1
|
Li W, Zhu H, Dong H, Shi B, Qin Z, Huang F, Yu Z, Liu S, Wang Z, Chen J. Body Composition Decrease and Impact on Clinical Outcome in Gastric Cancer Patients Undergoing Radical Gastrectomy After Neoadjuvant Treatment. Nutr Cancer 2024:1-12. [PMID: 39468458 DOI: 10.1080/01635581.2024.2418622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024]
Abstract
This study investigates the impact of neoadjuvant therapy (NT) on body composition and its correlation with long-term survival and other clinical outcomes in patients with advanced gastric cancer. We utilized Computed Tomography (CT) scans to measure body composition before and after NT, including Subcutaneous Adipose Tissue Index (SATI), Visceral Adipose Tissue Index (VATI), Skeletal Muscle Index (SMI), and Muscle Density (MA). We then analyzed the decrease in body composition in relation to tumor regression, inflammatory markers, nutritional scores, and long-term survival. Our findings reveal a negative correlation between the decrease in SATI and VATI after NT, and both tumor regression and nutritional score. Notably, patients who experienced a significant loss in SATI or VATI post-NT had shorter Recurrence-Free Survival (RFS) and Overall Survival (OS). Additionally, significant loss in SATI and VATI emerged as an independent risk factor for both RFS and OS. In conclusion, our study convincingly demonstrates that in patients with advanced gastric cancer, SATI and VATI decreases after NT and is negatively associated with tumor regression and nutritional score. A significant loss in SATI and VATI is a risk factor for shorter RFS and OS, thereby underscoring the importance of maintaining body composition during NT.
Collapse
Affiliation(s)
- Wei Li
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery after Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
| | - Hai Zhu
- Guangxi key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Haizheng Dong
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bo Shi
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery after Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
| | - Zhengkun Qin
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fuling Huang
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
| | - Zhu Yu
- Guangxi key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery after Surgery, Nanning, China
| | - Siyu Liu
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery after Surgery, Nanning, China
| | - Zhen Wang
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery after Surgery, Nanning, China
| | - Junqiang Chen
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery after Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
| |
Collapse
|
2
|
Xu X, Liu WH, Yates P. Changes in Skeletal Muscle Mass in the First 3 Months Following Gastrointestinal Cancer Surgery: A Prospective Study. Ann Surg Oncol 2024:10.1245/s10434-024-16109-8. [PMID: 39230851 DOI: 10.1245/s10434-024-16109-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Surgical resection is the primary treatment for gastrointestinal (GI) cancers, but postoperative skeletal muscle loss (SML) is common and linked to poor prognosis. This study aims to identify patterns of muscle change, examine its association with quality of life (QoL), and explore predictors of SML in the first 3 months. PATIENTS AND METHODS A prospective cohort study was conducted on patients newly diagnosed with GI cancer and undergoing surgery in China between September 2021 and May 2022. Skeletal muscle mass (SMM) and QoL were assessed at admission, 7 days, 1 month, and 3 months post-surgery. Demographic, clinical data, and biomarkers were collected. Missing data were imputed using multiple imputation. Data were analyzed using growth mixture modelling, bivariate analyses, and logistic regression. RESULTS A total of 483 patients completed baseline assessment. Of the 242 patients with complete muscle assessments, 92% experienced SML. Three distinct patterns of muscle change were identified: 57% had normal preoperative SMM with mild postoperative SML, 16% had low preoperative SMM with moderate SML, and 27% had normal preoperative mass but severe postoperative SML. Moderate/severe SML was associated with more postoperative complications, poorer health, and higher symptom burden. Independent predictors included advanced age, preoperative sarcopenia, advanced cancer stage, and low prognostic nutrition index (PNI ≤ 45). The results did not change when using imputed values. CONCLUSIONS Although SML is prevalent, patterns of muscle change are heterogeneous among patients. Advanced age, preoperative sarcopenia, advanced cancer stage, and cancer-related inflammation are predictors for moderate/severe SML, highlighting the need for early detection and management.
Collapse
Affiliation(s)
- Xinyi Xu
- Cancer and Palliative Care Outcomes Centre, Centre for Healthcare Transformation, School of Nursing, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Wei-Hong Liu
- Cancer and Palliative Care Outcomes Centre, Centre for Healthcare Transformation, School of Nursing, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Patsy Yates
- Cancer and Palliative Care Outcomes Centre, Centre for Healthcare Transformation, School of Nursing, Faculty of Health, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
3
|
Zhou D, Zuo J, Zeng C, Zhang L, Gao X, Li G, Wang X. Impact of body composition, grip strength, and physical performance on clinical outcomes for locally advanced gastric cancer during neoadjuvant chemotherapy: A prospective cohort study. Nutrition 2024; 125:112472. [PMID: 38810532 DOI: 10.1016/j.nut.2024.112472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/14/2024] [Accepted: 04/19/2024] [Indexed: 05/31/2024]
Abstract
OBJECTIVE Neoadjuvant chemotherapy (NC) is critical in treating locally advanced gastric cancer (LAGC). However, the effect of body composition, grip strength, and physical performance during neoadjuvant chemotherapy remains uncertain. This study aimed to investigate the impact of these factors on perioperative clinical outcomes in LAGC patients undergoing NC. METHODS A total of 162 consecutive patients receiving NC at two centers were prospectively registered between June 2022 and September 2023. The data on body composition parameters, grip strength, and physical performance during NC were collected, compared, and analyzed. The primary outcome was the tumor response after completion of NC. RESULTS Overall, we included 92 LAGC patients. No significant changes were observed in body composition, grip strength, and physical performance after NC. The change in skeletal muscle index and grip strength were both significantly lower in the patients with poor tumor response. According to the Youden index, the cutoff values of △SMI and △grip strength were -2.0 and -2.8, respectively. Based on these two parameters, the area under the curve to predict tumor response was 0.817 (P < 0.001). Furthermore, visceral fat index (VFI) loss >6.9 and 5-time chair stand test increase >2.4 independently predicted postoperative complication (OR: 3.82, 95% CI: 1.138-12.815, P = 0.030; OR: 5.01, 95% CI: 1.086-23.131, P = 0.039, respectively). CONCLUSIONS For LAGC patients receiving NC, changes in SMI, VFI, grip strength, and physical status can predict perioperative clinical outcomes. These patients should be given special nutritional intervention.
Collapse
Affiliation(s)
- Da Zhou
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Junbo Zuo
- Department of General Surgery, The Affiliated Jinling Hospital of Nanjing Medical University, Nanjing, China; Department of General Surgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Chenghao Zeng
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Zhang
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuejin Gao
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Guoli Li
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinying Wang
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Department of General Surgery, The Affiliated Jinling Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Svrcek M, Voron T, André T, Smyth EC, de la Fouchardière C. Improving individualised therapies in localised gastro-oesophageal adenocarcinoma. Lancet Oncol 2024; 25:e452-e463. [PMID: 39214116 DOI: 10.1016/s1470-2045(24)00180-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/05/2024] [Accepted: 03/20/2024] [Indexed: 09/04/2024]
Abstract
Despite our increased understanding of the biological and molecular aspects of gastro-oesophageal tumourigenesis, the identification of prognostic or predictive factors remains challenging. Patients with resectable gastric and oesophageal adenocarcinoma are often treated similarly after surgical resection, regardless of their tumour biology, clinical characteristics, and histological treatment response. Substantial progress has been made in the past 5 years in managing patients with gastric or oesophageal adenocarcinoma, including the use of immune checkpoint inhibitors and new targeted therapies, leading to substantial improvements in clinical outcomes. These advancements have primarily been established in advanced and metastatic disease, while the management framework for local and locoregional disease is just beginning to shift. We provide an overview of existing data on biomarkers and tumour-related and host-related factors that are relevant to stratify patients into low-risk and high-risk recurrence groups, both before and after surgery, paving the way for more personalised treatment approaches.
Collapse
Affiliation(s)
- Magali Svrcek
- Department of Pathology, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | - Thibault Voron
- Digestive Surgery Department, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | - Thierry André
- Department of Medical Oncology, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | | | | |
Collapse
|
5
|
Li W, Zhu H, Dong HZ, Qin ZK, Huang FL, Yu Z, Liu SY, Wang Z, Chen JQ. Impact of body composition parameters, age, and tumor staging on gastric cancer prognosis. Eur J Cancer Prev 2024:00008469-990000000-00167. [PMID: 39229969 DOI: 10.1097/cej.0000000000000917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
BACKGROUND Research studies on gastric cancer have not investigated the combined impact of body composition, age, and tumor staging on gastric cancer prognosis. To address this gap, we used machine learning methods to develop reliable prediction models for gastric cancer. METHODS This study included 1,132 gastric cancer patients, with preoperative body composition and clinical parameters recorded, analyzed using Cox regression and machine learning models. RESULTS The multivariate analysis revealed that several factors were associated with recurrence-free survival (RFS) and overall survival (OS) in gastric cancer. These factors included age (≥65 years), tumor-node-metastasis (TNM) staging, low muscle attenuation (MA), low skeletal muscle index (SMI), and low visceral to subcutaneous adipose tissue area ratios (VSR). The decision tree analysis for RFS identified six subgroups, with the TNM staging I, II combined with high MA subgroup showing the most favorable prognosis and the TNM staging III combined with low MA subgroup exhibiting the poorest prognosis. For OS, the decision tree analysis identified seven subgroups, with the subgroup featuring high MA combined with TNM staging I, II showing the best prognosis and the subgroup with low MA, TNM staging II, III, low SMI, and age ≥65 years associated with the worst prognosis. CONCLUSION Cox regression identified key factors associated with gastric cancer prognosis, and decision tree analysis determined prognoses across different risk factor subgroups. Our study highlights that the combined use of these methods can enhance intervention planning and clinical decision-making in gastric cancer.
Collapse
Affiliation(s)
- Wei Li
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer
- Guangxi Clinical Research Center for Enhanced Recovery after Surgery
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images and
| | - Hai Zhu
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hai-Zheng Dong
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University
| | - Zheng-Kun Qin
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University
| | - Fu-Ling Huang
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images and
| | - Zhu Yu
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images and
| | - Shi-Yu Liu
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer
- Guangxi Clinical Research Center for Enhanced Recovery after Surgery
| | - Zhen Wang
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer
- Guangxi Clinical Research Center for Enhanced Recovery after Surgery
| | - Jun-Qiang Chen
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer
- Guangxi Clinical Research Center for Enhanced Recovery after Surgery
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images and
| |
Collapse
|
6
|
Juez LD, Priego P, Cuadrado M, Blázquez LA, Sánchez-Picot S, Gil P, Longo F, Galindo J, Fernández-Cebrián JM, Botella-Carretero JI. Impact of Neoadjuvant Treatment on Body Composition in Patients with Locally Advanced Gastric Cancer. Cancers (Basel) 2024; 16:2408. [PMID: 39001470 PMCID: PMC11240361 DOI: 10.3390/cancers16132408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Neoadjuvant chemotherapy (NT) followed by radical surgery is the standard treatment for locally advanced gastric cancer (GC). The incidence of sarcopenia in upper gastrointestinal tract malignancies is very high, and it may be increased after NT. This study aimed to evaluate the impact of NT on body composition. A retrospective study of patients with locally advanced GC undergoing gastrectomy who had received NT in a tertiary hospital between 2012 and 2019 was conducted. CT measured the skeletal muscle index, total psoas area, and visceral and subcutaneous adipose tissue before and after NT. Of the 180 gastrectomies for GC, 61 patients received NT. During NT, changes in body composition were observed with a decrease in the skeletal muscle mass index (SMMI -2.5%; p < 0.001), and these changes were significantly greater in men (SMMI -10.55%). Before surgery, patients who received NT presented 15% more sarcopenia than those without NT (p = 0.048). In conclusion, patients with locally advanced gastric cancer who receive NT have significant changes in body composition during chemotherapy. These changes, which are at the expense of a loss of muscle mass, lead to an increased incidence of pre-surgical sarcopenia.
Collapse
Affiliation(s)
- Luz Divina Juez
- Department of General and Digestive Surgery, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria, IRyCIS, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Faculty of Medicine, University of Alcalá (UAH), Alcalá de Henares, 28801 Madrid, Spain
| | - Pablo Priego
- Department of General and Digestive Surgery, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Marta Cuadrado
- Department of General and Digestive Surgery, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria, IRyCIS, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Luis A Blázquez
- Department of General and Digestive Surgery, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria, IRyCIS, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Faculty of Medicine, University of Alcalá (UAH), Alcalá de Henares, 28801 Madrid, Spain
| | - Silvia Sánchez-Picot
- Department of General and Digestive Surgery, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Pablo Gil
- Department of General and Digestive Surgery, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Faculty of Medicine, University of Alcalá (UAH), Alcalá de Henares, 28801 Madrid, Spain
| | - Federico Longo
- Instituto Ramón y Cajal de Investigación Sanitaria, IRyCIS, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Department of Clinical Oncology, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Julio Galindo
- Department of General and Digestive Surgery, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria, IRyCIS, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Faculty of Medicine, University of Alcalá (UAH), Alcalá de Henares, 28801 Madrid, Spain
| | - José María Fernández-Cebrián
- Department of General and Digestive Surgery, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria, IRyCIS, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Faculty of Medicine, University of Alcalá (UAH), Alcalá de Henares, 28801 Madrid, Spain
| | - José I Botella-Carretero
- Instituto Ramón y Cajal de Investigación Sanitaria, IRyCIS, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Faculty of Medicine, University of Alcalá (UAH), Alcalá de Henares, 28801 Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| |
Collapse
|
7
|
Zheng K, Liu X, Li Y, Cui J, Li W. CT-based muscle and adipose measurements predict prognosis in patients with digestive system malignancy. Sci Rep 2024; 14:13036. [PMID: 38844600 PMCID: PMC11156914 DOI: 10.1038/s41598-024-63806-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
The role of skeletal muscle and adipose tissue in the progression of cancer has been gradually discussed, but it needs further exploration. The objective of this study was to provide an in-depth analysis of skeletal muscle and fat in digestive malignancies and to construct novel predictors for clinical management. This is a retrospective study that includes data from Cancer Center, the First Hospital of Jilin University. Basic characteristic information was analyzed by T tests. Correlation matrices were drawn to explore the relationship between CT-related indicators and other indicators. Cox risk regression analyses were performed to analyze the association between the overall survivals (OS) and various types of indicators. A new indicator body composition score (BCS) was then created and a time-dependent receiver operating characteristic curve was plotted to analyze the efficacy of the BCS. Finally, a nomogram was produced to develop a scored-CT system based on BCS and other indicators. C-index and calibration curve analyses were performed to validate the predictive accuracy of the scored-CT system. A total of 575 participants were enrolled in the study. Cox risk regression model revealed that VFD, L3 SMI and VFA/SFA were associated with prognosis of cancer patients. After adjustment, BCS index based on CT was significantly associated with prognosis, both in all study population and in subgroup analysis according to tumor types (all study population: HR 2.036, P < 0.001; colorectal cancer: HR 2.693, P < 0.001; hepatocellular carcinoma: HR 4.863, P < 0.001; esophageal cancer: HR 4.431, P = 0.008; pancreatic cancer: HR 1.905, P = 0.016; biliary system malignancies: HR 23.829, P = 0.035). The scored-CT system was constructed according to tumor type, stage, KPS, PG-SGA and BCS index, and it was of great predictive validity. This study identified VFD, L3 SMI and VFA/SFA associated with digestive malignancies outcomes. BCS was created and the scored-CT system was established to predict the OS of cancer patients.
Collapse
Affiliation(s)
- Kaiwen Zheng
- Cancer Center, The First Hospital of Jilin University, Xinmin St No 126, Changchun, 130021, Jilin, China
| | - Xiangliang Liu
- Cancer Center, The First Hospital of Jilin University, Xinmin St No 126, Changchun, 130021, Jilin, China
| | - Yuguang Li
- College of Instrumentation and Electrical Engineering, Jilin University, Changchun, Jilin, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Xinmin St No 126, Changchun, 130021, Jilin, China.
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Xinmin St No 126, Changchun, 130021, Jilin, China.
| |
Collapse
|
8
|
Rozynek M, Tabor Z, Kłęk S, Wojciechowski W. Body composition radiomic features as a predictor of survival in patients with non-small cellular lung carcinoma: A multicenter retrospective study. Nutrition 2024; 120:112336. [PMID: 38237479 DOI: 10.1016/j.nut.2023.112336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 02/24/2024]
Abstract
OBJECTIVES This study combined two novel approaches in oncology patient outcome predictions-body composition and radiomic features analysis. The aim of this study was to validate whether automatically extracted muscle and adipose tissue radiomic features could be used as a predictor of survival in patients with non-small cell lung cancer. METHODS The study included 178 patients with non-small cell lung cancer receiving concurrent platinum-based chemoradiotherapy. Abdominal imaging was conducted as a part of whole-body positron emission tomography/computed tomography performed before therapy. Methods used included automated assessment of the volume of interest using densely connected convolutional network classification model - DenseNet121, automated muscle and adipose tissue segmentation using U-net architecture implemented in nnUnet framework, and radiomic features extraction. Acquired body composition radiomic features and clinical data were used for overall and 1-y survival prediction using machine learning classification algorithms. RESULTS The volume of interest detection model achieved the following metric scores: 0.98 accuracy, 0.89 precision, 0.96 recall, and 0.92 F1 score. Automated segmentation achieved a median dice coefficient >0.99 in all segmented regions. We extracted 330 body composition radiomic features for every patient. For overall survival prediction using clinical and radiomic data, the best-performing feature selection and prediction method achieved areas under the curve-receiver operating characteristic (AUC-ROC) of 0.73 (P < 0.05); for 1-y survival prediction AUC-ROC was 0.74 (P < 0.05). CONCLUSION Automatically extracted muscle and adipose tissue radiomic features could be used as a predictor of survival in patients with non-small cell lung cancer.
Collapse
Affiliation(s)
- Miłosz Rozynek
- Department of Radiology, Jagiellonian University Medical College, Krakow, Poland
| | - Zbisław Tabor
- AGH University of Science and Technology, Krakow, Poland
| | - Stanisław Kłęk
- Surgical Oncology Clinic, Maria Skłodowska-Curie National Cancer Institute, Krakow, Poland
| | - Wadim Wojciechowski
- Department of Radiology, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
9
|
Yin L, Song C, Cui J, Lin X, Li N, Fan Y, Zhang L, Liu J, Chong F, Cong M, Li Z, Li S, Guo Z, Li W, Shi H, Xu H. Association of possible sarcopenia with all-cause mortality in patients with solid cancer: A nationwide multicenter cohort study. J Nutr Health Aging 2024; 28:100023. [PMID: 38216426 DOI: 10.1016/j.jnha.2023.100023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 10/25/2023] [Indexed: 01/14/2024]
Abstract
OBJECTIVES The concept of possible sarcopenia (PS) was recently introduced to enable timely intervention in settings without the technologies required to make a full diagnosis of sarcopenia. This study aimed to investigate the association between PS and all-cause mortality in patients with solid cancer. DESIGN Retrospective observational study. SETTING AND PARTICIPANTS 13,736 patients with 16 types of solid cancer who were ≥18 years old. MEASUREMENTS The presence of both a low calf circumference (men <34 cm or women <33 cm) and low handgrip strength (men <28 kg or women <18 kg) was considered to indicate PS. Harrell's C-index was used to assess prognostic value and the association of PS with mortality was estimated by calculating multivariable-adjusted hazard ratios (HRs). RESULTS The study enrolled 7207 men and 6529 women (median age = 57.8 years). During a median follow-up of 43 months, 3150 deaths occurred. PS showed higher Harrell's C-index (0.549, 95%CI = [0.541, 0.557]) than the low calf circumference (0.541, 95%CI = [0.531, 0.551], P = 0.037) or low handgrip strength (0.542, 95%CI = [0.532, 0.552], P = 0.026). PS was associated with increased mortality risk in both univariate (HR = 1.587, 95%CI = [1.476, 1.708]) and multivariable-adjusted models (HR = 1.190, 95%CI = [1.094, 1.293]). Sensitivity analyses showed that the association of PS with mortality was robust in different covariate subgroups, which also held after excluding those patients who died within the first 3 months (HR = 1.162, 95%CI = [1.060, 1.273]), 6 months (HR = 1.150, 95%CI = [1.039, 1.274]) and 12 months (HR = 1.139, 95%CI = [1.002, 1.296]) after enrollment. CONCLUSION PS could independently and robustly predict all-cause mortality in patients with solid cancer. These findings imply the importance of including PS assessment in routine cancer care to provide significant prognostic information to help mitigate sarcopenia-related premature deaths.
Collapse
Affiliation(s)
- Liangyu Yin
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China; Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chunhua Song
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Jiuwei Cui
- Cancer Center of the First Hospital of Jilin University, Changchun 130021, China
| | - Xin Lin
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Na Li
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Yang Fan
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Ling Zhang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Jie Liu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Feifei Chong
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Minghua Cong
- Department of Comprehensive Oncology, National Cancer Center or Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zengning Li
- Department of Clinical Nutrition, The First Hospital of Hebei Medical University, Shijiazhuang 050031, China
| | - Suyi Li
- Department of Nutrition and Metabolism of Oncology, The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Cancer Hospital), Hefei 230031, China
| | - Zengqing Guo
- Department of Medical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Wei Li
- Cancer Center of the First Hospital of Jilin University, Changchun 130021, China
| | - Hanping Shi
- Department of Gastrointestinal Surgery and Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing 100038, China.
| | - Hongxia Xu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China.
| |
Collapse
|
10
|
de Jongh C, van der Veen A, Brosens LAA, Nieuwenhuijzen GAP, Stoot JHMB, Ruurda JP, van Hillegersberg R. Distal Versus Total D2-Gastrectomy for Gastric Cancer: a Secondary Analysis of Surgical and Oncological Outcomes Including Quality of Life in the Multicenter Randomized LOGICA-Trial. J Gastrointest Surg 2023; 27:1812-1824. [PMID: 37340107 PMCID: PMC10511620 DOI: 10.1007/s11605-023-05683-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/10/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND Distal gastrectomy (DG) for gastric cancer can cause less morbidity than total gastrectomy (TG), but may compromise radicality. No prospective studies administered neoadjuvant chemotherapy, and few assessed quality of life (QoL). METHODS The multicenter LOGICA-trial randomized laparoscopic versus open D2-gastrectomy for resectable gastric adenocarcinoma (cT1-4aN0-3bM0) in 10 Dutch hospitals. This secondary LOGICA-analysis compared surgical and oncological outcomes after DG versus TG. DG was performed for non-proximal tumors if R0-resection was deemed achievable, TG for other tumors. Postoperative complications, mortality, hospitalization, radicality, nodal yield, 1-year survival, and EORTC-QoL-questionnaires were analyzed using Χ2-/Fisher's exact tests and regression analyses. RESULTS Between 2015 and 2018, 211 patients underwent DG (n = 122) or TG (n = 89), and 75% of patients underwent neoadjuvant chemotherapy. DG-patients were older, had more comorbidities, less diffuse type tumors, and lower cT-stage than TG-patients (p < 0.05). DG-patients experienced fewer overall complications (34% versus 57%; p < 0.001), also after correcting for baseline differences, lower anastomotic leakage (3% versus 19%), pneumonia (4% versus 22%), atrial fibrillation (3% versus 14%), and Clavien-Dindo grading compared to TG-patients (p < 0.05), and demonstrated shorter median hospital stay (6 versus 8 days; p < 0.001). QoL was better after DG (statistically significant and clinically relevant) in most 1-year postoperative time points. DG-patients showed 98% R0-resections, and similar 30-/90-day mortality, nodal yield (28 versus 30 nodes; p = 0.490), and 1-year survival after correcting for baseline differences (p = 0.084) compared to TG-patients. CONCLUSIONS If oncologically feasible, DG should be preferred over TG due to less complications, faster postoperative recovery, and better QoL while achieving equivalent oncological effectiveness. Distal D2-gastrectomy for gastric cancer resulted in less complications, shorter hospitalization, quicker recovery and better quality of life compared to total D2-gastrectomy, whereas radicality, nodal yield and survival were similar.
Collapse
Affiliation(s)
- Cas de Jongh
- Department of Surgery, University Medical Center (UMC) Utrecht, G04.228, 3508 GA Utrecht, The Netherlands
| | - Arjen van der Veen
- Department of Surgery, University Medical Center (UMC) Utrecht, G04.228, 3508 GA Utrecht, The Netherlands
| | | | | | - Jan H. M. B. Stoot
- Department of Surgery, Zuyderland Medical Center, Sittard, The Netherlands
| | - Jelle P. Ruurda
- Department of Surgery, University Medical Center (UMC) Utrecht, G04.228, 3508 GA Utrecht, The Netherlands
| | - Richard van Hillegersberg
- Department of Surgery, University Medical Center (UMC) Utrecht, G04.228, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
11
|
Correia M, Moreira I, Cabral S, Castro C, Cruz A, Magalhães B, Santos LL, Irving SC. Neoadjuvant Gastric Cancer Treatment and Associated Nutritional Critical Domains for the Optimization of Care Pathways: A Systematic Review. Nutrients 2023; 15:nu15102241. [PMID: 37242125 DOI: 10.3390/nu15102241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/26/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
(1) Background: Gastric cancer patients are known to be at a high risk of malnutrition, sarcopenia, and cachexia, and the latter impairs the patient's nutritional status during their clinical course and also treatment response. A clearer identification of nutrition-related critical points during neoadjuvant treatment for gastric cancer is relevant to managing patient care and predicting clinical outcomes. The aim of this systematic review was to identify and describe nutrition-related critical domains associated with clinical outcomes. (2) Methods: We performed a systematic review (PROSPERO ID:CRD42021266760); (3) Results: This review included 14 studies compiled into three critical domains: patient-related, clinical-related (disease and treatment), and healthcare-related. Body composition changes during neoadjuvant chemotherapy (NAC) accounted for the early termination of chemotherapy and reduced overall survival. Sarcopenia was confirmed to have an independent prognostic value. The role of nutritional interventions during NAC has not been fully explored. (4) Conclusions: Understanding critical domain exposures affecting nutritional status will enable better clinical approaches to optimize care plans. It may also provide an opportunity for the mitigation of poor nutritional status and sarcopenia and their deleterious clinical consequences.
Collapse
Affiliation(s)
- Marta Correia
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Ines Moreira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Sonia Cabral
- Portuguese Oncology Institute of Porto (IPO-Porto)-Nutrition, 4200-072 Porto, Portugal
| | - Carolina Castro
- Experimental Pathology and Therapeutics Group, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Andreia Cruz
- Experimental Pathology and Therapeutics Group, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
- Medical Oncology Department, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Bruno Magalhães
- School of Health, University of Trás-os-Montes e Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Oncology Nursing Research Unit IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
- Surgical Oncology Department, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Susana Couto Irving
- Portuguese Oncology Institute of Porto (IPO-Porto)-Nutrition, 4200-072 Porto, Portugal
| |
Collapse
|
12
|
Pedrosa MB, Barbosa S, Vitorino R, Ferreira R, Moreira-Gonçalves D, Santos LL. Chemotherapy-Induced Molecular Changes in Skeletal Muscle. Biomedicines 2023; 11:biomedicines11030905. [PMID: 36979884 PMCID: PMC10045751 DOI: 10.3390/biomedicines11030905] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Paraneoplastic conditions such as cancer cachexia are often exacerbated by chemotherapy, which affects the patient’s quality of life as well as the response to therapy. The aim of this narrative review was to overview the body-composition-related changes and molecular effects of different chemotherapy agents used in cancer treatment on skeletal-muscle remodeling. A literature search was performed using the Web of Science, Scopus, and Science Direct databases and a total of 77 papers was retrieved. In general, the literature survey showed that the molecular changes induced by chemotherapy in skeletal muscle have been studied mainly in animal models and mostly in non-tumor-bearing rodents, whereas clinical studies have essentially assessed changes in body composition by computerized tomography. Data from preclinical studies showed that chemotherapy modulates several molecular pathways in skeletal muscle, including the ubiquitin–proteasome pathway, autophagy, IGF-1/PI3K/Akt/mTOR, IL-6/JAK/STAT, and NF-κB pathway; however, the newest chemotherapy agents are underexplored. In conclusion, chemotherapy exacerbates skeletal-muscle wasting in cancer patients; however, the incomplete characterization of the chemotherapy-related molecular effects on skeletal muscle makes the development of new preventive anti-wasting strategies difficult. Therefore, further investigation on molecular mechanisms and clinical studies are necessary.
Collapse
Affiliation(s)
- Mafalda Barbosa Pedrosa
- Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (LAQV-REQUIMTE), Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
- Experimental Pathology and Therapeutics Group, Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal
- Correspondence: (M.B.P.); (L.L.S.)
| | - Samuel Barbosa
- Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (LAQV-REQUIMTE), Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
- Experimental Pathology and Therapeutics Group, Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine—iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rita Ferreira
- Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (LAQV-REQUIMTE), Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Daniel Moreira-Gonçalves
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), 4050-600 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal
- Correspondence: (M.B.P.); (L.L.S.)
| |
Collapse
|
13
|
Association of GLIM Defined Malnutrition According to Preoperative Chronic Inflammation with Long-Term Prognosis after Gastrectomy in Patients with Advanced Gastric Cancer. J Clin Med 2023; 12:jcm12041579. [PMID: 36836114 PMCID: PMC9966663 DOI: 10.3390/jcm12041579] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
This study aimed to investigate the association of malnutrition, defined by the Global Leadership Initiative on Malnutrition (GLIM) according to preoperative chronic inflammation with long-term prognosis after gastrectomy in patients with advanced gastric cancer. We included patients with primary stage I-III gastric cancer who underwent gastrectomy between April 2008 and June 2018. Patients were categorized as normal, moderate malnutrition, and severe malnutrition. Preoperative chronic inflammation was defined as a C-reactive protein level of >0.5 mg/dL. The primary endpoint was overall survival (OS), compared between the inflammation and non-inflammation groups. Among the 457 patients, 74 (16.2%) and 383 (83.8%) were included in the inflammation and non-inflammation groups, respectively. The prevalence of malnutrition was similar in both groups (p = 0.208). Multivariate analyses for OS showed that moderate malnutrition (hazard ratios: 1.749, 95% concordance interval: 1.037-2.949, p = 0.036) and severe malnutrition (hazard ratios: 1.971, 95% CI: 1.130-3.439, p = 0.017) were poor prognostic factors in the non-inflammation group, but malnutrition was not a prognostic factor in the inflammation group. In conclusion, preoperative malnutrition was a poor prognostic factor in patients without inflammation, but it was not a prognostic factor in patients with inflammation.
Collapse
|
14
|
Matsui R, Watanabe J, Banno M, Inaki N, Fukunaga T. Association of visceral adipose tissue with postoperative outcome in upper gastrointestinal cancer: a systematic review and meta-analysis. Am J Clin Nutr 2022; 116:1540-1552. [PMID: 36166841 DOI: 10.1093/ajcn/nqac273] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/22/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND There is no consensus on the relation between visceral fat mass and long-term postoperative prognosis in patients with upper gastrointestinal (GI) cancer. OBJECTIVES The purpose of this study was to investigate the association of preoperative visceral fat mass with postoperative complications and overall survival (OS) in patients with upper GI cancer. METHODS We searched MEDLINE, the Cochrane Central Register of Controlled Trials, EMBASE (Dialog), the WHO International Clinical Trials Platform Search Portal, and ClinicalTrials.gov and identified observational studies published from inception through 20 July 2022. We conducted a systematic review and random-effects meta-analysis of studies including patients who were surgically treated for upper GI cancer and whose visceral fat mass was assessed on the basis of body composition. We independently assessed the risk of bias and quality of evidence using the Quality In Prognosis Studies and the Grading of Recommendations, Assessment, Development, and Evaluation approach, respectively. The primary outcome was OS. HRs and 95% CIs for OS were pooled. RESULTS Ninety-one studies (n = 20,583) were included. All studies used computed tomography (CT) to assess the body composition of patients. Twenty-four studies reported the relation between high visceral fat and postoperative outcomes, and their results were synthesized. Compared with low visceral adipose tissue, high visceral adipose tissue assessed by CT may improve OS (HR: 0.69; 95% CI: 0.55, 0.87; I2 = 65%; n = 3407). The risk of bias for OS in each study was moderate or high. The certainty of evidence for OS was very low because of inconsistency in the forest plot, the moderate or high risk of bias, and publication bias. CONCLUSIONS High visceral fat may be associated with improved OS after radical resection in patients with upper GI cancer. Further studies are needed to confirm these findings and mitigate the risk.
Collapse
Affiliation(s)
- Ryota Matsui
- Department of Surgery, Juntendo University Urayasu Hospital, Urayasu City, Japan.,Department of Gastroenterology and Minimally Invasive Surgery, Juntendo University Hospital, Tokyo, Japan
| | - Jun Watanabe
- Department of Surgery, Division of Gastroenterological, General, and Transplant Surgery, Jichi Medical University, Shimotsuke City, Japan.,Center for Community Medicine, Jichi Medical University, Shimotsuke City, Japan.,Scientific Research WorkS Peer Support Group (SRWS-PSG), Osaka, Japan
| | - Masahiro Banno
- Scientific Research WorkS Peer Support Group (SRWS-PSG), Osaka, Japan.,Department of Psychiatry, Seichiryo Hospital, Nagoya, Japan.,Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noriyuki Inaki
- Department of Gastrointestinal Surgery/Breast Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa City, Japan
| | - Tetsu Fukunaga
- Department of Gastroenterology and Minimally Invasive Surgery, Juntendo University Hospital, Tokyo, Japan
| |
Collapse
|
15
|
Lin GT, Huang JB, Lin JL, Lin JX, Xie JW, Wang JB, Lu J, Zheng CH, Huang CM, Li P. Body composition parameters for predicting the efficacy of neoadjuvant chemotherapy with immunotherapy for gastric cancer. Front Immunol 2022; 13:1061044. [PMID: 36569876 PMCID: PMC9772614 DOI: 10.3389/fimmu.2022.1061044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Background Immune checkpoint inhibitors are increasingly used in neoadjuvant therapy for locally advanced gastric cancer. However, the effect of body composition on the efficacy of neoadjuvant therapy has not been reported. Methods The computed tomography (CT) images and clinicopathological data of 101 patients with locally advanced gastric cancer who received neoadjuvant chemotherapy combined with immunotherapy (NCI) from 2019 to 2021 were collected. The CT image of L3 vertebral body section was selected, and the body composition before and after the neoadjuvant treatment was calculated using the SliceOmatic software, mainly including skeletal muscle index (SMI), subcutaneous adipose index (SAI), and visceral adipose index (VAI). The relationship between body composition and the efficacy and adverse events of NCI was analyzed. Results Of the 101 patients, 81 with evaluable data were included in the analysis. Of the included patients, 77.8% were male; the median age of all the patients was 62 years, and the median neoadjuvant therapy cycle was three. After the neoadjuvant therapy, 62.9% of the tumors were in remission (residual tumor cells ≤ 50%), and 37.1% of the tumors had no remission (residual tumor cells>50%). Moreover, 61.7% of the patients had treatment-related adverse events (TRAEs), and 18.5% had immune-related adverse events (irAEs). After neoadjuvant therapy, the body mass index (from 23 to 22.6 cm2/m2, p=0.042), SAI (from 34.7 to 32.9 cm2/m2, p=0.01) and VAI (from 32.4 to 26.8 cm2/m2, p=0.005) were significantly lower than those before treatment, while the SMI had no significant change (44.7 vs 42.5 cm2/m2, p=0.278). The multivariate logistics regression analysis revealed that low SMI (odds ratio [OR]: 3.23,95% confidence interval [CI]: 1.06-9.81, p=0.047), SMI attenuation (△SMI) ≥ 1.8(OR: 1.45,95%CI: 1.20-3.48, p=0.048), and clinical node positivity (OR: 6.99,95%CI: 2.35-20.82, p=0.001) were independent risk factors for non-remission. Additionally, high SAI is an independent risk factor for irAEs (OR: 14, 95%CI: 1.73-112.7, p=0.013). Conclusion Low SMI and △SMI≥1.8 are independent risk factors for poor tumor regression in patients with advanced gastric cancer receiving NCI. Patients with a high SAI are more likely to develop irAEs.
Collapse
Affiliation(s)
- Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jiao-Bao Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ju-Li Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
16
|
Zhang Y, Jiang L, Su P, Yu T, Ma Z, Kang W, Liu Y, Jin Z, Yu J. Visceral Adipose Tissue Assessment Enhances the Prognostic Value of GLIM Criteria in Patients with Gastric Cancer Undergoing Radical Gastrectomy after Neoadjuvant Treatment. Nutrients 2022; 14:5047. [PMID: 36501076 PMCID: PMC9740239 DOI: 10.3390/nu14235047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/20/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The Global Leadership Initiative on Malnutrition (GLIM) criteria has been recently published for diagnosing malnutrition in adults. However, the validity of the GLIM criteria has not been well-established in patients with gastric cancer (GC) treated with neoadjuvant treatment (NT) followed by radical gastrectomy. The present study aimed to explore the prognostic value of GLIM-defined malnutrition before NT and after NT in GC patients and to investigate whether additional visceral adipose tissue (VAT) assessment could improve the predictive power of the GLIM criteria for NT-related adverse events (AEs) and long-term survival. METHODS GC patients who underwent radical surgery after NT from June 2016 to June 2020 were enrolled in this study. The cross-sectional areas of total skeletal muscle (TSM) and VAT were measured using computed tomography (CT) before NT and after NT. GLIM-defined malnutrition was diagnosed using the two-step approach, including nutritional risk screening and diagnostic assessment. Low VAT was also added to the diagnosis of malnutrition in this study. The predictive value of these malnutrition diagnoses for NT-related AEs, and long-term survival was evaluated in GC patients. RESULTS A total of 182 GC patients were included in this study, of which 66 (36.3%) patients before NT and 55 (30.2%) patients after NT were diagnosed with GLIM-defined malnutrition, respectively. In addition to GLIM-defined malnutrition, 54 (29.7%) patients had additional low VAT before NT, and 39 (21.4%) patients had additional low VAT after NT. GLIM-defined malnutrition alone before NT was not associated with NT-related AEs in GC patients. The addition of low VAT to GLIM-defined malnutrition led to a significant predictive value for NT-related AEs. Furthermore, GLIM-defined malnutrition before NT and after NT were both identified as independent risk factors for overall survival (OS) and disease-free survival (DFS). The combination of low VAT and GLIM-defined malnutrition showed a higher hazard ratio for the prediction of OS and DFS both before NT and after NT. CONCLUSIONS The addition of VAT assessment using CT improved the predictive value of GLIM-defined malnutrition for NT-related AEs and long-term survival in GC patients treated with NT followed by radical gastrectomy, which further supports the prognostic importance of assessing adipose tissue simultaneously during the routine nutritional assessment in patients with cancer.
Collapse
Affiliation(s)
- Yingjing Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lin Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Pengfei Su
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Tian Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhiqiang Ma
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Weiming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yuqin Liu
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhengyu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianchun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
17
|
Zhang Y, Jiang L, Su P, Ma Z, Kang W, Ye X, Liu Y, Yu J. Association between Plasma FGF21 Levels and Body Composition in Patients with Gastric Cancer. Nutr Cancer 2022; 75:349-356. [PMID: 36190321 DOI: 10.1080/01635581.2022.2118322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Accumulating evidence has suggested that Fibroblast growth factor 21 (FGF21) plays an important role in metabolic diseases. This study aimed to investigate the relationship between plasma FGF21 levels and body composition parameters in gastric cancer (GC) patients. METHODS This study was cross-sectional based on a prospective cohort of GC patients in a single center. Computer tomography (CT) and bioelectrical impedance analysis (BIA) were used to estimate skeletal muscle and adipose tissue mass. Blood samples were collected and plasma concentrations of FGF21 were measured by ELISA. Spearman's rank correlation test and logistic regression analysis were performed to assess associations between plasma FGF21 levels and these body composition parameters. RESULTS A total of 66 GC patients were enrolled in this study. Plasma FGF21 levels were significantly higher in women compared with men. The plasma FGF21 levels were positively correlated with fat mass index (FMI), fat mass percentage (FM%), and subcutaneous adipose tissue index (SATI). Furthermore, after adjustment for confounders, the lower plasma FGF21 levels were remain associated with increased odds for low SATI. CONCLUSIONS Plasma FGF21 levels were positively associated with FMI, FM%, and SATI in GC patients, suggesting a potential mechanistic link between FGF21 and subcutaneous adipose tissue in GC.
Collapse
Affiliation(s)
- Yingjing Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pengfei Su
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiqiang Ma
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Ye
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuqin Liu
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianchun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Cheng E, Kirley J, Cespedes Feliciano EM, Caan BJ. Adiposity and cancer survival: a systematic review and meta-analysis. Cancer Causes Control 2022; 33:1219-1246. [PMID: 35971021 PMCID: PMC10101770 DOI: 10.1007/s10552-022-01613-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 07/07/2022] [Indexed: 10/28/2022]
Abstract
PURPOSE The increasing availability of clinical imaging tests (especially CT and MRI) that directly quantify adipose tissue has led to a rapid increase in studies examining the relationship of visceral, subcutaneous, and overall adiposity to cancer survival. To summarize this emerging body of literature, we conducted a systematic review and meta-analysis of imaging-measured as well as anthropometric proxies for adipose tissue distribution and cancer survival across a wide range of cancer types. METHODS Using keywords related to adiposity, cancer, and survival, we conducted a systematic search of the literature in PubMed and MEDLINE, Embase, and Web of Science Core Collection databases from database inception to 30 June 2021. We used a random-effect method to calculate pooled hazard ratios (HR) and corresponding 95% confidence intervals (CI) within each cancer type and tested for heterogeneity using Cochran's Q test and the I2 test. RESULTS We included 203 records for this review, of which 128 records were utilized for quantitative analysis among 10 cancer types: breast, colorectal, gastroesophageal, head and neck, hepatocellular carcinoma, lung, ovarian, pancreatic, prostate, and renal cancer. We found that imaging-measured visceral, subcutaneous, and total adiposity were not significantly associated with increased risk of overall mortality, death from primary cancer, or cancer progression among patients diagnosed with these 10 cancer types; however, we found significant or high heterogeneity for many cancer types. For example, heterogeneity was similarly high when the pooled HRs (95% CI) for overall mortality associated with visceral adiposity were essentially null as in 1.03 (0.55, 1.92; I2 = 58%) for breast, 0.99 (0.81, 1.21; I2 = 71%) for colorectal, versus when they demonstrated a potential increased risk 1.17 (0.85, 1.60; I2 = 78%) for hepatocellular carcinoma and 1.62 (0.90, 2.95; I2 = 84%) for renal cancer. CONCLUSION Greater adiposity at diagnosis (directly measured by imaging) is not associated with worse survival among cancer survivors. However, heterogeneity and other potential limitations were noted across studies, suggesting differences in study design and adiposity measurement approaches, making interpretation of meta-analyses challenging. Future work to standardize imaging measurements and data analyses will strengthen research on the role of adiposity in cancer survival.
Collapse
Affiliation(s)
- En Cheng
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
| | - Jocelyn Kirley
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA
| | | | - Bette J Caan
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA, 94612, USA.
| |
Collapse
|
19
|
Karra P, Winn M, Pauleck S, Bulsiewicz-Jacobsen A, Peterson L, Coletta A, Doherty J, Ulrich CM, Summers SA, Gunter M, Hardikar S, Playdon MC. Metabolic dysfunction and obesity-related cancer: Beyond obesity and metabolic syndrome. Obesity (Silver Spring) 2022; 30:1323-1334. [PMID: 35785479 PMCID: PMC9302704 DOI: 10.1002/oby.23444] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/24/2022] [Accepted: 03/09/2022] [Indexed: 01/26/2023]
Abstract
OBJECTIVES The metabolic dysfunction driven by obesity, including hyperglycemia and dyslipidemia, increases risk for developing at least 13 cancer types. The concept of "metabolic dysfunction" is often defined by meeting various combinations of criteria for metabolic syndrome. However, the lack of a unified definition of metabolic dysfunction makes it difficult to compare findings across studies. This review summarizes 129 studies that evaluated variable definitions of metabolic dysfunction in relation to obesity-related cancer risk and mortality after a cancer diagnosis. Strategies for metabolic dysfunction management are also discussed. METHODS A comprehensive search of relevant publications in MEDLINE (PubMed) and Google Scholar with review of references was conducted. RESULTS Metabolic dysfunction, defined as metabolic syndrome diagnosis or any number of metabolic syndrome criteria out of clinical range, inflammatory biomarkers, or markers of metabolic organ function, has been associated with risk for, and mortality from, colorectal, pancreatic, postmenopausal breast, and bladder cancers. Metabolic dysfunction associations with breast and colorectal cancer risk have been observed independently of BMI, with increased risk in individuals with metabolically unhealthy normal weight or overweight/obesity compared with metabolically healthy normal weight. CONCLUSION Metabolic dysfunction is a key risk factor for obesity-related cancer, regardless of obesity status. Nonetheless, a harmonized definition of metabolic dysfunction will further clarify the magnitude of the relationship across cancer types, enable better comparisons across studies, and further guide criteria for obesity-related cancer risk stratification.
Collapse
Affiliation(s)
- Prasoona Karra
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah, USA
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Maci Winn
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Population Health Sciences, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Svenja Pauleck
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | | | - Lacie Peterson
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah, USA
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Adriana Coletta
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Health and Kinesiology, University of Utah, Salt Lake City, Utah, USA
| | - Jennifer Doherty
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Population Health Sciences, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Cornelia M. Ulrich
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Population Health Sciences, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Scott A. Summers
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah, USA
| | - Marc Gunter
- Nutrition and Metabolism Section, International Agency for Research on Cancer, World Health Organization, Lyon, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, United Kingdom
| | - Sheetal Hardikar
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Department of Population Health Sciences, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Mary C. Playdon
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah, USA
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| |
Collapse
|
20
|
Xu XY, Jiang XM, Xu Q, Xu H, Luo JH, Yao C, Ding LY, Zhu SQ. Skeletal Muscle Change During Neoadjuvant Therapy and Its Impact on Prognosis in Patients With Gastrointestinal Cancers: A Systematic Review and Meta-Analysis. Front Oncol 2022; 12:892935. [PMID: 35692760 PMCID: PMC9186070 DOI: 10.3389/fonc.2022.892935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundGastrointestinal cancers are the most common malignant tumors worldwide. As the improvement of survival by surgical resection alone for cancers is close to the bottleneck, recent neoadjuvant therapy has been emphasized and applied in the treatment. Despite the advantage on improving the prognosis, some studies have reported neoadjuvant therapy could reduce skeletal muscle and therefore affect postoperative outcomes. However, the conclusions are still controversial.MethodsPubMed, CINAHL, Embase, and Cochrane Library were searched from inception to September 2, 2021. The inclusion criteria were observational studies, published in English, of individuals aged ≥18 years who underwent neoadjuvant therapy with gastrointestinal cancers and were assessed skeletal muscle mass before and after neoadjuvant therapy, with sufficient data on skeletal muscle change or the association with clinical outcomes. Meta-analysis was conducted by using the STATA 12.0 package when more than two studies reported the same outcome.ResultsA total of 268 articles were identified, and 19 studies (1,954 patients) were included in the review. The fixed effects model showed that the risk of sarcopenia increased 22% after receiving neoadjuvant therapy (HR=1.22, 95% CI 1.14, 1.31, Z=4.286, P<0.001). In the random effects model, neoadjuvant therapy was associated with skeletal muscle loss, with a standardized mean difference of -0.20 (95% CI -0.31, -0.09, Z=3.49, P<0.001) and a significant heterogeneity (I2 =62.2%, P<0.001). Multiple meta regression indicated that population, neoadjuvant therapy type, and measuring tool were the potential sources of heterogeneity. The funnel plot revealed that there was no high publication bias in these studies (Begg’s test, P=0.544) and the sensitivity analysis showed stable results when separately excluding studies. For the postoperative outcomes, the results revealed that muscle loss during neoadjuvant therapy was significantly related to overall survival (HR=2,08, 95% CI =1.47, 2.95, Z=4.12, P<0.001, I2 = 0.0%), but not related to disease-free survival and other short-term outcomes.ConclusionsThis systematic review and meta-analysis revealed that skeletal muscle decreased significantly during neoadjuvant therapy in patients with gastrointestinal cancers and skeletal muscle loss was strongly associated with worse overall survival. More high-quality studies are needed to update and valid these conclusions in a more specific or stratified way.Systematic Review Registration[https://www.crd.york.ac.uk/PROSPERO/], identifier PROSPERO (CRD42021292118)
Collapse
Affiliation(s)
- Xin-Yi Xu
- Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Xiao-Man Jiang
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Qin Xu
- School of Nursing, Nanjing Medical University, Nanjing, China
- *Correspondence: Qin Xu,
| | - Hao Xu
- Department of Gastric Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin-Hua Luo
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cui Yao
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ling-Yu Ding
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Shu-Qin Zhu
- School of Nursing, Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Martin P. Cancer Cachexia Syndrome: Reflecting on 20 years of providing cancer cachexia care as the leader of an interdisciplinary team in an Australian Cancer Centre. Asia Pac J Oncol Nurs 2022; 9:100070. [PMID: 35637652 PMCID: PMC9142557 DOI: 10.1016/j.apjon.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/28/2022] Open
|
22
|
Li S, Qiu R, Yuan G, Wang Q, Li Z, Li Q, Zhang N. Body composition in relation to postoperative anastomotic leakage and overall survival in patients with esophageal cancer. Nutrition 2021; 94:111534. [PMID: 34952360 DOI: 10.1016/j.nut.2021.111534] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Body composition was reported to be related to the prognosis of patients with cancer. This study aimed to investigate the influence of preoperative body composition on anastomotic leakage and overall survival in patients with esophageal cancer. METHODS In this retrospective study, 93 patients with esophageal cancers were evaluated. Skeletal muscle area, intermuscular adipose tissue, visceral adipose tissue (VAT), and subcutaneous adipose tissue were measured on computed tomography images at the level of the third lumbar vertebra. Subsequently, each body composition index was also calculated by dividing the body composition by the square of the height. The cut-off values of body compositions were defined using X-tile software (version 3.6.1; Yale University, New Haven, CTA). Univariate and multivariate analyses were performed to evaluate the risk factors of anastomotic leakage. Kaplan-Meier method and Cox regression analysis were used to evaluate the risk factors of overall survival. RESULTS VAT and visceral fat index (VFI) were higher in patients with anastomotic fistula than in those without anastomotic fistula, but none of them were independent risk factors. Patients with higher body mass index (BMI), higher VFI, and higher subcutaneous fat index (SFI) had better overall survival. By multivariate analysis, SFI >27.6 cm2/m2 was still significantly associated with overall survival. CONCLUSION Patients with higher VAT and VFI were prone to have an anastomotic leakage. Lower BMI, VFI, and SFI were associated with a reduction in overall survival.
Collapse
Affiliation(s)
- Shichao Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruixing Qiu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guanjie Yuan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Wang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Ni Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
23
|
Zhang FM, Zhang XZ, Zhu GL, Lv LQ, Yan XL, Wu WX, Wang SL, Chen XL, Zhuang CL, Yu Z. Impact of sarcopenia on clinical outcomes of patients with stage I gastric cancer after radical gastrectomy: A prospective cohort study. Eur J Surg Oncol 2021; 48:541-547. [PMID: 34420825 DOI: 10.1016/j.ejso.2021.08.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/27/2021] [Accepted: 08/15/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The relationships between sarcopenia and postoperative outcomes in patients with early-stage gastric cancer who undergo radical gastrectomy is unclear. We aimed to investigate the predictive value of sarcopenia on adverse outcomes for stage I gastric cancer. METHODS The clinical data of patients who underwent radical gastrectomy for stage I gastric cancer between July 2013 and May 2019 were prospectively collected. Basic sarcopenia components were measured preoperatively. Univariate and multivariate analyses were conducted to evaluate the risk factors for short- and long-term outcomes. RESULTS A total of 507 patients with early-stage gastric cancer were included in the study, and 73 (14.4%) patients were diagnosed as sarcopenia. Patients with sarcopenia had significantly higher incidence of postoperative complications (32.9% vs. 17.5%, P = 0.002), longer postoperative hospital stays (13 days vs. 12 days, P < 0.001), higher hospitalization costs (65210 yuan vs. 55197 yuan, P < 0.001) and one-year mortality (8.2% vs. 1.8%, P = 0.002). During the median follow-up time of 38.8 months, 12 (16.4%) patients dead in the sarcopenic group and 25 (5.8%) patients dead in the non-sarcopenic group. Sarcopenia was an independent risk factor for both short- and long-term clinical outcomes. Moreover, we found that low muscle quantity and low handgrip strength mediated the adverse impacts of sarcopenia on postoperative complications while low muscle quality mediated the adverse impacts of sarcopenia on overall survival. CONCLUSION Sarcopenia was strongly associated with worse short- and long-term clinical outcomes in patients with stage I gastric cancer who undergo radical gastrectomy.
Collapse
Affiliation(s)
- Feng-Min Zhang
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xian-Zhong Zhang
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guang-Lou Zhu
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lu-Qing Lv
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xia-Lin Yan
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Xue Wu
- Department of General Surgery, Shanghai Tenth People's Hospital Chongming Branch Affiliated to Tongji University, Shanghai, China
| | - Su-Lin Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao-Lei Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cheng-Le Zhuang
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Zhen Yu
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
24
|
Meyer HJ, Wienke A, Surov A. Computed tomography-defined low skeletal muscle mass as a prognostic marker for short-term mortality in critically ill patients: A systematic review and meta-analysis. Nutrition 2021; 91-92:111417. [PMID: 34399402 DOI: 10.1016/j.nut.2021.111417] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/28/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Low skeletal muscle mass (LSMM) can be assessed by imaging modalities and is associated with several clinically relevant factors in critically ill patients. Our aim was to establish the effect of computed tomography (CT)-defined LSMM on short-term mortality in critically ill patients based on a large patient sample. METHODS The MedLine library and the Cochrane and SCOPUS databases were screened for associations between CT-defined LSMM and short-term mortality in critically ill patients up to May 2021. The primary endpoint of the systematic review was the odds ratio of sarcopenia on mortality. In total, nine studies were selected as suitable for the analysis and included into the present analysis. RESULTS The studies included a total of 1563 critically ill patients with different underlying diagnoses. The pooled overall prevalence of LSMM was 50.9%. The pooled odds ratio for the effect of sarcopenia on short-term mortality was 2.78 (95% confidence interval, 2.05-3.75). CONCLUSIONS CT-defined LSMM is highly prevalent in critically ill patients, has a relevant effect on short-term mortality, and should be included as a relevant prognostic biomarker in clinical routines.
Collapse
Affiliation(s)
- Hans-Jonas Meyer
- Department of Diagnostic and Interventional Radiology, University of Leipzig, Leipzig, Germany.
| | - Andreas Wienke
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Alexey Surov
- Department of Radiology and Nuclear Medicine, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|