. J Cachexia Sarcopenia Muscle. 2018;9:326-334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 95]  [Article Influence: 15.8]  [Reference Citation Analysis (1)]
19.  Sugimoto M, Farnell MB, Nagorney DM, Kendrick ML, Truty MJ, Smoot RL, Chari ST, Moynagh MR, Petersen GM, Carter RE, Takahashi N. Decreased Skeletal Muscle Volume Is a Predictive Factor for Poorer Survival in Patients Undergoing Surgical Resection for Pancreatic Ductal Adenocarcinoma. J Gastrointest Surg. 2018;22:831-839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 38]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
20.  Chen LK, Liu LK, Woo J, Assantachai P, Auyeung TW, Bahyah KS, Chou MY, Chen LY, Hsu PS, Krairit O, Lee JS, Lee WJ, Lee Y, Liang CK, Limpawattana P, Lin CS, Peng LN, Satake S, Suzuki T, Won CW, Wu CH, Wu SN, Zhang T, Zeng P, Akishita M, Arai H. Sarcopenia in Asia: consensus report of the Asian Working Group for Sarcopenia. J Am Med Dir Assoc. 2014;15:95-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2330]  [Cited by in F6Publishing: 2708]  [Article Influence: 270.8]  [Reference Citation Analysis (0)]
21.  Uemura S, Iwashita T, Ichikawa H, Iwasa Y, Mita N, Shiraki M, Shimizu M. The impact of sarcopenia and decrease in skeletal muscle mass in patients with advanced pancreatic cancer during FOLFIRINOX therapy. Br J Nutr. 2021;125:1140-1147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
22.  Asama H, Ueno M, Kobayashi S, Fukushima T, Kawano K, Sano Y, Tanaka S, Nagashima S, Morimoto M, Ohira H, Maeda S. Sarcopenia: Prognostic Value for Unresectable Pancreatic Ductal Adenocarcinoma Patients Treated With Gemcitabine Plus Nab-Paclitaxel. Pancreas. 2022;51:148-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
23.  Emori T, Itonaga M, Ashida R, Tamura T, Kawaji Y, Hatamaru K, Yamashita Y, Shimokawa T, Koike M, Sonomura T, Kawai M, Kitano M. Impact of sarcopenia on prediction of progression-free survival and overall survival of patients with pancreatic ductal adenocarcinoma receiving first-line gemcitabine and nab-paclitaxel chemotherapy. Pancreatology. 2022;22:277-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
24.  Peng P, Hyder O, Firoozmand A, Kneuertz P, Schulick RD, Huang D, Makary M, Hirose K, Edil B, Choti MA, Herman J, Cameron JL, Wolfgang CL, Pawlik TM. Impact of sarcopenia on outcomes following resection of pancreatic adenocarcinoma. J Gastrointest Surg. 2012;16:1478-1486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 377]  [Cited by in F6Publishing: 409]  [Article Influence: 34.1]  [Reference Citation Analysis (0)]
25.  Rom H, Tamir S, Van Vugt JLA, Berger Y, Perl G, Morgenstern S, Tovar A, Brenner B, Benchimol D, Kashtan H, Sadot E. Sarcopenia as a Predictor of Survival in Patients with Pancreatic Adenocarcinoma After Pancreatectomy. Ann Surg Oncol. 2022;29:1553-1563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
26.  Rovesti G, Valoriani F, Rimini M, Bardasi C, Ballarin R, Di Benedetto F, Menozzi R, Dominici M, Spallanzani A. Clinical Implications of Malnutrition in the Management of Patients with Pancreatic Cancer: Introducing the Concept of the Nutritional Oncology Board. Nutrients. 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
27.  Vujasinovic M, Valente R, Del Chiaro M, Permert J, Löhr JM. Pancreatic Exocrine Insufficiency in Pancreatic Cancer. Nutrients. 2017;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 78]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
28.  Ninomiya G, Fujii T, Yamada S, Yabusaki N, Suzuki K, Iwata N, Kanda M, Hayashi M, Tanaka C, Nakayama G, Sugimoto H, Koike M, Fujiwara M, Kodera Y. Clinical impact of sarcopenia on prognosis in pancreatic ductal adenocarcinoma: A retrospective cohort study. Int J Surg. 2017;39:45-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 71]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
29.  Okumura S, Kaido T, Hamaguchi Y, Kobayashi A, Shirai H, Yao S, Yagi S, Kamo N, Hatano E, Okajima H, Takaori K, Uemoto S. Visceral Adiposity and Sarcopenic Visceral Obesity are Associated with Poor Prognosis After Resection of Pancreatic Cancer. Ann Surg Oncol. 2017;24:3732-3740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 101]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
30.  Ryu Y, Shin SH, Kim JH, Jeong WK, Park DJ, Kim N, Heo JS, Choi DW, Han IW. The effects of sarcopenia and sarcopenic obesity after pancreaticoduodenectomy in patients with pancreatic head cancer. HPB (Oxford). 2020;22:1782-1792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
31.  Bundred J, Kamarajah SK, Roberts KJ. Body composition assessment and sarcopenia in patients with pancreatic cancer: a systematic review and meta-analysis. HPB (Oxford). 2019;21:1603-1612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 64]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
32.  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. Oncol Rep. 2016;35:1727-1731.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 69]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
33.  Gugenheim J, Crovetto A, Petrucciani N. Neoadjuvant therapy for pancreatic cancer. Updates Surg. 2022;74:35-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 21]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
34.  Sugimoto M, Takahashi N, Farnell MB, Smyrk TC, Truty MJ, Nagorney DM, Smoot RL, Chari ST, Carter RE, Kendrick ML. Survival benefit of neoadjuvant therapy in patients with non-metastatic pancreatic ductal adenocarcinoma: A propensity matching and intention-to-treat analysis. J Surg Oncol. 2019;120:976-984.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
35.  Cooper AB, Slack R, Fogelman D, Holmes HM, Petzel M, Parker N, Balachandran A, Garg N, Ngo-Huang A, Varadhachary G, Evans DB, Lee JE, Aloia T, Conrad C, Vauthey JN, Fleming JB, Katz MH. Characterization of Anthropometric Changes that Occur During Neoadjuvant Therapy for Potentially Resectable Pancreatic Cancer. Ann Surg Oncol. 2015;22:2416-2423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 117]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
36.  Cloyd JM, Nogueras-González GM, Prakash LR, Petzel MQB, Parker NH, Ngo-Huang AT, Fogelman D, Denbo JW, Garg N, Kim MP, Lee JE, Tzeng CD, Fleming JB, Katz MHG. Anthropometric Changes in Patients with Pancreatic Cancer Undergoing Preoperative Therapy and Pancreatoduodenectomy. J Gastrointest Surg. 2018;22:703-712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
37.  Jin K, Tang Y, Wang A, Hu Z, Liu C, Zhou H, Yu X. Body Composition and Response and Outcome of Neoadjuvant Treatment for Pancreatic Cancer. Nutr Cancer. 2022;74:100-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
38.  Kurita Y, Kobayashi N, Tokuhisa M, Goto A, Kubota K, Endo I, Nakajima A, Ichikawa Y. Sarcopenia is a reliable prognostic factor in patients with advanced pancreatic cancer receiving FOLFIRINOX chemotherapy. Pancreatology. 2019;19:127-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 61]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
39.  Lee HS, Kim SY, Chung MJ, Park JY, Bang S, Park SW, Song SY. Skeletal Muscle Mass Predicts Poor Prognosis in Patients with Advanced Pancreatic Cancer Undergoing Second-Line FOLFIRINOX Chemotherapy. Nutr Cancer. 2019;71:1100-1107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
40.  Williet N, Fovet M, Maoui K, Chevalier C, Maoui M, Le Roy B, Roblin X, Hag B, Phelip JM. A Low Total Psoas Muscle Area Index Is a Strong Prognostic Factor in Metastatic Pancreatic Cancer. Pancreas. 2021;50:579-586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Reference Citation Analysis (0)]
41.  Solheim TS, Laird BJA, Balstad TR, Stene GB, Bye A, Johns N, Pettersen CH, Fallon M, Fayers P, Fearon K, Kaasa S. A randomized phase II feasibility trial of a multimodal intervention for the management of cachexia in lung and pancreatic cancer. J Cachexia Sarcopenia Muscle. 2017;8:778-788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 205]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
42.  Griffin OM, Bashir Y, O'Connor D, Peakin J, McMahon J, Duggan SN, Geoghegan J, Conlon KC. Measurement of body composition in pancreatic cancer: a systematic review, meta-analysis and recommendations for future study design. Dig Surg. 2022;Online ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
43.  Wu CH, Chang MC, Lyadov VK, Liang PC, Chen CM, Shih TT, Chang YT. Comparing Western and Eastern criteria for sarcopenia and their association with survival in patients with pancreatic cancer. Clin Nutr. 2019;38:862-869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
44.  Hsu TH, Schawkat K, Berkowitz SJ, Wei JL, Makoyeva A, Legare K, DeCicco C, Paez SN, Wu JSH, Szolovits P, Kikinis R, Moser AJ, Goehler A. Artificial intelligence to assess body composition on routine abdominal CT scans and predict mortality in pancreatic cancer- A recipe for your local application. Eur J Radiol. 2021;142:109834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]