1
|
Zhang ZQ, Zhong Q, Sun YQ, Ma YB, Ding FH, Wu D, Xue MQ, Desiderio J, Yu JH, Wu J, Zheng CH, Li P, Chen QY, Huang CM, Xie JW. The minimum number of lymph node retrieval in gastric cancer patients after neoadjuvant therapy: An international multi-institute cohort study. Surgery 2025; 183:109373. [PMID: 40328160 DOI: 10.1016/j.surg.2025.109373] [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: 09/23/2024] [Revised: 02/22/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND The minimum number of retrieved lymph nodes for radical gastrectomy after neoadjuvant therapy remains controversial. The objective of this study was to determine the minimum threshold for retrieved lymph nodes in patients with gastric cancer after neoadjuvant therapy to accurately evaluate staging and prognosis. METHODS Multivariate models were employed to investigate the correlation between the number of retrieved lymph nodes and survival outcomes and stage migration. The hazard ratio curves for each retrieved lymph node count compared with 1 retrieved lymph node (as a reference), including overall survival and disease-specific survival, were fitted using a LOWESS smoother, and the structural break points were determined by Chow test. RESULTS This international multicenter study analyzed the clinicopathological data of 2,490 patients with gastric cancer who underwent gastrectomy after neoadjuvant therapy. The median follow-up period for the overall population was 90.0 months, with a median of 22 retrieved lymph nodes. The study demonstrated that a greater number of retrieved lymph nodes was linked to a greater probability of detecting positive lymph nodes (odds ratio, 1.005; P = .030) as well as improved overall survival and disease-specific survival (overall survival: hazard ratio, 0.988; P < .001 and disease-specific survival: hazard ratio, 0.987; P < .001). The cutoff point analysis identified 24 as the minimum number of retrieved lymph nodes. The 5-year overall survival rate was significantly greater in the group with ≥24 retrieved lymph nodes (46.0%) compared with the group with 16-23 retrieved lymph nodes (35.3%) and the group with <16 retrieved lymph nodes group (29.3%) (P < .001). Similar trends were observed with regard to disease-specific survival. The time-dependent area under the curve analysis revealed that the group with ≥24 retrieved lymph nodes exhibited superior predictive performance for overall survival and disease-specific survival compared to the group with <24 retrieved lymph nodes, on the basis of ypN staging (overall survival: time-dependent areas under the receiver operating characteristic curve≥24, 0.71 vs time-dependent areas under the receiver operating characteristic curve<24, 0.67; disease-specific survival: time-dependent areas under the receiver operating characteristic curve≥24, 0.72 vs time-dependent areas under the receiver operating characteristic curve<24, 0.68). CONCLUSION The minimum number of retrieved lymph nodes for evaluation prognosis and reducing stage migration in patients with gastric cancer who undergo radical gastrectomy after neoadjuvant therapy was 24.
Collapse
Affiliation(s)
- Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Yu-Qin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China; Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Yu-Bin Ma
- Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Fang-Hui Ding
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, China
| | - Dong Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Meng-Qi Xue
- Public Health School, Fujian Medical University, Fuzhou, Fujian, China
| | - Jacopo Desiderio
- Department of Digestive Surgery, St. Mary's Hospital, University of Perugia, Terni, Italy
| | - Jun-Hua Yu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China; Department of General Surgery, The Quzhou Affiliated Hospital of Wenzhou University, Quzhou, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China; Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
| |
Collapse
|
2
|
Jeong JY, Yoon JK, Hwang J, Park SH, Cho M, Kim YM, Kim HI, Kim H, Hyung WJ. Diagnostic performance of fluorescent lymphography-guided lymph node dissection during minimally invasive gastrectomy following chemotherapy. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109738. [PMID: 40048959 DOI: 10.1016/j.ejso.2025.109738] [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: 02/23/2025] [Accepted: 03/01/2025] [Indexed: 05/26/2025]
Abstract
INTRODUCTION Fluorescent lymphography-guided lymph node dissection (FL) using indocyanine green (ICG) during radical gastrectomy for gastric cancer has shown enhanced lymph node (LN) retrieval and high sensitivity in detecting LN metastases. However, the impact of FL during gastrectomy following chemotherapy remains uncertain because changes in the ICG injection site due to tumor shrinkage may potentially visualize different lymphatic drainage from the tumor. This study aimed to assess the diagnostic performance of FL during gastrectomy after preoperative chemotherapy. MATERIALS AND METHODS This retrospective study included patients who underwent minimally invasive gastrectomy with FL following chemotherapy between January 2013 and February 2024. Patients were categorized according to their tumor response after chemotherapy based on endoscopic, radiologic, and pathological findings. RESULTS Of 29 patients, 9.4 (range 8-12) LN stations containing 6.9 (range 3-11) fluorescent LN stations, which had 56.3 (range 33-99) LNs including 33.4 (range 11-68) fluorescent LNs, were retrieved per patient. While 52 metastatic LN stations were fluorescent, three non-fluorescent metastatic LN stations were identified in one patient (3.4 %). FL showed 94.5 % (52/55) sensitivity and 95.9 % (70/73) negative predictive value for detecting metastatic LN stations. There was no significant difference in the number of retrieved LNs and the sensitivity for detecting metastatic LN stations between responders and non-responders. CONCLUSION Tumor response after chemotherapy did not influence the diagnostic performance of FL. The diagnostic performance of FL during gastrectomy following chemotherapy was acceptable. Similar to upfront surgery, FL can be safely applied even after chemotherapy.
Collapse
Affiliation(s)
- Ji Yoon Jeong
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea; Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Ja Kyung Yoon
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jawon Hwang
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea; Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Sung Hyun Park
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea; Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Minah Cho
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea; Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Yoo Min Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea; Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Hyoung-Il Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea; Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woo Jin Hyung
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea; Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea; Department of Faculty Surgery No. 1, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
3
|
Zhang H, Lin JP, Chen XF, Wang F. The optimal number of lymph node dissections in three-field lymphadenectomy for esophageal squamous cell carcinoma: a large retrospective study. Langenbecks Arch Surg 2025; 410:121. [PMID: 40186668 PMCID: PMC11972225 DOI: 10.1007/s00423-025-03686-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/19/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Currently, there is no consensus on the optimal number of lymph node dissections (LNDs) in three-field lymphadenectomy for esophageal squamous cell carcinoma (ESCC). This study aimed to explore the relationship between the LND count and overall survival (OS) in ESCC patients to determine the optimal number of LNDs that confer a survival benefit. METHODS A retrospective analysis was conducted on ESCC patients who underwent three-field lymphadenectomy at Fujian Cancer Hospital from February 2004 to January 2018. The optimal LND number was determined using X-Tile software. Kaplan‒Meier survival curves and Cox regression analyses were used to evaluate the relationship between LND count and OS. RESULTS A total of 1053 ESCC patients who underwent three-field lymphadenectomy were included in this study (median age 58 years [IQR: 52-65], 781 males [74.2%]). Using X-Tile software, 27 was identified as the optimal cutoff value for the number of LNDs. The 5-year OS for the > 27 LNDs group was significantly better than that for the ≤ 27 LNDs group (67.8% vs. 59.8%, P = 0.042). Multivariate Cox regression analysis confirmed that LND count (≤ 27 and > 27) was an independent protective factor for OS (HR = 0.724; P = 0.004). Stratified analysis on the basis of TNM stage revealed that in patients with T3-4N0M0 disease (HR = 0.412; P = 0.001) and T1-2 N + M0 disease (HR = 0.503; P = 0.025), a greater number of dissected lymph nodes was closely associated with a survival benefit. CONCLUSION For ESCC patients undergoing three-field lymphadenectomy, dissecting more than 27 lymph nodes is associated with better prognosis, especially for patients with T3-4N0M0 and T1-2 N + M0 stages.
Collapse
Affiliation(s)
- Huaiyuan Zhang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jun-Peng Lin
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xiao-Feng Chen
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Feng Wang
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
- Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, China.
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, China.
| |
Collapse
|
4
|
Zhang H, Sun N, Li F, Wang Q, Sun Z, Zhang Y, Wang L, Zhao C, Fu Y. Construction of a modified TNM staging system and prediction model based on examined lymph node counts for gastric cancer patients at pathological stage N3. Front Oncol 2025; 15:1569736. [PMID: 40248208 PMCID: PMC12003143 DOI: 10.3389/fonc.2025.1569736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/13/2025] [Indexed: 04/19/2025] Open
Abstract
Background Examined lymph node (ELN) count is a critical factor affecting the number of metastatic lymph nodes (MLNs). The impact of the ELN number on survival and staging remains unclear. Methods This study included 4,291 stage N3 GC patients from the SEER database (training cohort) and 567 stage N3 GC patients from the FAHZZU database (validation cohort). The optimal ELN count and stage migration were investigated, and a modified TNM (mTNM) staging system including the ELN count was proposed. LASSO regression and random forest analyses were used to screen and evaluate the variables associated with survival, and an mTNM-based nomogram was constructed. The performance of the mTNM staging system and mTNM-based nomogram were compared with that of the 8th edition of the TNM staging system. Results The optimal threshold of the ELN count was identified as 21. An insufficient number of ELNs (≤ 21) was associated with poorer survival outcomes and led to stage migration in all N3 patients. A new mTNM staging system was proposed, integrating the ELN count into the TNM staging system (8th edition). LASSO regression analysis revealed that age, tumor size, adjuvant chemotherapy, adjuvant radiotherapy, and the mTNM system were associated with overall survival (OS) outcomes, and random forest analysis revealed that the mTNM system was the most important variable for predicting survival. An mTNM-based nomogram was constructed to predict 1-, 3-, and 5-year OS rates. Compared with the TNM staging system (8th edition), the mTNM staging system and mTNM-based nomogram showed superior prognosis discriminative ability, better predictive accuracy, and greater net improvement in survival outcomes. Conclusions The optimal ELN count for N3 GC patients was 21. The mTNM staging system and mTNM-based nomogram showed superior discriminative ability, predictive accuracy, and greater net benefit for OS outcomes.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nan Sun
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feng Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiyang Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhao Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yawei Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunlin Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Zhang XQ, Huang ZN, Wu J, Liu XD, Xie RZ, Wu YX, Zheng CY, Zheng CH, Li P, Xie JW, Wang JB, He QC, Qiu WW, Tang YH, Zhang HX, Zhou YB, Lin JX, Huang CM. Machine Learning Prediction of Early Recurrence in Gastric Cancer: A Nationwide Real-World Study. Ann Surg Oncol 2025; 32:2637-2650. [PMID: 39738899 DOI: 10.1245/s10434-024-16701-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 11/28/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Patients with gastric cancer (GC) who experience early recurrence (ER) within 2 years postoperatively have poor prognoses. This study aimed to analyze and predict ER after curative surgery for patients with GC using machine learning (ML) methods. PATIENTS AND METHODS This multicenter population-based cohort study included data from ten large tertiary regional medical centers in China. The clinical, pathological, and laboratory parameters were retrospectively collected from the records of 11,615 patients. The patients were randomly divided into training (70%) and test (30%) cohorts. A total of ten ML models were developed and validated to predict the ER. Model performance was evaluated using area under the receiver operating characteristic curve (AUC), calibration plots, and Brier score (BS). SHapley Additive exPlanations (SHAP) was used to rank the input features and interpret predictions. RESULTS ER was reported in 1794 patients (15%) during follow-up. The stacking ensemble model achieved AUCs of 1.0 and 0.8 in the training and testing cohorts, respectively, with a BS of 0.113. SHAP dependency plots revealed that tumor staging, elevated tumor marker levels, lymphovascular invasion, perineural invasion, and tumor size > 5 cm were associated with higher ER risk. The impact of age and the number of lymph nodes harvested on ER risk exhibited a "U-shaped distribution." Additionally, an online prediction tool based on the best model was developed to facilitate clinical applications. CONCLUSIONS We developed a robust clinical model for predicting the risk of ER after surgery for GC, which may aid in individualized clinical decision-making.
Collapse
Affiliation(s)
- Xing-Qi Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Xiao-Dong Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Rong-Zhen Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Ying-Xin Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Section for Gastrointestinal Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University and The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| | - Chang-Yue Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Putian University, Putian, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qi-Chen He
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Wen-Wu Qiu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Yi-Hui Tang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Hao-Xiang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Yan-Bing Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
6
|
Calì M, Aiolfi A, Sato S, Hwang J, Bonitta G, Albanesi F, Bonavina G, Cavalli M, Campanelli G, Biondi A, Bonavina L, Bona D. Effect of Indocyanine Green-Guided Lymphadenectomy During Gastrectomy on Survival: Individual Patient Data Meta-Analysis. Cancers (Basel) 2025; 17:980. [PMID: 40149314 PMCID: PMC11940200 DOI: 10.3390/cancers17060980] [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: 12/29/2024] [Revised: 02/14/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Indocyanine green-guided (ICG-guided) lymphadenectomy during gastrectomy for cancer has been proposed to enhance the accuracy of lymphadenectomy. The impact of ICG-guided lymphadenectomy on patient survival remains debated. METHODS The findings of the systematic review were reconstructed into an individual patient data (IDP) meta-analysis with restricted mean survival time difference (RMSTD). Overall survival (OS) and disease-free (DFS) survival were primary outcomes. RMSTD, standardized mead difference (SMD), and 95% confidence intervals (CI) were used as pooled effect size measures. RESULTS Three studies (6325 patients) were included; 42% of patients underwent ICG-guided lymphadenectomy. The patients' age ranged from 47 to 72 years and 58% were males. Proximal, distal, and total gastrectomy were completed in 6.8%, 80.4%, and 12.8% of patients, respectively. The surgical approach was laparoscopic (62.3%) and robotic (37.7%). ICG-guided lymphadenectomy was associated with a higher number of harvested lymph nodes compared to non-ICG-guided lymphadenectomy (SMD 0.50; 95% CI 0.45-0.55). At the 42-month follow-up, OS and DFS estimates for ICG-guided vs. non-ICG-guided lymphadenectomy were 0.5 months (95% CI -0.01, 1.1) and 1.3 months (95% CI 0.39, 2.15), respectively. CONCLUSIONS Our analysis suggests that ICG-guided lymphadenectomy offers equivalent long-term OS and DFS compared to non-ICG-guided lymphadenectomy.
Collapse
Affiliation(s)
- Matteo Calì
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, 20122 Milan, Italy; (M.C.); (M.C.); (G.C.); (D.B.)
| | - Alberto Aiolfi
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, 20122 Milan, Italy; (M.C.); (M.C.); (G.C.); (D.B.)
| | - Sho Sato
- Department of Surgery, Minimally Invasive UGI Surgery and Oncology, Yokohama City University Gastroenterological Center, Yokohama 232-0024, Japan;
| | - Jawon Hwang
- Minimally Invasive UGI Surgery, Severance Hospital–Division of General Surgery, Seoul 03722, Republic of Korea;
| | - Gianluca Bonitta
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, 20122 Milan, Italy; (M.C.); (M.C.); (G.C.); (D.B.)
| | - Francesca Albanesi
- Department of Oncologic Surgery 1–HPB, Division of General Surgery, Fondazione I.R.C.C.S Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Giulia Bonavina
- Department of Obstetrics and Gynecology, IRCCS MultiMedica, 20138 Milan, Italy;
| | - Marta Cavalli
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, 20122 Milan, Italy; (M.C.); (M.C.); (G.C.); (D.B.)
| | - Giampiero Campanelli
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, 20122 Milan, Italy; (M.C.); (M.C.); (G.C.); (D.B.)
| | - Antonio Biondi
- G. Rodolico Hospital, Surgical Division, Department of General Surgery and Medical Surgical Specialties, University of Catania, 95131 Catania, Italy;
| | - Luigi Bonavina
- IRCCS Policlinico San Donato, Division of General and Foregut Surgery, Department of Biomedical Sciences for Health, University of Milan, 20097 Milan, Italy;
| | - Davide Bona
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, 20122 Milan, Italy; (M.C.); (M.C.); (G.C.); (D.B.)
| |
Collapse
|
7
|
Dehal A, Woo Y, Glazer ES, Davis JL, Strong VE. D2 Lymphadenectomy for Gastric Cancer: Advancements and Technical Considerations. Ann Surg Oncol 2025; 32:2129-2140. [PMID: 39589578 DOI: 10.1245/s10434-024-16545-6] [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: 09/09/2024] [Accepted: 11/05/2024] [Indexed: 11/27/2024]
Abstract
Lymphadenectomy (LND) is a crucial component of the curative surgical treatment of gastric cancer (GC). The LND serves to both accurately stage the disease and offer therapeutic benefits. At the time of "curative-intent" gastrectomy, D2 LND is the optimal treatment for patients with locally advanced GC due to its survival benefits and acceptable morbidity. Mastery of the technical aspects of LND, especially D2, requires significant training, adequate case volume, and expertise. This review discusses key aspects of D2 LND, including its status as the standard treatment for locally advanced GC, definition and anatomic borders, technical details, and controversial topics such as splenic hilar dissection and omentectomy. The application of indocyanine green (ICG) fluorescence imaging to elucidate the drainage patterns of GC and to facilitate lymph node (LN) identification is briefly reviewed. Finally, GC standardization and centralization, including surgical treatment, are discussed.
Collapse
Affiliation(s)
- Ahmed Dehal
- Department of General Surgery, Southern California Permanente Medical Group, Department of Clinical Sciences, Kaiser Permanente School of Medicine, Los Angeles, CA, USA.
| | - Yanghee Woo
- Division of Surgical Oncology, Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Evan S Glazer
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jeremey L Davis
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vivian E Strong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
8
|
Tang Y, Huang Z, Zhang X, Li P, Xie J, Wang J, Chen Q, Cao L, Lin M, Tu R, Lin G, Zheng H, Zhong Q, Lin J, Yao Z, Wu D, Zheng C, Lin J, Huang C. Effect of visceral obesity on outcomes of fluorescence-guided lymphadenectomy during laparoscopic gastrectomy for gastric cancer: Post hoc analysis of a randomized phase 3 trial. Chin J Cancer Res 2024; 36:503-516. [PMID: 39539814 PMCID: PMC11555203 DOI: 10.21147/j.issn.1000-9604.2024.05.04] [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: 06/28/2024] [Accepted: 09/08/2024] [Indexed: 11/16/2024] Open
Abstract
Objective To explore the impact of visceral fat area (VFA) on the short- and long-term efficacy of indocyanine green (ICG)-guided D2 lymphadenectomy for gastric cancer (GC). Methods A post hoc analysis was performed in patients who participated in a phase 3 randomized clinical trial of ICG-guided laparoscopic radical gastrectomy vs. conventional laparoscopic radical gastrectomy from November 2018 to July 2019. The VFA was calculated based on preoperative computed tomography images. Short-term efficacy included the quality of lymph node (LN) dissection and surgical outcomes, while long-term efficacy included overall survival (OS) and recurrence-free survival (RFS). Results This study included 126 patients each in the ICG (high-VFA, n=43) and non-ICG groups (high-VFA, n=38). Compared with the non-ICG group, the ICG group had significantly more retrieved LNs (low-VFA: 50.1 vs. 43.9, P=0.001; high-VFA: 49.6 vs. 37.5, P<0.001) and a significantly lower LN noncompliance rate (low-VFA: 32.5% vs. 50.0%, P=0.020; high-VFA: 32.6% vs. 73.7%, P<0.001), regardless of the VFA. The ICG group had a shorter postoperative hospital stay and fewer intra-abdominal infections than the ICG group in the high-VFA patients (P=0.025 and P=0.020, respectively) but not in the low-VFA patients. Regardless of the VFA, the 3-year OS (RFS) was better in the ICG group than in the non-ICG group [low-VFA: 83.1% (76.9%) vs. 73.9% (67.0%); high-VFA: 90.7% (90.7%) vs. 73.7% (73.5%); P for interaction =0.474 (0.547)]. Conclusions The short- and long-term efficacies of ICG tracing were not influenced by visceral obesity.
Collapse
Affiliation(s)
- Yihui Tang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zening Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Xingqi Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Jianwei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Jiabin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Qiyue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Longlong Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Ruhong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Guangtan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Hualong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Juli Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Zihao Yao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Dong Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Chaohui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Jianxian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| | - Changming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou 350001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou 350108, China
| |
Collapse
|
9
|
Batra S, Bhandare MS, Chaudhari V, Esha P, Nikhil M, Ostwal V, Ramaswamy A, Ramadwar M, Kulkarni S, Shrikhande SV. 1657 Resected Gastric Adenocarcinomas at a Single Institution: Outcomes and Trends over 17 Years. Ann Surg Oncol 2024; 31:7142-7156. [PMID: 39014164 DOI: 10.1245/s10434-024-15842-4] [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: 02/09/2024] [Accepted: 07/02/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Outside of clinical trials, real-world data of advanced gastric cancers (AGCs) managed with perioperative or adjuvant chemotherapy with a backbone of D2 lymphadenectomy is limited. PATIENTS AND METHODS Curative resections for gastric adenocarcinoma between January 2003 and January 2020 at the Tata Memorial Centre were analyzed, comparing three time periods marking major increments in annual gastric resections (GRs). RESULTS 1657 radical gastric resections were performed with a morbidity and mortality rate of 34.9% and 1.4%, respectively. Over three consecutive periods, the number of annual GRs increased from 56/year to 97/year to 156/year (P < 0.001) with a significant escalation in surgical magnitude and complexity. Improvement in surgical quality indicators (median lymph node yield from 15 to 25, P < 0.001 and margin negativity from 8.2 to 5.5%, P = 0.002) was observed with no corresponding increase in severe complications (6.9%) or mortality (1.4%). The proportion of distal and signet ring cancers was found to decrease over time, with an increase in proximal cancers and younger age at presentation. Overall, 90% of GRs were for AGCs with a median overall survival (OS) of 4.4 years (± 6 months), and 5-year OS rate of 47.6% (± 1.9%). CONCLUSIONS Change in pattern of tumor characteristics was observed. Aggressive treatment options for AGC were employed progressively with excellent survival. With increase in volumes, improvements in surgical quality indicators, and a relative improvement in postoperative mortality was observed. These results provide a roadmap for developing dedicated gastric cancer centers.
Collapse
Affiliation(s)
- Swati Batra
- Department of GI and HPB Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. Ernest Borges Road, Parel, Mumbai, India
| | - Manish S Bhandare
- Department of GI and HPB Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. Ernest Borges Road, Parel, Mumbai, India
| | - Vikram Chaudhari
- Department of GI and HPB Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. Ernest Borges Road, Parel, Mumbai, India
| | - Pai Esha
- Department of GI and HPB Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. Ernest Borges Road, Parel, Mumbai, India
| | - Mehta Nikhil
- Department of GI and HPB Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. Ernest Borges Road, Parel, Mumbai, India
| | - Vikas Ostwal
- Department of GI and HPB Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. Ernest Borges Road, Parel, Mumbai, India
| | - Anant Ramaswamy
- Department of GI and HPB Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. Ernest Borges Road, Parel, Mumbai, India
| | - Mukta Ramadwar
- Department of Oncopathology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. Ernest Borges Road, Parel, Mumbai, India
| | - Suyash Kulkarni
- Department of Radiology and Intervention Radiology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. Ernest Borges Road, Parel, Mumbai, India
| | - Shailesh Vinayak Shrikhande
- Department of GI and HPB Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr. Ernest Borges Road, Parel, Mumbai, India.
| |
Collapse
|
10
|
Liu Y, Zhou C, Gan L, Zhang Q, Chang J, Wang X, Wang X, Xu M, Chen J, Sheng W, Liu F. Finding the minimum number of retrieved lymph nodes and negative lymph nodes in gastric cancer surgery: a real-world study. Int J Surg 2024; 110:5574-5584. [PMID: 38752518 PMCID: PMC11392185 DOI: 10.1097/js9.0000000000001626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/07/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Lymph node retrieval deficiency can lead to understagement and postoperative cancer recurrence, it is crucial to establish the standard number of retrieved lymph nodes (rLNs) and negative lymph nodes (nLNs) for patients undergoing gastrectomy. METHODS Patients who has gastric adenocarcinoma and underwent either radical subtotal gastrectomy (RSG) or radical total gastrectomy (RTG) between 2000 and 2022 were retrospectively included. The authors utilized restricted cubic spline (RCS) analysis to determine the ideal threshold for rLNs and nLNs. Survival analysis was conducted using Kaplan-Meier (KM) curves, log-rank tests and forest plots. Propensity score matching (PSM) was utilized to balance parameters between two groups. The median follow-up time for this study was 3095 days. RESULTS Our study found that there are significant tumor characteristic differences between RSG and RTG. For patients with N0-N3a stage undergoing RSG, retrieving greater than or equal to 24 lymph nodes intraoperatively were associated with better prognosis both before and after PSM [overall survival (OS): P <0.001, P =0.019]; whereas for N3b stage, at least 32 rLNs were required (OS: P =0.006, P =0.023). Similarly, for patients with N0-N3a stage undergoing RTG, retrieving greater than or equal to 27 lymph nodes intraoperatively were associated with better prognosis both before and after PSM (OS: P <0.001, P =0.047); whereas for N3b stage, at least 34 rLNs were required (OS: P <0.001, P =0.003). Additionally, for patients undergoing RSG, having greater than or equal to 21 nLNs (OS: P <0.001, P =0.013), and for those undergoing RTG, having greater than or equal to 22 nLNs (OS: P <0.001, P <0.001), were also associated with better prognosis both before and after PSM. CONCLUSIONS For patients receiving RSG, rLNs should reach 24 when lymph nodes are limited, and 32 when lymph node metastasis is more extensive, with a minimum number of nLNs ideally reaching 21. Similarly, for patients receiving RTG, rLNs should reach 27 when lymph nodes are limited, 34 when lymph node metastasis is more extensive, and a minimum number of nLNs ideally reaching 22.
Collapse
Affiliation(s)
- Yingxue Liu
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Medical Oncology, Shanghai Medical College, Fudan University
- Institute of Pathology, Fudan University
| | - Changming Zhou
- Department of Cancer Prevention, Clinical Statistics Center, Fudan University Shanghai Cancer Center
- Department of Medical Oncology, Shanghai Medical College, Fudan University
| | - Lu Gan
- Department of Medical Oncology, Fudan University Zhongshan Hospital, Shanghai, China
| | | | - Jinjia Chang
- Department ofMedical Oncology, Fudan University Shanghai Cancer Center
| | - Xin Wang
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Medical Oncology, Shanghai Medical College, Fudan University
- Institute of Pathology, Fudan University
| | - Xu Wang
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Medical Oncology, Shanghai Medical College, Fudan University
- Institute of Pathology, Fudan University
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Medical Oncology, Shanghai Medical College, Fudan University
- Institute of Pathology, Fudan University
| | - Jie Chen
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center
- Department of Medical Oncology, Shanghai Medical College, Fudan University
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Medical Oncology, Shanghai Medical College, Fudan University
- Institute of Pathology, Fudan University
| | - Fenglin Liu
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center
- Department of Medical Oncology, Shanghai Medical College, Fudan University
| |
Collapse
|
11
|
Stroobant EE, Strong VE. Advances in Gastric Cancer Surgical Management. Hematol Oncol Clin North Am 2024; 38:547-557. [PMID: 38402138 DOI: 10.1016/j.hoc.2024.01.003] [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] [Indexed: 02/26/2024]
Abstract
The goal of a gastric cancer operation is a microscopically negative resection margin and D2 lymphadenectomy. Minimally invasive techniques (laparoscopic and robotic) have been proven to be equivalent for oncologic care, yet with faster recovery. Endoscopic mucosal resection can be used for T1a N0 tumor resection. Better understanding of hereditary gastric cancer and molecular subtypes has led to specialized recommendations for MSI-high tumors and patients with pathogenic CDH1 mutations. In the future, surgical management will support minimally invasive approaches and personalized cancer care based on subtype.
Collapse
Affiliation(s)
- Emily E Stroobant
- Gastric and Mixed Tumor Service, Department of Surgery - H1216, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Vivian E Strong
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Weill Cornell Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
12
|
Chan KS, Oo AM. Learning curve of laparoscopic and robotic total gastrectomy: A systematic review and meta-analysis. Surg Today 2024; 54:509-522. [PMID: 36912987 DOI: 10.1007/s00595-023-02672-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/13/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE Minimally-invasive total gastrectomy (MITG) is associated with lower morbidity in comparison to open total gastrectomy but requires a learning curve (LC). We aimed to perform a pooled analysis of the number of cases required to surmount the LC (NLC) in MITG. METHODS A systematic review of PubMed, Embase, Scopus and the Cochrane Library from inception until August 2022 was performed for studies reporting the LC in laparoscopic total gastrectomy (LTG) and/or robotic total gastrectomy (RTG). Poisson mean (95% confidence interval [CI]) was used to determine the NLC. Negative binomial regression was performed as a comparative analysis. RESULTS There were 12 articles with 18 data sets: 12 data sets (n = 1202 patients) on LTG and 6 data sets (n = 318 patients) on RTG. The majority of studies were conducted in East Asia (94.4%). The majority of the data sets (n = 12/18, 66.7%) used non-arbitrary analyses. The NLC was significantly smaller in RTG in comparison to LTG [RTG 20.5 (95% CI 17.0-24.5); LTG 43.9 (95% CI 40.2-47.8); incidence rate ratio 0.47, p < 0.001]. The NLC was comparable between totally-laparoscopic total gastrectomy (TLTG) and laparoscopic-assisted total gastrectomy (LATG) [LATG 39.0 (95% CI 30.8-48.7); TLTG 36.0 (95% CI 30.4-42.4)]. CONCLUSIONS The LC for RTG was significantly shorter for LTG. However existing studies are heterogeneous.
Collapse
Affiliation(s)
- Kai Siang Chan
- Department of General Surgery, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore.
| | - Aung Myint Oo
- Department of General Surgery, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
13
|
Yoo H, Hong KY, Min S, Lee HS, Park DJ, Kim YS, Kong SH, Chang H. Distribution of Perigastric Station 4d Lymph Nodes in Vascularized Gastroepiploic Lymph Node Transfer: An Anatomic Study and Case Series. Ann Surg Oncol 2024; 31:3694-3704. [PMID: 38530528 DOI: 10.1245/s10434-024-15113-2] [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: 12/27/2023] [Accepted: 02/12/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Vascularized gastroepiploic lymph node transfer (VGLNT) is a well-accepted surgical treatment for restoring physiological function in chronic lymphedema. However, the inclusion of substantial lymph nodes (LNs) in the flap remains uncertain. This study aimed to identify the anatomical basis for reliable flap harvest for VGLNT. PATIENTS AND METHODS The anatomy of perigastric station 4d LNs was studied in healthy cadavers (n = 15) and patients with early gastric cancer (EGC) (n = 27). The omentum was divided into three segments: proximal, middle, and distal from the origin of the right gastroepiploic vessels. The flap dimension, number, location, size of LNs, and caliber of the vessels were reviewed. Eight patients underwent VGLNT for upper/lower limb lymphedema. RESULTS The mean numbers of LNs in the proximal, middle, and distal segment were 2.5, 1.4, 0.5 in the cadavers, and 4.9, 2.7, 0.7 in the gastrectomy specimens, respectively. The proximal third included a significantly greater number of LNs than the distal third in the cadaveric (p = 0.024) and ECG (p = 0.016) specimens. A total of 95% of the LNs were located within proximal two-thirds of the flap from the vessel origin both in the cadavers (21.0 × 5.0 cm) and in the gastrectomy specimens (20 × 3.5 cm). In VGLNT, the transferred flap was 25.5 ± 6.9 × 4.1 + 0.7 cm in dimension, containing a mean number of 6.5 ± 1.9 LNs. At postoperative 6 months, the volumetric difference was significantly reduced by 22.8 ± 9.2% (p < 0.001). CONCLUSIONS This study provides a distinct distribution pattern of station 4d LNs. Inclusion of the proximal two-thirds of the flap, which carries majority of the LNs, is recommended for VGLNT.
Collapse
Affiliation(s)
- Hyokyung Yoo
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ki Yong Hong
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sally Min
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Do-Joong Park
- Department of Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yi-Suk Kim
- Department of Anatomy, Catholic Institute for Applied Anatomy, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hak Chang
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Bittar V, Boneli MF, Reis PCA, Felix N, Braga MAP, Rocha KM, Fogaroli LO, Costa GB, Comini AC, Amaral G, Marini DC, Camandaroba MPG. Laparoscopic Versus Open Gastrectomy for Advanced Gastric Cancer: A Meta-Analysis of Randomized Controlled Trials. J Gastrointest Cancer 2024; 55:652-661. [PMID: 38564116 DOI: 10.1007/s12029-024-01048-0] [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] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Laparoscopy-assisted gastrectomy (LAG) is a well-established surgical technique in treating patients with early gastric cancer. However, the efficacy and safety of LAG versus open gastrectomy (OG) in patients with advanced gastric cancer (AGC) remains unclear. METHODS We systematically searched PubMed, Embase, and Cochrane Library in June 2023 for RCTs comparing LAG versus OG in patients with AGC. We pooled risk ratios (RR) and mean differences (MD) with 95% confidence intervals (CI) for binary and continuous endpoints, respectively. We performed all statistical analyses using R software version 4.3.1 and a random-effects model. RESULTS Nine RCTs comprising 3827 patients were included. There were no differences in terms of intraoperative complications (RR 1.14; 95% CI 0.72 to 1.82), number of retrieved lymph nodes (MD -0.54 lymph nodes; 95% CI -1.18 to 0.09), or mortality (RR 0.91; 95% CI 0.30 to 2.83). LAG was associated with a longer operative time (MD 49.28 minutes; 95% CI 30.88 to 67.69), lower intraoperative blood loss (MD -51.24 milliliters; 95% CI -81.41 to -21.06), shorter length of stay (MD -0.83 days; 95% CI -1.60 to -0.06), and higher incidence of pancreatic fistula (RR 2.44; 95% CI 1.08 to 5.50). Postoperatively, LAG was also superior to OG in reducing bleeding rates (RR 0.44; 95% CI 0.22 to 0.86) and time to first flatus (MD -0.27 days; 95% CI -0.47 to -0.07), with comparable results in anastomotic leakage, wound healing issues, major complications, time to ambulation, or time to first liquid intake. In the long-term analyses at 3 and 5 years, there were no significant differences between LAG and OG in terms of overall survival (RR 0.99; 95% CI 0.96 to 1.03) or relapse-free survival (RR 0.99; 95% CI 0.94 to 1.04). CONCLUSION This meta-analysis of RCTs suggests that LAG may be an effective and safe alternative to OG for treating AGC; albeit, it may be associated with an increased risk for pancreatic fistula.
Collapse
Affiliation(s)
- Vinicius Bittar
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil.
| | - Mauricio Ferreira Boneli
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil
| | | | - Nicole Felix
- Universidade Federal de Campina Grande, Campina Grande, Brazil
| | | | - Kian M Rocha
- Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo O Fogaroli
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil
| | - Gamaliel B Costa
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil
| | | | - Gustavo Amaral
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil
| | - Danyelle Cristine Marini
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil
| | | |
Collapse
|
15
|
Kim KY, Hwang J, Park SH, Cho M, Kim YM, Kim HI, Hyung WJ. Superior lymph node harvest by fluorescent lymphography during minimally invasive gastrectomy for gastric cancer patients with high body mass index. Gastric Cancer 2024; 27:622-634. [PMID: 38502275 DOI: 10.1007/s10120-024-01482-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Fluorescent lymphography (FL) using indocyanine green (ICG) allows for the visualization of all draining lymph nodes (LNs), thereby increasing LN retrieval. However, no studies have assessed the efficacy of FL in high body mass index (BMI) gastric cancer patients, even as LN yield decreases with increasing BMI in gastrectomy. This study aimed to investigate the influence of FL on LN retrieval in high BMI gastric cancer patients. METHODS Gastric cancer patients who underwent laparoscopic or robotic gastrectomies from 2013 to 2021 were included. Patients were classified into two groups, with FL (FL group) or without FL (non-FL group). The effect of FL on LN retrieval was assessed by BMI. Inverse probability of treatment weighting (IPTW) was used to ensure comparability between groups. RESULTS Retrieved LN number decreased as BMI increased regardless of FL application (P < 0.001). According to the IPTW analysis, the mean retrieved LN number was significantly higher in the FL group (48.4 ± 18.5) than in the non-FL group (39.8 ± 16.3, P < 0.001), irrespective of BMI. The FL group exhibited a significantly higher proportion of patients with 16 or more LNs (99.5%) than the non-FL group (98.1%, P < 0.001). The FL group also had a significantly higher proportion of patients with 30 or more LNs (86.6%) than the non-FL group (72.2%, P < 0.001). In both the normal and high-BMI patients, the FL group had a significantly larger percentage of patients with a higher nodal classification than the non-FL group. CONCLUSION FL resulted in more LN retrieval, even in high BMI patients. FL ensures accurate staging by maintaining the appropriate retrieved LN number in high BMI gastric cancer patients.
Collapse
Affiliation(s)
- Ki-Yoon Kim
- Department of Surgery, Yonsei University College of Medicine, and Gastric Cancer Center, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Jawon Hwang
- Department of Surgery, Yonsei University College of Medicine, and Gastric Cancer Center, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Sung Hyun Park
- Department of Surgery, Yonsei University College of Medicine, and Gastric Cancer Center, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Minah Cho
- Department of Surgery, Yonsei University College of Medicine, and Gastric Cancer Center, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Yoo Min Kim
- Department of Surgery, Yonsei University College of Medicine, and Gastric Cancer Center, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Hyoung-Il Kim
- Department of Surgery, Yonsei University College of Medicine, and Gastric Cancer Center, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Woo Jin Hyung
- Department of Surgery, Yonsei University College of Medicine, and Gastric Cancer Center, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Manara M, Aiolfi A, Sozzi A, Calì M, Grasso F, Rausa E, Bonitta G, Bonavina L, Bona D. Short-Term Outcomes Analysis Comparing Open, Laparoscopic, Laparoscopic-Assisted, and Robotic Distal Gastrectomy for Locally Advanced Gastric Cancer: A Randomized Trials Network Analysis. Cancers (Basel) 2024; 16:1620. [PMID: 38730574 PMCID: PMC11083793 DOI: 10.3390/cancers16091620] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Minimally invasive surgery for the treatment of locally advanced gastric cancer (AGC) is debated. The aim of this study was to execute a comprehensive assessment of principal surgical treatments for resectable distal gastric cancer. METHODS Systematic review and randomized controlled trials (RCTs) network meta-analysis. Open (Op-DG), laparoscopic-assisted (LapAs-DG), totally laparoscopic (Lap-DG), and robotic distal gastrectomy (Rob-DG) were compared. Pooled effect-size measures were the risk ratio (RR), the weighted mean difference (WMD), and the 95% credible intervals (CrIs). RESULTS Ten RCTs (3823 patients) were included. Overall, 1012 (26.5%) underwent Lap-DG, 902 (23.6%) LapAs-DG, 1768 (46.2%) Op-DG, and 141 (3.7%) Rob-DG. Anastomotic leak, severe complications (Clavien-Dindo > 3), and in-hospital mortality were comparable. No differences were observed for reoperation rate, pulmonary complications, postoperative bleeding requiring transfusion, surgical-site infection, cardiovascular complications, number of harvested lymph nodes, and tumor-free resection margins. Compared to Op-DG, Lap-DG and LapAs-DG showed a significantly reduced intraoperative blood loss with a trend toward shorter time to first flatus and reduced length of stay. CONCLUSIONS LapAs-DG, Lap-DG, and Rob-DG performed in referral centers by dedicated surgeons have comparable short-term outcomes to Op-DG for locally AGC.
Collapse
Affiliation(s)
- Michele Manara
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, Via C. Belgioioso, 173, 20157 Milan, Italy; (M.M.); (D.B.)
| | - Alberto Aiolfi
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, Via C. Belgioioso, 173, 20157 Milan, Italy; (M.M.); (D.B.)
| | - Andrea Sozzi
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, Via C. Belgioioso, 173, 20157 Milan, Italy; (M.M.); (D.B.)
| | - Matteo Calì
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, Via C. Belgioioso, 173, 20157 Milan, Italy; (M.M.); (D.B.)
| | - Federica Grasso
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, Via C. Belgioioso, 173, 20157 Milan, Italy; (M.M.); (D.B.)
| | - Emanuele Rausa
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, Via C. Belgioioso, 173, 20157 Milan, Italy; (M.M.); (D.B.)
| | - Gianluca Bonitta
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, Via C. Belgioioso, 173, 20157 Milan, Italy; (M.M.); (D.B.)
| | - Luigi Bonavina
- Department of Biomedical Sciences for Health, Division of General and Foregut Surgery, IRCCS Policlinico San Donato, University of Milan, 20097 Milan, Italy;
| | - Davide Bona
- I.R.C.C.S. Ospedale Galeazzi–Sant’Ambrogio, Division of General Surgery, Department of Biomedical Science for Health, University of Milan, Via C. Belgioioso, 173, 20157 Milan, Italy; (M.M.); (D.B.)
| |
Collapse
|
17
|
Jiang Y, Shao X, Li W, Hu H, Lu Y, Li Y, Tian Y. Impact of Removal of Lymph Nodes on Survival in Stage I-III Gastric Signet-Ring Cell Cancer: The More, the Better? Ann Surg Oncol 2024; 31:783-791. [PMID: 37991582 DOI: 10.1245/s10434-023-14590-1] [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: 10/12/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND There is an ongoing debate over the prognostic value of the number of examined lymph nodes (ELNs) in cases of gastric signet-ring cell cancer (GSRCC). In this study, we sought to evaluate the correlation between the number of ELNs and the prognosis of GSRCC and identify the optimal number of ELNs. METHODS A total of 1020 patients diagnosed with GSRCC between 2011 and 2018 in the National Cancer Center database were identified. Clinicopathological characteristics were retrospectively collected, and optimal cutoff values of ELNs were calculated by using X-tile. The impact of different ELNs on overall survival (OS) was compared by using Kaplan-Meier curves. We used univariate and multivariate Cox and subgroup analyses to explore the relationship between ELNs and OS. Furthermore, nonlinear correlations were investigated by using restricted cubic splines (RCSs). RESULTS X-tile showed that the optimal cutoff value of ELNs was 22. The 5-year OS was higher for patients with ELNs > 22 (vs. ELNs ≤ 22, 66.9% vs. 74.9%, P = 0.026). Multivariate Cox analyses showed that high ELNs were associated with superior OS (hazard ratio = 0.56, 95% confidence interval 0.43-0.74, P < 0.001). In subgroup analyses, the significant association between tumor size > 4 cm, and TNM III stage was still observed. The RCS regression model showed a U-shaped dose-response nonlinear relationship between ELNs and OS; the inflection point, as well as the lowest risk points, corresponded to 44-52 ELNs. CONCLUSIONS A U-shaped, nonlinear correlation with inflection points of 44-52 ELNs between ELNs and prognosis in GSRCC was identified.
Collapse
Affiliation(s)
- Yujuan Jiang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinxin Shao
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weikun Li
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haitao Hu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiming Lu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Li
- Senior Department of General Surgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Yantao Tian
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
18
|
Senent-Boza A, García-Fernández N, Alarcón-Del Agua I, Socas-Macías M, de Jesús-Gil Á, Morales-Conde S. Impact of tumor stage and neoadjuvant chemotherapy in fluorescence-guided lymphadenectomy during laparoscopic gastrectomy for gastric cancer: A propensity score-matched study in a western center. Surgery 2024; 175:380-386. [PMID: 38040597 DOI: 10.1016/j.surg.2023.10.032] [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: 04/12/2023] [Revised: 07/12/2023] [Accepted: 10/25/2023] [Indexed: 12/03/2023]
Abstract
BACKGROUND The use of indocyanine green fluorescence has been shown to be a safe and effective method for improving lymph node retrieval in patients with gastric cancer. However, previous studies have focused on early-stage tumors and/or the Asian population and excluded patients who received neoadjuvant treatment. METHODS In this study, 142 patients with gastric adenocarcinoma underwent laparoscopic gastrectomy at a Spanish hospital between January 2017 and December 2022. Of these, 42 patients received preoperative indocyanine green injection to guide lymphadenectomy. Their outcomes were compared to a retrospective cohort of 42 patients after 1:1 propensity score matching. RESULTS The feasibility of indocyanine green lymphatic mapping was 95.5%. No complications associated with indocyanine green injection were observed. The indocyanine green group had a significantly higher number of retrieved lymph nodes than the non-indocyanine green group (32.67 vs 25.14; P = .013). This statistically significant difference was maintained across subgroups of neoadjuvant treatment, non-obese patients, pT0 to 2 stage, and pN0 stage. In 47.6% of patients from the indocyanine green group, lymphadenectomy was extended outside the standard D2 dissection area based on indocyanine green uptake, but none of the retrieved lymph nodes were metastatic. There were no differences in postoperative complications and length of hospital stay between the 2 groups. CONCLUSION Indocyanine green-guided lymphadenectomy is safe and feasible and increases the number of retrieved lymph nodes compared to conventional lymphatic dissection, as well as in patients receiving neoadjuvant chemotherapy. The use of indocyanine green should be routine if available for guiding lymph node dissection in gastric cancer, regardless of tumor stage or previous neoadjuvant treatment. However, further studies are needed to determine the impact of this technique on disease-free and overall survival.
Collapse
Affiliation(s)
- Ana Senent-Boza
- Unit of Esophagogastric and Bariatric Surgery, Department of General and Digestive Surgery, Virgen del Rocio University Hospital, Sevilla, Spain.
| | - Noelia García-Fernández
- Unit of Esophagogastric and Bariatric Surgery, Department of General and Digestive Surgery, Virgen del Rocio University Hospital, Sevilla, Spain. https://twitter.com/ngarciafdez
| | - Isaías Alarcón-Del Agua
- Unit of Esophagogastric and Bariatric Surgery, Department of General and Digestive Surgery, Virgen del Rocio University Hospital, Sevilla, Spain
| | - María Socas-Macías
- Unit of Esophagogastric and Bariatric Surgery, Department of General and Digestive Surgery, Virgen del Rocio University Hospital, Sevilla, Spain
| | - Ángela de Jesús-Gil
- Unit of Esophagogastric and Bariatric Surgery, Department of General and Digestive Surgery, Virgen del Rocio University Hospital, Sevilla, Spain. https://twitter.com/angeladejesuss
| | - Salvador Morales-Conde
- Unit of Esophagogastric and Bariatric Surgery, Department of General and Digestive Surgery, Virgen del Rocio University Hospital, Sevilla, Spain; Department of Surgery, School of Medicine, University of Sevilla, Spain. https://twitter.com/smoralesconde
| |
Collapse
|
19
|
De Ruvo N, Fenocchi S, Veronesi L, Missori G, Ricciardolo AA, Rossi EG, Sorrentino L, Cautero N, Serra F, Gelmini R. Prognostic factors in gastric cancer patients: a 10-year mono-institutional experience. Front Surg 2024; 11:1321981. [PMID: 38362460 PMCID: PMC10867173 DOI: 10.3389/fsurg.2024.1321981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/11/2024] [Indexed: 02/17/2024] Open
Abstract
Introduction Gastric cancer (GC) is one of the main causes of death from cancer globally. Long-term survival, especially in Western countries, remains dismal, with no significant improvements in recent years. Therefore, precise identification of clinical and pathological risk factors is crucial for prognosis, as it allows a better selection of patients suitable for oncologically radical treatments and contributes to longer survivals. Methods We devised a retrospective observational longitudinal study over 10 years of experience with GC patients operated with curative intent. Results Several factors were thoroughly investigated in a multivariate analysis to look for significance as independent risk factors for disease-free survival. Our results showed that only BMI, pTNM, and lymph node ratio expressed hazard ratios with implications for survival in our series of patients. Discussion Although limited by the retrospective nature of the study, this is one of the few cancer reports from Northern Italy showing results over 10 years, which may in our view, have an impact on decision-making processes for multidisciplinary teams dedicated to the care of gastric cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - F. Serra
- General, Oncological and Emergency Surgery Unit, Department of Surgery, AOU Policlinico di Modena, University of Modena and Reggio Emilia, Modena, Italy
| | | |
Collapse
|
20
|
Liu F, Yang W, He Y, Yang W, Chen L, Xu R, Liu Z, Ke J, Hou B, Zhang L, Lin M, Liang L, Huang Y, Zhang L, Zhang F, Cai F, Xu H, Liu M, Pan Y, Liu Y, He Z, Ke Y. Surgical quality determines the long-term survival superiority of right over left thoracic esophagectomy for localized esophageal squamous cell carcinoma patients: a real-world multicenter study. Int J Surg 2024; 110:675-683. [PMID: 37983771 PMCID: PMC10871567 DOI: 10.1097/js9.0000000000000897] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 10/28/2023] [Indexed: 11/22/2023]
Abstract
OBJECTIVE The objective was to compare the long-term overall survival (OS) of right versus left thoracic esophagectomy, and to evaluate whether surgical quality impacts comparison result. BACKGROUND Controversy regarding the optimal thoracic esophagectomy approach persists for esophageal squamous cell carcinoma (ESCC). No study has assessed the effect of surgical quality in comparison between right and left approaches. METHODS The authors consecutively recruited 5556 operable ESCC patients from two high-volume centers in China, of whom 2220 and 3336 received right and left thoracic esophagectomy, respectively. Cumulative sum was used to evaluate the learning curve for operation time of right approach, as the indicator of surgical proficiency. RESULTS With a median follow-up of 83.1 months, right approach, harvesting more lymph nodes, tended to have a better OS than left approach (Mean: 23.8 vs. 16.7 nodes; adjusted hazard ratio (HR)=0.93, 95% CI: 0.85-1.02). Subset analysis by the extent of lymphadenectomy demonstrated that right approach with adequate lymphadenectomy (≥15 nodes) resulted in statistically significant OS benefit compared with left approach (adjusted HR=0.86, 95% CI: 0.77-0.95), but not with limited lymphadenectomy. Subset analysis by surgical proficiency showed that proficient right approach conferred a better OS than left approach (adjusted HR=0.75, 95% CI: 0.64-0.88), but improficient right approach did not have such survival advantage. CONCLUSIONS Surgical quality plays a crucial role in survival comparison between surgical procedures. Right thoracic esophagectomy performed with adequate lymphadenectomy and surgical proficiency, conferring more favorable survival than left approach, should be recommended as the preferred surgical procedure for localized ESCC.
Collapse
Affiliation(s)
- Fangfang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Wenlei Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yu He
- Chinese Preventive Medicine Association, Beijing, People's Republic of China
- Department of Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Wei Yang
- Cancer Hospital of Shantou University Medical College, Guangdong Province, People's Republic of China
| | - Lei Chen
- Cancer Hospital of Shantou University Medical College, Guangdong Province, People's Republic of China
| | - Ruiping Xu
- Anyang Cancer Hospital, Henan Province, People's Republic of China
| | - Zhen Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Ji Ke
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Bolin Hou
- Linkdoc AI Research (LAIR), Beijing, People's Republic of China
| | - Liqun Zhang
- Cancer Hospital of Shantou University Medical College, Guangdong Province, People's Republic of China
| | - Miaoping Lin
- Cancer Hospital of Shantou University Medical College, Guangdong Province, People's Republic of China
| | - Linlin Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yi Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Lixin Zhang
- Anyang Cancer Hospital, Henan Province, People's Republic of China
| | - Fan Zhang
- Cancer Hospital of Shantou University Medical College, Guangdong Province, People's Republic of China
| | - Fen Cai
- Cancer Hospital of Shantou University Medical College, Guangdong Province, People's Republic of China
| | - Huawen Xu
- Cancer Hospital of Shantou University Medical College, Guangdong Province, People's Republic of China
| | - Mengfei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yaqi Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Ying Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Zhonghu He
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yang Ke
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| |
Collapse
|
21
|
Huang ZN, He QC, Qiu WW, Wu J, Zheng CY, Lin GS, Li P, Wang JB, Lin JX, Lu J, Cao LL, Lin M, Tu RH, Zheng CH, Chen QY, Huang CM, Xie JW. OSATS scoring confirms ICG enhancement of performance in laparoscopic radical gastrectomy: a post-hoc analysis of a randomized controlled trial. Int J Surg 2024; 110:342-352. [PMID: 37939147 PMCID: PMC10793768 DOI: 10.1097/js9.0000000000000830] [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: 06/13/2023] [Accepted: 09/28/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Indocyanine green (ICG) fluorescence imaging is effective in increasing the number of lymph node dissections during laparoscopic radical gastrectomy; however, no studies have attempted to explain this phenomenon. METHODS This study utilized the data from a previous randomized controlled trial (FUGES-012 study) investigating ICG-guided laparoscopic radical gastrectomy performed between November 2018 and July 2019. The Objective Structured Assessments of Technical Skills (OSATS) scoring system was used to grade videos from the ICG and non-ICG groups. Patients with an OSATS score greater than 29 were classified as the high-OSATS population, while those with an OSATS score less than or equal to 29 were classified as the low-OSATS population. RESULTS A total of 258 patients were included in the modified intention-to-treat analysis: 129 in the ICG group and 129 in the non-ICG group. The OSATS score of the ICG group was higher than that of the non-ICG group (29.6±2.6 vs. 26.6±3.6; P <0.001). The ICG group underwent a significantly higher mean total number of lymph node dissections than the non-ICG group (50.5±15.9 vs. 42.0±10.3; adjusted P <0.001). The group assigned to ICG use, better OSATS (high-OSATS) scores were observed, which correlated with greater D2 lymph node retrieval (54.1±15.0 vs. 47.2±8.7; adjusted P =0.039). Finally, the ICG group had a lower rate of lymph node noncompliance than that of the non-ICG group (31.8 vs. 57.4%; P <0.001). CONCLUSIONS By applying the ICG fluorescence navigation technique, better OSATS scores were observed, which correlated with greater lymph node retrieval and a lower lymph node noncompliance rate, as recommended for individualized laparoscopic radical gastrectomy.
Collapse
Affiliation(s)
- Ze-Ning Huang
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Qi-Chen He
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Wen-Wu Qiu
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Ju Wu
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian
| | - Chang-Yue Zheng
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Putian University, Putian, People’s Republic of China
| | - Guo-Sheng Lin
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Ping Li
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Jia-Bin Wang
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Jian-Xian Lin
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Jun Lu
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Long-Long Cao
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Mi Lin
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Ru-Hong Tu
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Chao-Hui Zheng
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Qi-Yue Chen
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Chang-Ming Huang
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| | - Jian-Wei Xie
- Department of Gastric Surgery
- Department of General Surgery, Fujian Medical University Union Hospital
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou
| |
Collapse
|
22
|
Zhang S, Zheng L, Zhang Y, Gao Y, Liu L, Jiang Z, Wang L, Ma Z, Wu J, Chen J, Lu Y, Wang D. A web-based prediction model for long-term cancer-specific survival of middle-aged patients with early-stage gastric cancer: a multi-institutional retrospective study. J Cancer Res Clin Oncol 2023; 149:16551-16561. [PMID: 37712958 DOI: 10.1007/s00432-023-05405-7] [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: 06/18/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND This study constructed and validated a prognostic model to evaluate long-term cancer-specific survival (CSS) in middle-aged patients with early gastric cancer (EGC). METHODS We extracted clinicopathological data from relevant patients between 2004 and 2015 from Surveillance, Epidemiology, and End Results (SEER) database, and randomly divided the patients into a training group (N = 688) and a validation group (N = 292). In addition, 102 Chinese patients were enrolled for external validation. Univariate and multivariate Cox regression models were used to screen for independent prognostic factors, and a nomogram was constructed to predict CSS. We used the concordance index (C-index), calibration curve, receiver operating characteristic (ROC) curve, and decision curve analysis (DCA) to evaluate the predictive performance of the model. RESULTS Univariate and multivariate COX regression analyses showed that tumor location, differentiation grade, N stage, chemotherapy, and number of regional nodes examined were independent risk factors for prognosis, and these factors were used to construct the nomogram. The C-index of the model in the training cohort, internal validation cohort, and external validation cohort was 0.749 (95% CI 0.699-0.798), 0.744 (95% CI 0.671-0.818), and 0.807 (95% CI 0.721-0.893), respectively. The calibration curve showed that the model had an excellent fit. The DCA curve showed that the model had good predictive performance and practical clinical value. CONCLUSION This study developed and validated a new nomogram to predict CSS in middle-aged patients with EGC. The prediction model has unique and practical value and can help doctors carry out individualized treatment and judge prognosis.
Collapse
Affiliation(s)
- Simeng Zhang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Longbo Zheng
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266400, Shandong, China
| | - Yuxia Zhang
- Department of Rehabilitation Pain, Shanghe County People's Hospital, Jinan, Shandong, China
| | - Yuan Gao
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266400, Shandong, China
| | - Lei Liu
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Zinian Jiang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Liang Wang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Zheng Ma
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Jinhui Wu
- Department of Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Jiansheng Chen
- Department of Gastrointestinal Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Yun Lu
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266400, Shandong, China
| | - Dongsheng Wang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China.
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266400, Shandong, China.
| |
Collapse
|
23
|
Mahenthiran AK, Logan CD, Janczewski LM, Valukas C, Warwar S, Silver CM, Feinglass J, Merkow RP, Bentrem DJ, Odell DD. Evaluation of Nationwide Trends in Nodal Sampling Guideline Adherence for Gastric Cancer: 2005-2017. J Surg Res 2023; 291:514-526. [PMID: 37540969 PMCID: PMC10529819 DOI: 10.1016/j.jss.2023.07.011] [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: 03/22/2023] [Revised: 06/15/2023] [Accepted: 07/04/2023] [Indexed: 08/06/2023]
Abstract
INTRODUCTION Surgical resection is the primary curative treatment for localized gastric cancer. A multitude of research supports surgical nodal sampling guidelines. Though there are known disparities in adherence to nodal sampling, it is unclear how hospital program-level disparities have changed over time. The purpose of this study is to evaluate trends in program-level disparities in adherence to gastric cancer nodal sampling guidelines. METHODS Patients who underwent resection of gastric cancer from 2005 to 2017 were identified in the National Cancer Database. Patients treated at academic programs were compared to those treated at nonacademic programs, and rates and trends of adherence to nodal sampling guidelines (defined as ≥15 lymph nodes) were determined. Adjusted multivariable analysis was used to determine likelihood of nodal sampling adherence while controlling for sociodemographic, clinical, hospital, and travel distance characteristics. RESULTS A total of 55,421 patients were included with 27,201 (49.1%) of patients meeting adherence criteria for lymph node sampling. Academic programs treated 44.4% of the total cohort. Overall, lymph node sampling criteria were met in 59.2% of patients treated at high-volume academic programs and 37.0% of patients treated at low-volume nonacademic programs (incidence rate ratios 0.67, 95% confidence interval 0.63-0.72 versus high-volume academic programs). Adherence rates improved from 2005 to 2017 for both low-volume nonacademic programs (27.8% in 2005 to 50.1% in 2017) and high-volume academic programs (46.0% in 2005 to 69.8% in 2017, P < 0.001). CONCLUSIONS Though adherence rates have improved from 2005 to 2017, high-volume academic programs were more likely to adhere to lymph node sampling guidelines for gastric cancer.
Collapse
Affiliation(s)
- Ashorne K Mahenthiran
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Charles D Logan
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Surgery, Canning Thoracic Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Lauren M Janczewski
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Catherine Valukas
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Samantha Warwar
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Casey M Silver
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joe Feinglass
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ryan P Merkow
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David J Bentrem
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David D Odell
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Surgery, Canning Thoracic Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
24
|
Park SH, Kim KY, Cho M, Kim YM, Kim HI, Hyung WJ. Prognostic impact of fluorescent lymphography on gastric cancer. Int J Surg 2023; 109:2926-2933. [PMID: 37352518 PMCID: PMC10583912 DOI: 10.1097/js9.0000000000000572] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/11/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND Fluorescent lymphography-guided lymphadenectomy (FL) for gastric cancer is gaining popularity. However, its impact on prognosis is not known. This study aimed to assess the prognostic impact of FL in gastric cancer patients. MATERIALS AND METHODS This study retrospectively analyzed 5678 gastric cancer patients who underwent gastrectomy from 2013 to 2017. The survival was compared between the FLFL group and the conventional lymphadenectomy (non-FL group) using 1:1 propensity score matching after exclusion. Patients in the FL group underwent gastrectomy with systematic lymphadenectomy after endoscopic peritumoral injection of indocyanine green the day before surgery. RESULTS After propensity score matching, the FL and non-FL groups each had 1064 patients with similar demographic and clinicopathological characteristics. All matched variables had a standardized mean difference under 0.1. The FL group showed a significantly higher number of retrieved lymph nodes (56.2±20.1) than the non-FL group (46.2±18.2, P <0.001). The FL group also had more stage III patients ( P= 0.044) than the non-FL group. The FL group demonstrated higher overall survival ( P= 0.038) and relapse-free survival ( P= 0.036) in stage III compared with the non-FL group. However, no significant differences in overall and relapse-free survival were observed between the two groups for stages I ( P= 0.420 and P= 0.120, respectively) and II ( P= 0.200 and P= 0.280, respectively). CONCLUSION FL demonstrated a higher survival in stage III gastric cancer patients by the more accurate staging resulting from larger lymph node retrieval. Thus, given its potential to improve prognostication by enhancing staging accuracy, it is recommended as an option to consider the use of FL in clinical practice.
Collapse
Affiliation(s)
- Sung Hyun Park
- Department of Surgery, Yonsei University College of Medicine
- Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Ki-Yoon Kim
- Department of Surgery, Yonsei University College of Medicine
- Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Minah Cho
- Department of Surgery, Yonsei University College of Medicine
- Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Yoo Min Kim
- Department of Surgery, Yonsei University College of Medicine
- Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Hyoung-Il Kim
- Department of Surgery, Yonsei University College of Medicine
- Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Woo Jin Hyung
- Department of Surgery, Yonsei University College of Medicine
- Gastric Cancer Center, Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
| |
Collapse
|
25
|
van der Wielen N, Daams F, Rosati R, Parise P, Weitz J, Reissfelder C, Del Val ID, Loureiro C, Parada-González P, Pintos-Martínez E, Vallejo FM, Achirica CM, Sánchez-Pernaute A, Campos AR, Bonavina L, Asti ELG, Poza AA, Gilsanz C, Nilsson M, Lindblad M, Gisbertz SS, van Berge Henegouwen MI, Romario UF, De Pascale S, Akhtar K, Cuesta MA, van der Peet DL, Straatman J. Three-year survival and distribution of lymph node metastases in gastric cancer following neoadjuvant chemotherapy: results from a European randomized clinical trial. Surg Endosc 2023; 37:7317-7324. [PMID: 37468751 PMCID: PMC10462494 DOI: 10.1007/s00464-023-10278-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/02/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Adequate lymphadenectomy is an important step in gastrectomy for cancer, with a modified D2 lymphadenectomy being recommended for advanced gastric cancers. When assessing a novel technique for the treatment of gastric cancer, lymphadenectomy should be non-inferior. The aim of this study was to assess completeness of lymphadenectomy and distribution patterns between open total gastrectomy (OTG) and minimally invasive total gastrectomy (MITG) in the era of peri-operative chemotherapy. METHODS This is a retrospective analysis of the STOMACH trial, a randomized clinical trial in thirteen hospitals in Europe. Patients were randomized between OTG and MITG for advanced gastric cancer after neoadjuvant chemotherapy. Three-year survival, number of resected lymph nodes, completeness of lymphadenectomy, and distribution patterns were examined. RESULTS A total of 96 patients were included in this trial and randomized between OTG (49 patients) and MITG (47 patients). No difference in 3-year survival was observed, this was 57.1% in OTG group versus 46.8% in MITG group (P = 0.186). The mean number of examined lymph nodes per patient was 44.3 ± 16.7 in the OTG group and 40.7 ± 16.3 in the MITG group (P = 0.209). D2 lymphadenectomy of 71.4% in the OTG group and 74.5% in the MITG group was performed according to the surgeons; according to the pathologist compliance to D2 lymphadenectomy was 30% in the OTG group and 36% in the MITG group. Tier 2 lymph node metastases (stations 7-12) were observed in 19.6% in the OTG group versus 43.5% in the MITG group (P = 0.024). CONCLUSION No difference in 3-year survival was observed between open and minimally invasive gastrectomy. No differences were observed for lymph node yield and type of lymphadenectomy. Adherence to D2 lymphadenectomy reported by the pathologist was markedly low.
Collapse
Affiliation(s)
- Nicole van der Wielen
- Department of Gastrointestinal Surgery, Amsterdam University Medical Center Location VU University Medical Center, De Boelelaan 1117, ZH 7F020, 1081 HV, Amsterdam, The Netherlands.
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| | - Freek Daams
- Department of Gastrointestinal Surgery, Amsterdam University Medical Center Location VU University Medical Center, De Boelelaan 1117, ZH 7F020, 1081 HV, Amsterdam, The Netherlands
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Riccardo Rosati
- Department of Gastrointestinal Surgery, San Raffaele Hospital, Milan, Italy
| | - Paolo Parise
- Department of Gastrointestinal Surgery, San Raffaele Hospital, Milan, Italy
| | - Jürgen Weitz
- Department of of Visceral-, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Christoph Reissfelder
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | | | - Carlos Loureiro
- Department of Surgery, Hospital Universitario de Basurto, Bilbao, Spain
| | | | - Elena Pintos-Martínez
- Department of Surgery, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | | | | | - Luigi Bonavina
- Department of Surgery and Division of Foregut Surgery, IRCCS Policlinico San Donato, University of Milan, Milan, Italy
| | - Emanuele L G Asti
- Department of Surgery and Division of Foregut Surgery, IRCCS Policlinico San Donato, University of Milan, Milan, Italy
| | | | - Carlos Gilsanz
- Department of Surgery, Hospital del Sureste, Madrid, Spain
| | - Magnus Nilsson
- Division of Surgery, Department of Clinical Science Intervention and Technology, Karolinska Institutet and Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Mats Lindblad
- Division of Surgery, Department of Clinical Science Intervention and Technology, Karolinska Institutet and Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Suzanne S Gisbertz
- Department of Surgery, Amsterdam UMC, Location AMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Mark I van Berge Henegouwen
- Department of Surgery, Amsterdam UMC, Location AMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Stefano De Pascale
- Digestive Surgery, European Institute of Oncology - IRCCS - Milan, Milan, Italy
| | - Khurshid Akhtar
- Department of Surgery, Salford Royal NHS Foundation Trust, Manchester, UK
| | - Miguel A Cuesta
- Department of Gastrointestinal Surgery, Amsterdam University Medical Center Location VU University Medical Center, De Boelelaan 1117, ZH 7F020, 1081 HV, Amsterdam, The Netherlands
| | - Donald L van der Peet
- Department of Gastrointestinal Surgery, Amsterdam University Medical Center Location VU University Medical Center, De Boelelaan 1117, ZH 7F020, 1081 HV, Amsterdam, The Netherlands
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jennifer Straatman
- Department of Gastrointestinal Surgery, Amsterdam University Medical Center Location VU University Medical Center, De Boelelaan 1117, ZH 7F020, 1081 HV, Amsterdam, The Netherlands
- Department of Clinical Epidemiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
26
|
He Q, Huangfu L, Fan B, Zhuang Q, He L, Li L, You W, Xing X. T-cells infiltration mediates the association between neutrophil/lymphocyte ratio and survival in gastric cancer. Cancer Med 2023; 12:15893-15902. [PMID: 37306187 PMCID: PMC10469634 DOI: 10.1002/cam4.6228] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/15/2023] [Accepted: 06/01/2023] [Indexed: 06/13/2023] Open
Abstract
BACKGROUND Neutrophil/lymphocyte ratio (NLR) is a vital index for systemic inflammation and a prognostic indicator for gastric cancer (GC). Despite the abundant literature on NLR's prognostic value for GC, the underlying factors mediating its impact on survival remain unclear. The objective of this study was to analyze the role of NLR in different prognostic models and subgroups, and investigate the mediating effects of immune infiltrates between NLR and survival. METHODS A total of 924 patients who underwent D2 lymph node resection were enrolled in this study. According to the level of NLR, patients were divided into two groups, the high and low NLR groups. Clinical parameters, indexes related to immune infiltrates, and survival were compared between the two groups. Prognostic models, interaction analysis, and mediating effects analysis were performed to investigate the clinical association of NLR, immune infiltrates, and survival. RESULTS The infiltration of CD3+ and CD8+ T cells was significantly different in the two NLR groups. The level of NLR was an independent prognostic predictor of GC. In addition, an interaction effect exists between NLR and MMR status on the prognosis of GC (p-interaction <0.01). Lastly, the mediating effect analysis revealed that the infiltration level of CD3+ T cells was the mediating factor between NLR and survival (p < 0.001). CONCLUSIONS The level of NLR is an independent prognostic predictor of GC. The effect of NLR on prognosis is partly mediated by CD3+ T-cell infiltration.
Collapse
Affiliation(s)
- Qifei He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer CenterPeking University Cancer Hospital and InstituteBeijingChina
- Department of Bone Joint and Musculoskeletal TumorThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's HospitalShenzhenChina
| | - Longtao Huangfu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer CenterPeking University Cancer Hospital and InstituteBeijingChina
| | - Biao Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer CenterPeking University Cancer Hospital and InstituteBeijingChina
| | - Qianzheng Zhuang
- Department of Bone Joint and Musculoskeletal TumorThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's HospitalShenzhenChina
| | - Liu He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer CenterPeking University Cancer Hospital and InstituteBeijingChina
| | - Lin Li
- Department of Gastroenterology, Aerospace Center HospitalPeking University Aerospace School of Clinical MedicineBeijingChina
| | - Wei You
- Department of Bone Joint and Musculoskeletal TumorThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's HospitalShenzhenChina
| | - Xiaofang Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer CenterPeking University Cancer Hospital and InstituteBeijingChina
| |
Collapse
|
27
|
Racz K, Legner A, Böhme F, Sebesta C. [Gastric cancer]. Wien Med Wochenschr 2023; 173:227-231. [PMID: 37071301 DOI: 10.1007/s10354-023-01011-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/06/2023] [Indexed: 04/19/2023]
Abstract
Gastric cancers are relativ commonly cancer types. The therapy options have changed in the last years as well in the surgery as in the oncology, it is worth to look at the etiology, diagnosis and therapy.
Collapse
Affiliation(s)
- Krisztina Racz
- 2. Medizinische Abteilung, Klinik Donaustadt, Donaustadt, Österreich.
| | - Andras Legner
- Chirurgische Abteilung, Klinik Donaustadt, Donaustadt, Österreich
| | - Felix Böhme
- 2. Medizinische Abteilung, Klinik Donaustadt, Donaustadt, Österreich
| | - Christian Sebesta
- 2. Medizinische Abteilung, Klinik Donaustadt, Donaustadt, Österreich
| |
Collapse
|
28
|
Yan Y, Ou C, Cao S, Hua Y, Sha Y. Laparoscopic vs. open distal gastrectomy for locally advanced gastric cancer: A systematic review and meta-analysis of randomized controlled trials. Front Surg 2023; 10:1127854. [PMID: 36874456 PMCID: PMC9982133 DOI: 10.3389/fsurg.2023.1127854] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/23/2023] [Indexed: 02/19/2023] Open
Abstract
Objective The aim of this systematic review and meta-analysis is to compare the short- and long-term outcomes of laparoscopic distal gastrectomy (LDG) with those of open distal gastrectomy (ODG) for patients with advanced gastric cancer (AGC) who exclusively underwent distal gastrectomy and D2 lymphadenectomy in randomized controlled trials (RCTs). Background Data in published meta-analyses that included different gastrectomy types and mixed tumor stages prevented an accurate comparison between LDG and ODG. Recently, several RCTs that compared LDG with ODG included AGC patients specifically for distal gastrectomy, with D2 lymphadenectomy being reported and updated with the long-term outcomes. Methods PubMed, Embase, and Cochrane databases were searched to identify RCTs for comparing LDG with ODG for advanced distal gastric cancer. Short-term surgical outcomes and mortality, morbidity, and long-term survival were compared. The Cochrane tool and GRADE approach were used for evaluating the quality of evidence (Prospero registration ID: CRD42022301155). Results Five RCTs consisting of a total of 2,746 patients were included. Meta-analyses showed no significant differences in terms of intraoperative complications, overall morbidity, severe postoperative complications, R0 resection, D2 lymphadenectomy, recurrence, 3-year disease-free survival, intraoperative blood transfusion, time to first liquid diet, time to first ambulation, distal margin, reoperation, mortality, or readmission between LDG and ODG. Operative times were significantly longer for LDG [weighted mean difference (WMD) 49.2 min, p < 0.05], whereas harvested lymph nodes, intraoperative blood loss, postoperative hospital stay, time to first flatus, and proximal margin were lower for LDG (WMD -1.3, p < 0.05; WMD -33.6 mL, p < 0.05; WMD -0.7 day, p < 0.05; WMD -0.2 day, p < 0.05; WMD -0.4 mm, p < 0.05). Intra-abdominal fluid collection and bleeding were found to be less after LDG. Certainty of evidence ranged from moderate to very low. Conclusions Data from five RCTs suggest that LDG with D2 lymphadenectomy for AGC has similar short-term surgical outcomes and long-term survival to ODG when performed by experienced surgeons in hospitals contending with high patient volumes. It can be concluded that RCTs should highlight the potential advantages of LDG for AGC. Systematic Review Registration PROSPERO, registration number CRD42022301155.
Collapse
Affiliation(s)
- Yong Yan
- Department of General Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Caiwen Ou
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shunwang Cao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yinggang Hua
- Department of General Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Yanhua Sha
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
29
|
Li H, Lin D, Yu Z, Li H, Zhao S, Hainisayimu T, Liu L, Wang K. A nomogram model based on the number of examined lymph nodes-related signature to predict prognosis and guide clinical therapy in gastric cancer. Front Immunol 2022; 13:947802. [PMID: 36405735 PMCID: PMC9667298 DOI: 10.3389/fimmu.2022.947802] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/30/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Increasing evidence suggests that the number of examined lymph nodes (ELNs) is strongly linked to the survivorship of gastric cancer (GC). The goal of this study was to assess the prognostic implications of the ELNs number and to construct an ELNs-based risk signature and nomogram model to predict overall survival (OS) characteristics in GC patients. METHODS This inception cohort study included 19,317 GC patients from the U.S. Surveillance, Epidemiology, and End Results (SEER) database, who were separated into a training group and an internal validation group. The nomogram was built with the training set, then internally verified with SEER data, and externally validated with two different data sets. Based on the RNA-seq data, ELNs-related DERNAs (DElncRNAs, DEmiRNAs, andDEmRNAs) and immune cells were identified. The LASSO-Cox regression analysis was utilized to construct ELNs-related DERNAs and immune cell prognostic signature in The Cancer Genome Atlas (TCGA) cohort. The OS of subgroups with high- and low-ELN signature was compared using the Kaplan-Meier (K-M) analysis. A nomogram was successfully constructed based on the ELNs signature and other clinical characteristics. The concordance index (C-index), calibration plot, receiver operating characteristic curve, and decision curve analysis (DCA) were all used to evaluate the nomogram model. The meta-analysis, the Gene Expression Profiling Interactive Analysis database, and reverse transcription-quantitative PCR (RT-qPCR) were utilized to validate the RNA expression or abundance of prognostic genes and immune cells between GC tissues and normal gastric tissues, respectively. Finally, we analyzed the correlations between immune checkpoints, chemotherapy drug sensitivity, and risk score. RESULTS The multivariate analysis revealed that the high ELNs improved OS compared with low ELNs (hazard ratio [HR] = 0.659, 95% confidence interval [CI]: 0.626-0.694, p < 0.0001). Using the training set, a nomogram incorporating ELNs was built and proven to have good calibration and discrimination (C-index [95% CI], 0.714 [0.710-0.718]), which was validated in the internal validation set (C-index [95% CI], 0.720 [0.714-0.726]), the TCGA set (C-index [95% CI], 0.693 [0.662-0.724]), and the Chinese set (C-index [95% CI], 0.750 [0.720-0.782]). An ELNs-related signature model based on ELNs group, regulatory T cells (Tregs), neutrophils, CDKN2B-AS1, H19, HOTTIP, LINC00643, MIR663AHG, TMEM236, ZNF705A, and hsa-miR-135a-5p was constructed by the LASSO-Cox regression analysis. The result showed that OS was remarkably lower in patients with high-ELNs signature compared with those with low-ELN signature (HR = 2.418, 95% CI: 1.804-3.241, p < 0.001). This signature performed well in predicting 1-, 3-, and 5-year survival (AUC [95% CI] = 0.688 [0.612-0.763], 0.744 [0.659-0.830], and 0.778 [0.647-0.909], respectively). The multivariate Cox analysis illustrated that the risk score was an independent predictor of survival for patients with GC. Moreover, the expression of prognostic genes (LINC00643, TMEM236, and hsa-miR-135a-5p) displayed differences between GC tissues and adjacent non-tumor tissues. The C-index of the nomogram that can be used to predict the OS of GC patients was 0.710 (95% CI: 0.663-0.753). Both the calibration plots and DCA showed that the nomogram has good predictive performance. Moreover, the signature was significantly correlated with the N stage and T stage. According to our analysis, GC patients in the low-ELN signature group may have a better immunotherapy response and OS outcome. CONCLUSIONS We explored the prognostic role of ELNs in GC and successfully constructed an ELNs signature linked to the GC prognosis in TCGA. The findings manifested that the signature is a powerful predictive indicator for patients with GC. The signature might contain potential biomarkers for treatment response prediction for GC patients. Additionally, we identified a novel and robust nomogram combining the characteristics of ELNs and clinical factors for predicting 1-, 3-, and 5-year OS in GC patients, which will facilitate personalized survival prediction and aid clinical decision-making in GC patients.
Collapse
Affiliation(s)
- Huling Li
- School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Dandan Lin
- School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Zhen Yu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| | - Hui Li
- Central Laboratory of Xinjiang Medical University, Urumqi, China
| | - Shi Zhao
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Tuersun Hainisayimu
- Department of Biochemistry and Molecular Biology, Basic Medicine School, Xinjiang Medical University, Urumqi, China
| | - Lin Liu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital, Xinjiang Medical University, Urumqi, China,*Correspondence: Kai Wang, ; Lin Liu,
| | - Kai Wang
- Department of Medical Engineering and Technology, Xinjiang Medical University, Urumqi, China,*Correspondence: Kai Wang, ; Lin Liu,
| |
Collapse
|
30
|
Yu C, Zhou Z, Liu B, Yao D, Huang Y, Wang P, Li Y. Pathological Nodal Staging Score for Gastric Signet Ring Cell Carcinoma: A Clinical Tool of Adequate Nodal Staging. Diagnostics (Basel) 2022; 12:diagnostics12102289. [PMID: 36291978 PMCID: PMC9600920 DOI: 10.3390/diagnostics12102289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/11/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Gastric signet ring cell carcinoma (GSRCC) is a subset of gastric cancer with distinct histological and inconsistent prognosis outcome. Currently, the association between the adequate regional lymph node and proper nodal staging in GSRCC is rarely noticed. Materials and methods: Clinical data of GSRCC were retrieved from the Surveillance, Epidemiology, and End Results database. Beta-binomial distribution model was employed for the estimation of the probability of missing nodal disease, followed by the development of a nodal staging score (NSS). Results: A total of 561 GSRCC patients were included in this study, with 193 in lymph node-negative and 368 in lymph node-positive diagnoses. As the number of examined lymph nodes increased, the probability of missing nodal disease decreased rapidly, with T stage-specific curves. The probability of missing nodal disease in T4 was lower than that in T1. NSS calculation indicated that T1 stage patients commonly had NSS > 0.8. However, with the NSS of T2−T4 to reach 0.8, the number of examined lymph node was required to be larger than 12 in T2, 17 in T3 and 27 in T4. NSS ≥ 0.75 (quantile 75%) subgroup in T2−T4 subgroups tended to have better outcome; however, without significant prognostic value. Conclusions: NSS is served as a reliable and feasible tool in adequate nodal staging of GSRCC with statistical basis and provides further evidence for clinical decision making.
Collapse
|
31
|
Puccetti F, Cinelli L, Genova L, Battaglia S, Barbieri LA, Treppiedi E, Cossu A, Elmore U, Rosati R. Applicative Limitations of Indocyanine Green Fluorescence Assistance to Laparoscopic Lymph Node Dissection in Total Gastrectomy for Cancer. Ann Surg Oncol 2022; 29:5875-5882. [PMID: 35729291 DOI: 10.1245/s10434-022-11940-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/08/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Indocyanine green (ICG) fluorescence has been recently introduced as a novel imaging technique improving the accuracy of lymph node (LN) dissection in gastric cancer (GC) surgery, although procedure standardization and achievements have not been clearly defined. This study analyzed the feasibility and effectiveness of ICG-guidance for laparoscopic D2-lymphadenectomy during total gastrectomy for cancer. METHODS This study retrospectively analyzed a single-center series of patients who underwent laparoscopic total gastrectomy for cancer between April 2015 and August 2021. All patients underwent surgery with standard D2 LN dissection. Intraoperative ICG-fluorescence was institutionally implemented in April 2018 and was performed routinely afterward. Primary outcomes were LN harvest and ratio. Secondary endpoints included operative time and subgroup analysis to assess variables potentially affecting LN retrieval. RESULTS The study population included 102 patients, and ICG-fluorescence was applied in 38 (37.3%). ICG and no-ICG groups presented similar median age, gender proportions, ASA score and comorbidities (age-adjusted Charlson Comorbidity Index), body mass index, and advanced pathological stage. The median of LNs retrieved was significantly higher after the intraoperative ICG-guidance (44 vs. 32; p = 0.004), although this association was not significant after neoadjuvant therapy or among patients with positive LNs. Lymph node ratio and operative time were not significantly impacted by ICG fluorescence. Multivariate analysis identified the ICG-assistance as the only independent determinant for LN harvest (p = 0.029). CONCLUSIONS ICG-guidance contributes to a significantly wider LN retrieval after laparoscopic D2-lymphadenectomy during total gastrectomy for cancer. However, neoadjuvant therapy and positive LN stage appeared to limit the procedural effectiveness to ICG-assisted LN identification.
Collapse
Affiliation(s)
- Francesco Puccetti
- Department of Gastrointestinal Surgery, San Raffaele Research Hospital, 20132, Milan, Italy.,Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Lorenzo Cinelli
- Department of Gastrointestinal Surgery, San Raffaele Research Hospital, 20132, Milan, Italy
| | - Luana Genova
- Department of Gastrointestinal Surgery, San Raffaele Research Hospital, 20132, Milan, Italy
| | - Silvia Battaglia
- Department of Gastrointestinal Surgery, San Raffaele Research Hospital, 20132, Milan, Italy
| | - Lavinia A Barbieri
- Department of Gastrointestinal Surgery, San Raffaele Research Hospital, 20132, Milan, Italy
| | - Elio Treppiedi
- Department of Gastrointestinal Surgery, San Raffaele Research Hospital, 20132, Milan, Italy
| | - Andrea Cossu
- Department of Gastrointestinal Surgery, San Raffaele Research Hospital, 20132, Milan, Italy
| | - Ugo Elmore
- Department of Gastrointestinal Surgery, San Raffaele Research Hospital, 20132, Milan, Italy. .,Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132, Milan, Italy.
| | - Riccardo Rosati
- Department of Gastrointestinal Surgery, San Raffaele Research Hospital, 20132, Milan, Italy.,Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132, Milan, Italy
| |
Collapse
|
32
|
Mranda GM, Xue Y, Zhou XG, Yu W, Wei T, Xiang ZP, Liu JJ, Ding YL. Revisiting the 8th AJCC system for gastric cancer: A review on validations, nomograms, lymph nodes impact, and proposed modifications. Ann Med Surg (Lond) 2022; 75:103411. [PMID: 35386808 PMCID: PMC8977912 DOI: 10.1016/j.amsu.2022.103411] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/23/2022] [Indexed: 12/20/2022] Open
Abstract
Gastric cancer is the fifth most frequently diagnosed cancer worldwide, behind breast, lung, colorectal, and prostate cancers. In gastric cancer, multimodality treatment shows prospective benefits and also improves survival. Surgery, however, is the mainstay of curative treatment. The staging of gastric cancer patients is critical for harmonization of care. Accurate stages assure that informed clinical decisions are timely made. The American Joint Committee on Cancer (AJCC) staging system is the most widely applied system in to determine the disease's prognosis and survival prediction. The recently adopted 8th AJCC TNM staging system has been revised to enhance its survival predictive power. Subsequent studies have established the validity of the current edition, demonstrating improved stage stratification, discriminatory power, and survival prediction. However, other studies have cast doubt on the superiority of the new edition. Innovations aimed at further improving its prognosis have resulted in developing of novel models. Advances in our understanding of the tumor microenvironment and molecular categorization of cancer have resulted in proposals for their inclusion in TNM staging as potential complementary factors that enhance survival prediction and prognostic assessment ability. The purpose of this study is to conduct a review of the published literature regarding the validity of the 8th AJCC TNM staging system, proposed modifications, and nomograms.
The 8th AJCC is valid in prognostic stratification of gastric cancer, however, revisions are still required. The yPT staging requires some modifications and inclusion of stages that currently don't exist in the 8th AJCC. High lymph nodes count and anatomical localization improve the prediction ability of the current AJCC. Nomograms comprising of individual prognostic factors are crucial to the current AJCC. Molecular markers positively influence survival prediction of gastric cancer.
Collapse
Affiliation(s)
- Geofrey Mahiki Mranda
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
- Department of General Surgery, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Ying Xue
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Xing-Guo Zhou
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Wang Yu
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Tian Wei
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Zhi-Ping Xiang
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Jun-Jian Liu
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Yin-Lu Ding
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
- Corresponding author. Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247 Beiyuan Street, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
33
|
Ambrosio MR, Perotti B, Battini A, Fattorini C, Cavazzana A, Pasqua R, Palumbo P, Gia L, Arganini M. Surgeon-Pathologist Team Approach Dramatically Affects Lymph Nodes Detection and Improves Patients' Short-Term Outcome. Cancers (Basel) 2022; 14:cancers14041034. [PMID: 35205783 PMCID: PMC8870551 DOI: 10.3390/cancers14041034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/07/2022] [Accepted: 02/11/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Appropriate lymph node harvesting for patients with gastric cancer is fundamental for a correct staging and is strongly related to survival. In this study, we present a new protocol for on-site macroscopic evaluation and sampling of lymph nodes for gastric cancer patients. With the joint collaboration of surgeons and pathologists, our method aims to provide the largest possible number of analyzed lymph nodes per patient, allowing for a better staging. We are convinced that this approach is routinely feasible, and our preliminary results seem to confirm better patient stratification compared to other lymph node dissection methods. Abstract The downstaging of gastric cancer has recently gained particular attention in the field of gastric cancer surgery. The phenomenon is mainly due to an inappropriate sampling of lymph nodes during standard lymphadenectomy. Hence, collection of the maximum number of lymph nodes is a critical factor affecting the outcome of patients. None of the techniques proposed so far have demonstrated a real efficiency in increasing the number of identified lymph nodes. To harvest the maximum number of lymph nodes, we designed a protocol for on-site macroscopic evaluation and sampling of lymph nodes according to the Japanese Gastric Cancer Association protocol. The procedure was carried out by a surgeon/pathologist team in the operating room. We enrolled one hundred patients, 50 of whom belonged to the study group and 50 to a control group. The study group included patients who underwent lymph node dissection following the proposed protocol; the control group encompassed patients undergoing standard procedures for sampling. We compared the number and maximum diameter of lymph nodes collected in both groups, as well as some postoperative variables, the 30-day mortality and the overall survival. In the study group, the mean number of lymph nodes harvested was higher than the control one (p = 0.001). Moreover, by applying the proposed technique, we sampled lymph nodes with a very small diameter, some of which were metastatic. Noticeably, no difference in terms of postoperative course was identified between the two groups, again supporting the feasibility of an extended lymphadenectomy. By comparing the prognosis of patients, a better overall survival (p = 0.03) was detected in the study group; however, to date, no long-term follow-up is available. Interestingly, patients with metastasis in node stations number 8, 9, 11 or with skip metastasis, experienced a worse outcome and died. Based on our preliminary results, the pathologist/surgeon team approach seems to be a reliable option, despite of a slight increase in sfaff workload and technical cost. It allows for the harvesting of a larger number of lymph nodes and improves the outcome of the patients thanks to more precise staging and therapy. Nevertheless, since a higher number of patients are necessary to confirm our findings and assess the impact of this technique on oncological outcome, our study could serve as a proof-of-concept for a larger, multicentric collaboration.
Collapse
Affiliation(s)
- Maria Raffaella Ambrosio
- Pathology Unit, Azienda Sanitaria Toscana Nord-Ovest, Via Cocchi 1, 56121 Pisa, Italy; (C.F.); (A.C.)
- Correspondence:
| | - Bruno Perotti
- Surgery Unit, Ospedale Unico Versilia and Nuovo Ospedale Apuane, Azienda Sanitaria Toscana Nord Ovest, 56121 Pisa, Italy; (B.P.); (A.B.); (L.G.); (M.A.)
| | - Alda Battini
- Surgery Unit, Ospedale Unico Versilia and Nuovo Ospedale Apuane, Azienda Sanitaria Toscana Nord Ovest, 56121 Pisa, Italy; (B.P.); (A.B.); (L.G.); (M.A.)
| | - Caterina Fattorini
- Pathology Unit, Azienda Sanitaria Toscana Nord-Ovest, Via Cocchi 1, 56121 Pisa, Italy; (C.F.); (A.C.)
| | - Andrea Cavazzana
- Pathology Unit, Azienda Sanitaria Toscana Nord-Ovest, Via Cocchi 1, 56121 Pisa, Italy; (C.F.); (A.C.)
| | - Rocco Pasqua
- Department of Surgical Sciencies, University “La Sapienza”, 00100 Roma, Italy; (R.P.); (P.P.)
| | - Piergaspare Palumbo
- Department of Surgical Sciencies, University “La Sapienza”, 00100 Roma, Italy; (R.P.); (P.P.)
| | - Liano Gia
- Surgery Unit, Ospedale Unico Versilia and Nuovo Ospedale Apuane, Azienda Sanitaria Toscana Nord Ovest, 56121 Pisa, Italy; (B.P.); (A.B.); (L.G.); (M.A.)
| | - Marco Arganini
- Surgery Unit, Ospedale Unico Versilia and Nuovo Ospedale Apuane, Azienda Sanitaria Toscana Nord Ovest, 56121 Pisa, Italy; (B.P.); (A.B.); (L.G.); (M.A.)
| |
Collapse
|
34
|
Gong S, Li X, Tian H, Song S, Lu T, Jing W, Huang X, Xu Y, Wang X, Zhao K, Yang K, Guo T. Clinical efficacy and safety of robotic distal gastrectomy for gastric cancer: a systematic review and meta-analysis. Surg Endosc 2022; 36:2734-2748. [PMID: 35020057 DOI: 10.1007/s00464-021-08994-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/31/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Robotic distal gastrectomy (RDG) is a new technique that is rapidly gaining popularity and may help overcome the limitations of laparoscopic distal gastrectomy (LDG); however, its safety and therapeutic efficacy remain controversial. Therefore, this meta-analysis was performed to evaluate the safety and efficacy of RDG. METHODS We searched PubMed, EMBASE, the Cochrane Library, and Web of Science for studies that compared RDG and LDG and were published between the time of database inception and May 2021. We assessed the bias risk of the observational studies using ROBIN-I, and a random effect model was always applied. RESULTS The meta-analysis included 22 studies involving 5386 patients. Compared with LDG, RDG was associated with longer operating time (Mean Difference [MD] = 43.88, 95% CI = 35.17-52.60), less intraoperative blood loss (MD = - 24.84, 95% CI = - 41.26 to - 8.43), a higher number of retrieved lymph nodes (MD = 2.41, 95% CI = 0.77-4.05), shorter time to first flatus (MD = - 0.09, 95% CI = - 0.15 to - 0.03), shorter postoperative hospital stay (MD = - 0.68, 95% CI = - 1.27 to - 0.08), and lower incidence of pancreatic fistula (OR = 0.23, 95% CI = 0.07-0.79). Mean proximal and distal resection margin distances, time to start liquid and soft diets, and other complications were not significantly different between RDG and LDG groups. However, in the propensity-score-matched meta-analysis, the differences in time to first flatus and postoperative hospital stay between the two groups lost significance. CONCLUSIONS Based on the available evidence, RDG appears feasible and safe, shows better surgical and oncological outcomes than LDG and, comparable postoperative recovery and postoperative complication outcomes.
Collapse
Affiliation(s)
- Shiyi Gong
- Ningxia Medical University, Yinchuan, 750000, Ningxia, China.,Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China.,Institution of Clinical Research and Evidence-Based Medicine, The Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 222 West Donggang R.D., Lanzhou, 730000, Gansu, China
| | - Xiong Li
- Ningxia Medical University, Yinchuan, 750000, Ningxia, China.,Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China.,Institution of Clinical Research and Evidence-Based Medicine, The Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 222 West Donggang R.D., Lanzhou, 730000, Gansu, China
| | - Hongwei Tian
- Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China.,Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Shaoming Song
- Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China.,Institution of Clinical Research and Evidence-Based Medicine, The Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 222 West Donggang R.D., Lanzhou, 730000, Gansu, China
| | - Tingting Lu
- Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China.,Institution of Clinical Research and Evidence-Based Medicine, The Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 222 West Donggang R.D., Lanzhou, 730000, Gansu, China.,Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Wutang Jing
- Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China
| | - Xianbin Huang
- Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China.,Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Yongcheng Xu
- Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China
| | - Xingqiang Wang
- Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China
| | - Kaixuan Zhao
- Ningxia Medical University, Yinchuan, 750000, Ningxia, China.,Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China
| | - Kehu Yang
- Institution of Clinical Research and Evidence-Based Medicine, The Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China. .,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 222 West Donggang R.D., Lanzhou, 730000, Gansu, China. .,Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, Gansu, China.
| | - Tiankang Guo
- Ningxia Medical University, Yinchuan, 750000, Ningxia, China. .,Department of General Surgery, Gansu Provincial Hospital, 204 West Donggang R.D., Lanzhou, 730000, Gansu, China. .,Department of Clinical Medicine, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
35
|
Survival impact of compliance in extra-perigastric lymphadenectomy for gastric cancer: 20 years of real-world data from a single institution. Surgery 2022; 171:948-954. [DOI: 10.1016/j.surg.2021.09.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/16/2021] [Accepted: 09/19/2021] [Indexed: 12/18/2022]
|
36
|
Spolverato G, Paro A, Capelli G, Dalmacy D, Poultsides GA, Fields RC, Weber SM, Votanopoulos KI, He J, Maithel SK, Pucciarelli S, Pawlik TM. Surgical treatment of gastric adenocarcinoma: Are we achieving textbook oncologic outcomes for our patients? J Surg Oncol 2021; 125:621-630. [PMID: 34964983 DOI: 10.1002/jso.26778] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/28/2021] [Accepted: 11/30/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND AND OBJECTIVES Composite measures are increasingly used to assess quality of care in surgical oncology. We sought to define the incidence of "textbook oncologic outcome" (TOO) following resection of gastric adenocarcinoma among a large, international cohort of patients. METHODS Gastric adenocarcinoma patients undergoing resection between 2000 and 2020 were identified from an international database. TOO was defined as margin-negative resection, examination of ≥16 lymph nodes, no prolonged length-of-stay (LOS), no 30-day mortality, and stage-appropriate receipt of chemotherapy. RESULTS Among a total of 910 patients, 321 patients (35.3%) achieved a postoperative TOO. While failure to evaluate ≥16 lymph nodes (n = 591, 65.0%) and receipt of chemotherapy (n = 651, 71.5%) had the greatest negative impact on the ability to obtain a TOO, no 30-day mortality (n = 880, 96.7%), margin-negative resection (n = 831, 91.3%), and no extended LOS (n = 706, 77.6%) were more commonly achieved. No postoperative complications (OR: 0.44; 95% CI: 0.31-0.63) and T1a/T1b-stage disease (OR: 2.87; 95% CI: 1.59-5.18) were independently associated with achieving a TOO (p < 0.05). The odds of achieving a TOO improved over time (p-trend < 0.05), which was largely attributable to improved odds of evaluating ≥16 lymph nodes (2010-2014 vs. 2000-2004: OR, 5.21; 95% CI: 3.22-8.45). CONCLUSIONS Only about one in three patients achieved a TOO following resection of gastric adenocarcinoma. Odds of TOO increased over time, largely due to improved lymph node evaluation.
Collapse
Affiliation(s)
- Gaya Spolverato
- Department of Surgical Oncological and Gastrointestinal Science, University of Padova, Padova, Italy
| | - Alessandro Paro
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Giulia Capelli
- Department of Surgical Oncological and Gastrointestinal Science, University of Padova, Padova, Italy
| | - Djhenne Dalmacy
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | | | - Ryan C Fields
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Sharon M Weber
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin, Madison, Wisconsin, USA
| | | | - Jin He
- Department of Surgery, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | | | - Salvatore Pucciarelli
- Department of Surgical Oncological and Gastrointestinal Science, University of Padova, Padova, Italy
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
37
|
Desiderio J, Sagnotta A, Terrenato I, Garofoli E, Mosillo C, Trastulli S, Arteritano F, Tozzi F, D'Andrea V, Fong Y, Woo Y, Bracarda S, Parisi A. Long-term survival of patients with stage II and III gastric cancer who underwent gastrectomy with inadequate nodal assessment. World J Gastrointest Surg 2021; 13:1463-1483. [PMID: 34950434 PMCID: PMC8649557 DOI: 10.4240/wjgs.v13.i11.1463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/30/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer is an aggressive disease with frequent lymph node (LN) involvement. The NCCN recommends a D2 lymphadenectomy and the harvesting of at least 16 LNs. This threshold has been the subject of great debate, not only for the extent of surgery but also for more appropriate staging. The reclassification of stage IIB through IIIC based on N3b nodal staging in the eighth edition of the American Joint Committee on Cancer (AJCC) staging system highlights the efforts to more accurately discriminate survival expectancy based on nodal number. Furthermore, studies have suggested that pathologic assessment of 30 or more LNs improve prognostic accuracy and is required for proper staging of gastric cancer.
AIM To evaluate the long-term survival of advanced gastric cancer patients who deviated from expected survival curves because of inadequate nodal evaluation.
METHODS Eligible patients were identified from the Surveillance, Epidemiology, and End Results database. Those with stage II–III gastric cancer were considered for inclusion. Three groups were compared based on the number of analyzed LNs. They were inadequate LN assessment (ILA, < 16 LNs), adequate LN assessment (ALA, 16-29 LNs), and optimal LN assessment (OLA, ≥ 30 LNs). The main outcomes were overall survival (OS) and cancer-specific survival. Data were analyzed by the Kaplan-Meier product-limit method, log-rank test, hazard risk, and Cox proportional univariate and multivariate models. Propensity score matching (PSM) was used to compare the ALA and OLA groups.
RESULTS The analysis included 11607 patients. Most had advanced T stages (T3 = 48%; T4 = 42%). The pathological AJCC stage distribution was IIA = 22%, IIB = 18%, IIIA = 26%, IIIB = 22%, and IIIC = 12%. The overall sample divided by the study objective included ILA (50%), ALA (35%), and OLA (15%). Median OS was 24 mo for the ILA group, 29 mo for the ALA group, and 34 mo for the OLA group (P < 0.001). Univariate analysis showed that the ALA and OLA groups had better OS than the ILA group [ALA hazard ratio (HR) = 0.84, 95% confidence interval (CI): 0.79–0.88, P < 0.001 and OLA HR = 0.73, 95%CI: 0.68–0.79, P < 0.001]. The OS outcome was confirmed by multivariate analysis (ALA HR = 0.68, 95%CI: 0.64–0.71, P < 0.001 and OLA: HR = 0.48, 95%CI: 0.44–0.52, P < 0.001). A 1:1 PSM analysis in 3428 patients found that the OLA group had better survival than the ALA group (OS: OLA median = 34 mo vs ALA median = 26 mo, P < 0.001, which was confirmed by univariate analysis (HR = 0.81, 95%CI: 0.75–0.89, P < 0.001) and multivariate analysis: (HR = 0.71, 95%CI: 0.65–0.78, P < 0.001).
CONCLUSION Proper nodal staging is a critical issue in gastric cancer. Assessment of an inadequate number of LNs places patients at high risk of adverse long-term survival outcomes.
Collapse
Affiliation(s)
- Jacopo Desiderio
- Department of Digestive Surgery, St. Mary’s Hospital, Terni 05100, Italy
- Department of Surgical Sciences, Sapienza University of Rome, Rome 00161, Italy
| | - Andrea Sagnotta
- Department of General Surgery and Surgical Oncology, San Filippo Neri Hospital, Rome 00135, Italy
| | - Irene Terrenato
- Biostatistics and Bioinformatic Unit, Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome 00144, Italy
| | - Eleonora Garofoli
- Department of Medical Oncology, St. Mary’s Hospital, Terni 05100, Italy
| | - Claudia Mosillo
- Department of Medical Oncology, St. Mary’s Hospital, Terni 05100, Italy
| | - Stefano Trastulli
- Department of Digestive Surgery, St. Mary’s Hospital, Terni 05100, Italy
| | | | - Federico Tozzi
- Division of Surgical Oncology and Endocrine Surgery, Mays Cancer Center, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, United States
| | - Vito D'Andrea
- Department of Surgical Sciences, Sapienza University of Rome, Rome 00161, Italy
| | - Yuman Fong
- Division of Surgical Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, LA, 91010, United States
| | - Yanghee Woo
- Division of Surgical Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, LA, 91010, United States
| | - Sergio Bracarda
- Department of Medical Oncology, St. Mary’s Hospital, Terni 05100, Italy
| | - Amilcare Parisi
- Department of Digestive Surgery, St. Mary’s Hospital, Terni 05100, Italy
| |
Collapse
|
38
|
Chen QY, Zhong Q, Li P, Xie JW, Liu ZY, Huang XB, Lin GT, Wang JB, Lin JX, Lu J, Cao LL, Lin M, Zheng QL, Tu RH, Huang ZN, Zheng CH, Huang CM. Comparison of submucosal and subserosal approaches toward optimized indocyanine green tracer-guided laparoscopic lymphadenectomy for patients with gastric cancer (FUGES-019): a randomized controlled trial. BMC Med 2021; 19:276. [PMID: 34702260 PMCID: PMC8549272 DOI: 10.1186/s12916-021-02125-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/13/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Application of indocyanine green (ICG) fluorescence imaging is effective in guiding laparoscopic radical lymphadenectomy for gastric cancer. However, the optimal approach for indocyanine green injection is controversial. Therefore, the objective of this study was aimed to compare the efficacy and ICG injection between the preoperative submucosal and intraoperative subserosal approaches for lymph node (LN) tracing during laparoscopic gastrectomy. METHOD This randomized controlled trial (ClinicalTrials.gov, NCT04219332) included 266 patients with potentially resectable gastric cancer (cT1-T4a, N0/+, M0) enrolled from a tertiary teaching center between December 2019 and October 2020. The primary endpoint was total number of retrieved LNs. RESULTS In total, 259 patients (n = 130 and n = 129 in the submucosal and subserosal groups, respectively) were included in the per-protocol analysis. There are no significant differences in total number of retrieved LNs between the two groups (49.8 vs. 49.2, P = 0.713). The rate of LN noncompliance in the submucosal group was comparable to that in the subserosal group (32.3% vs. 33.3%, P = 0.860). No significant difference was found between the submucosal and subserosal groups in terms of the incidence (17.7% vs. 16.3%; P = 0.762) or severity of postoperative complications. The mean fluorescence cost in the submucosal group was higher than that in the subserosal group ($335.3 vs. $182.4; P < 0.001). The overall treatment satisfaction score was lower in the submucosal group than in the subserosal group (70.5 vs. 76.1%, P = 0.048). CONCLUSION ICG administered by subserosal injection was comparable to that administered by submucosal injection for lymph node tracing in gastric cancer. However, the former approach imposed a lower economic and mental burden on patients undergoing laparoscopic D2 lymphadenectomy. TRIAL REGISTRATION ClinicalTrials.gov, NCT04219332 .
Collapse
Affiliation(s)
- Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Xiao-Bo Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Qiao-Ling Zheng
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
39
|
Lino-Silva LS, Mendoza-Lara HE, León-Takahashi AM, Zepeda-Najar C, Salcedo-Hernández RA. Lymph node retrieval in radical gastrectomy: the pathologist alone or the surgeon-pathologist team? PRZEGLAD GASTROENTEROLOGICZNY 2021; 16:224-228. [PMID: 34584584 PMCID: PMC8456766 DOI: 10.5114/pg.2021.108987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/18/2020] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Lymph node (LN) dissection is an important prognostic factor in gastric cancer. There is little information comparing the LN count depending on whether they are dissected in the operating room or in the pathology laboratory. AIM To establish if the LN count is greater in either of them. MATERIAL AND METHODS From 2015 to 2017 all consecutive gastrectomies with D2 dissection were prospectively evaluated based in either of 2 protocols: One started in the operating room where the surgeon separated the LN levels and then submitted the entire adipose tissue with LNs (undissected) to pathology in separate containers; the pathologist dissected the LNs from the specimens. The second protocol consisted of sending the tissue/LNs to pathology as usual (adipose tissue and LN attached to the stomach). RESULTS A total of 83 patients were analysed. The mean age was 58.4 years. The median number of LNs dissected in the protocol starting in the operating room was 56 (IQR: 37-74), whereas the pathology laboratory dissected a median of 39 LNs (IQR 26-53) (p = 0.005). The survival of cases dissected by both protocols were comparable (median survival of 48 and 43 months, p = 0.316). CONCLUSIONS The LN final count is significantly higher when LN levels are separated beforehand in the operating room compared to dissection only in pathology; however, this does not impact survival, perhaps because the number of dissected nodes in both groups is high and the quality of the surgery is good.
Collapse
Affiliation(s)
- Leonardo S. Lino-Silva
- Department of Surgical Pathology, Instituto Nacional de Cancerología, Tlalpan, Mexico City, Mexico
| | | | | | - César Zepeda-Najar
- Department of Surgical Oncology, Hospital Angeles Tijuana, Tijuana, Baja California Norte, Mexico
| | | |
Collapse
|
40
|
Trastulli S, Desiderio J, Lin JX, Reim D, Zheng CH, Borghi F, Cianchi F, Norero E, Nguyen NT, Qi F, Coratti A, Cesari M, Bazzocchi F, Alimoglu O, Brower ST, Pernazza G, D’Imporzano S, Azagra JS, Zhou YB, Cao SG, Garofoli E, Mosillo C, Guerra F, Liu T, Arcuri G, González P, Staderini F, Marano A, Terrenato I, D’Andrea V, Bracarda S, Huang CM, Parisi A. Laparoscopic Compared with Open D2 Gastrectomy on Perioperative and Long-Term, Stage-Stratified Oncological Outcomes for Gastric Cancer: A Propensity Score-Matched Analysis of the IMIGASTRIC Database. Cancers (Basel) 2021; 13:4526. [PMID: 34572753 PMCID: PMC8465518 DOI: 10.3390/cancers13184526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/14/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The laparoscopic approach in gastric cancer surgery is being increasingly adopted worldwide. However, studies focusing specifically on laparoscopic gastrectomy with D2 lymphadenectomy are still lacking in the literature. This retrospective study aimed to compare the short-term and long-term outcomes of laparoscopic versus open gastrectomy with D2 lymphadenectomy for gastric cancer. METHODS The protocol-based, international IMIGASTRIC (International study group on Minimally Invasive surgery for Gastric Cancer) registry was queried to retrieve data on patients undergoing laparoscopic or open gastrectomy with D2 lymphadenectomy for gastric cancer with curative intent from January 2000 to December 2014. Eleven predefined, demographical, clinical, and pathological variables were used to conduct a 1:1 propensity score matching (PSM) analysis to investigate intraoperative and recovery outcomes, complications, pathological findings, and survival data between the two groups. Predictive factors of long-term survival were also assessed. RESULTS A total of 3033 patients from 14 participating institutions were selected from the IMIGASTRIC database. After 1:1 PSM, a total of 1248 patients, 624 in the laparoscopic group and 624 in the open group, were matched and included in the final analysis. The total operative time (median 180 versus 240 min, p < 0.0001) and the length of the postoperative hospital stay (median 10 versus 14.8 days, p < 0.0001) were longer in the open group than in the laparoscopic group. The conversion to open rate was 1.9%. The proportion of patients with in-hospital complications was higher in the open group (21.3% versus 15.1%, p = 0.004). The median number of harvested lymph nodes was higher in the laparoscopic approach (median 32 versus 28, p < 0.0001), and the proportion of positive resection margins was higher (p = 0.021) in the open group (5.9%) than in the laparoscopic group (3.2%). There was no significant difference between the groups in five-year overall survival rates (77.4% laparoscopic versus 75.2% open, p = 0.229). CONCLUSION The adoption of the laparoscopic approach for gastric resection with D2 lymphadenectomy shortened the length of hospital stay and reduced postoperative complications with respect to the open approach. The five-year overall survival rate after laparoscopy was comparable to that for patients who underwent open D2 resection. The types of surgical approaches are not independent predictive factors for five-year overall survival.
Collapse
Affiliation(s)
- Stefano Trastulli
- Department of Digestive Surgery, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (S.T.); (A.P.)
| | - Jacopo Desiderio
- Department of Digestive Surgery, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (S.T.); (A.P.)
- Department of Surgical Sciences—PhD Program in Advanced Surgical Technologies, Sapienza University of Rome, 00161 Rome, Italy;
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China; (J.-X.L.); (C.-H.Z.); (C.-M.H.)
| | - Daniel Reim
- Klinik und Poliklinik für Chirurgie, Klinikum Rechts der Isar, Technische Universität München, 81675 Munich, Germany;
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China; (J.-X.L.); (C.-H.Z.); (C.-M.H.)
| | - Felice Borghi
- General and Oncologic Surgery Unit, Department of Surgery, Santa Croce e Carle Hospital, 12100 Cuneo, Italy; (F.B.); (A.M.)
| | - Fabio Cianchi
- Digestive Surgery Unit, Department of Experimental and Clinical Medicine, “Careggi” Hospital, University of Florence, 50134 Florence, Italy; (F.C.); (F.S.)
| | - Enrique Norero
- Esophagogastric Surgery Unit, Digestive Surgery Department, Hospital Dr. Sotero del Rio, Pontificia Universidad Catolica de Chile, Santiago 8207257, Chile; (E.N.); (P.G.)
| | - Ninh T. Nguyen
- Irvine Medical Center, Department of Surgery, Division of Gastrointestinal Surgery, University of California, Orange, CA 92868, USA;
| | - Feng Qi
- Gastrointestinal Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (F.Q.); (T.L.)
| | - Andrea Coratti
- Department of General and Emergency Surgery, Division of General and Emergency Surgery, School of Robotic Surgery, Misericordia Hospital of Grosseto, 58100 Grosseto, Italy; (A.C.); (F.G.)
| | - Maurizio Cesari
- Department of General Surgery, Hospital of Città di Castello, USL1 Umbria, 06012 Città di Castello, Italy;
| | - Francesca Bazzocchi
- Department of Surgery, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Orhan Alimoglu
- Department of General Surgery, School of Medicine, Istanbul Medeniyet University, 34000 Istanbul, Turkey;
| | - Steven T. Brower
- Department of Surgical Oncology and HPB Surgery, Englewood Hospital and Medical Center, Englewood, NJ 07631, USA;
| | - Graziano Pernazza
- Robotic General Surgery Unit, Department of Surgery, San Giovanni Addolorata Hospital, 00184 Rome, Italy;
| | - Simone D’Imporzano
- Esophageal Surgery Unit, Tuscany Regional Referral Center for the Diagnosis and Treatment of Esophageal Disease, Medical University of Pisa, 56124 Pisa, Italy;
| | - Juan-Santiago Azagra
- Unité des Maladies de l’Appareil Digestif et Endocrine, Centre Hospitalier de Luxembourg, 1210 Luxembourg, Luxembourg;
| | - Yan-Bing Zhou
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China; (Y.-B.Z.); (S.-G.C.)
| | - Shou-Gen Cao
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China; (Y.-B.Z.); (S.-G.C.)
| | - Eleonora Garofoli
- Medical and Translational Oncology, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (E.G.); (C.M.); (S.B.)
| | - Claudia Mosillo
- Medical and Translational Oncology, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (E.G.); (C.M.); (S.B.)
| | - Francesco Guerra
- Department of General and Emergency Surgery, Division of General and Emergency Surgery, School of Robotic Surgery, Misericordia Hospital of Grosseto, 58100 Grosseto, Italy; (A.C.); (F.G.)
| | - Tong Liu
- Gastrointestinal Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (F.Q.); (T.L.)
| | - Giacomo Arcuri
- Division of Surgery, S. Maria della Misericordia Hospital, 06129 Perugia, Italy;
| | - Paulina González
- Esophagogastric Surgery Unit, Digestive Surgery Department, Hospital Dr. Sotero del Rio, Pontificia Universidad Catolica de Chile, Santiago 8207257, Chile; (E.N.); (P.G.)
| | - Fabio Staderini
- Digestive Surgery Unit, Department of Experimental and Clinical Medicine, “Careggi” Hospital, University of Florence, 50134 Florence, Italy; (F.C.); (F.S.)
| | - Alessandra Marano
- General and Oncologic Surgery Unit, Department of Surgery, Santa Croce e Carle Hospital, 12100 Cuneo, Italy; (F.B.); (A.M.)
| | - Irene Terrenato
- Biostatistics and Bioinformatic Unit, Scientific Direction, IRCCS Regina Elena National Cancer Institute, 00128 Rome, Italy;
| | - Vito D’Andrea
- Department of Surgical Sciences—PhD Program in Advanced Surgical Technologies, Sapienza University of Rome, 00161 Rome, Italy;
| | - Sergio Bracarda
- Medical and Translational Oncology, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (E.G.); (C.M.); (S.B.)
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China; (J.-X.L.); (C.-H.Z.); (C.-M.H.)
| | - Amilcare Parisi
- Department of Digestive Surgery, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (S.T.); (A.P.)
| |
Collapse
|
41
|
Ye Z, Wei S, Zeng Y, Wang Y, Lin Z, Chen S, Xie Y, Zheng Q, Chen L. Prognostic value of preoperative body mass index for diabetic patients with non-metastasis gastric cancer: a single center experience. BMC Surg 2021; 21:320. [PMID: 34372800 PMCID: PMC8351091 DOI: 10.1186/s12893-021-01316-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 08/02/2021] [Indexed: 12/20/2022] Open
Abstract
Aim This study was designed to investigate the prognostic effect of preoperative body mass index (BMI) for Type 2 diabetes mellitus (T2DM) patients with non-metastasis gastric cancer (GC) who underwent D2 gastrectomy. Methods T2DM patients with pT1–4bN0–3bM0 GC were retrospectively collected in Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital from January, 2000 to December, 2010. These patients underwent D2 radical resection of the stomach combined with regional lymphadenectomy. Chi-square test was used to analyze unordered categorical variables and ranked data, followed by Kaplan–Meier analysis as well as Cox regression models to detect risk factors for survival outcomes. In addition, the cut-off point was determined by the X-tile program. All analyses were carried out using survival package of R and SPSS Software. Results A total of 302 T2DM patients with pT1–4bN0–3bM0 GC were collected and analyzed. The cut-off points of BMI, identified by the X-tile program, was 19 kg/m2. Patients with low BMI (< 19 kg/m2) had a higher percentage of advanced T stage (T4a and T4b), more advanced TNM stage (stage IIIA, IIIB and IIIC), and more elevated level of serum carcinoembryonic antigen (CEA), compared to those with high BMI (> 19 kg/m2) (all P < 0.05). In the low BMI subgroup, the 5-year overall survival rate was 39.02%, which was as high as 58.11% in the high BMI subgroup (P < 0.05). In the multivariate Cox regression model revealed that IIIC stage (OR = 3.101), N3b stage (OR = 3.113) were the most important prognostic indicators, followed by pretreatment BMI (OR = 2.136). Conclusion Low preoperative BMI (< 19 kg/m2) was a poor prognostic marker for T2DM patients with pT1–4bN0–3bM0 GC.
Collapse
Affiliation(s)
- Zaisheng Ye
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital, and Fujian Medical University Cancer Hospital, No. 420 Fu-ma Road, Jin-An District, Fuzhou, 350014, China
| | - Shenghong Wei
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital, and Fujian Medical University Cancer Hospital, No. 420 Fu-ma Road, Jin-An District, Fuzhou, 350014, China
| | - Yi Zeng
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital, and Fujian Medical University Cancer Hospital, No. 420 Fu-ma Road, Jin-An District, Fuzhou, 350014, China
| | - Yi Wang
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital, and Fujian Medical University Cancer Hospital, No. 420 Fu-ma Road, Jin-An District, Fuzhou, 350014, China
| | - Zhitao Lin
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital, and Fujian Medical University Cancer Hospital, No. 420 Fu-ma Road, Jin-An District, Fuzhou, 350014, China
| | - Shu Chen
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital, and Fujian Medical University Cancer Hospital, No. 420 Fu-ma Road, Jin-An District, Fuzhou, 350014, China
| | - Yunqing Xie
- Department of Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350014, Fujian, China
| | - Qiuhong Zheng
- Department of Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350014, Fujian, China
| | - Luchuan Chen
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital, and Fujian Medical University Cancer Hospital, No. 420 Fu-ma Road, Jin-An District, Fuzhou, 350014, China.
| |
Collapse
|
42
|
Liao XL, Liang XW, Pang HY, Yang K, Chen XZ, Chen XL, Liu K, Zhao LY, Zhang WH, Hu JK. Safety and Efficacy of Laparoscopic Versus Open Gastrectomy in Patients With Advanced Gastric Cancer Following Neoadjuvant Chemotherapy: A Meta-Analysis. Front Oncol 2021; 11:704244. [PMID: 34422658 PMCID: PMC8377369 DOI: 10.3389/fonc.2021.704244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/19/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Given the expanding clinical applications of laparoscopic surgery and neoadjuvant chemotherapy in advanced gastric cancer treatment, there is an emerging need to summarize the few evidences that evaluated the safety and efficacy of laparoscopic versus open gastrectomy in patients with advanced gastric cancer (AGC) following neoadjuvant chemotherapy (NAC). METHODS From January 1 to 2, 2021, we searched Ovid Embase, PubMed, Cochrane central register Trials (Ovid), and web of science to find relevant studies published in English, and two authors independently performed literature screening, quality assessment of the included studies, data extraction, and data analysis. This study was registered with PROSPERO (CRD42021228845). RESULTS The initial search retrieved 1567 articles, and 6 studies were finally included in the meta-analysis review, which comprised 2 randomized control trials and 4 observational studies involving 288 laparoscopic gastrectomy (LG) and 416 open gastrectomy (OG) AGC patients treated with NAC. For intraoperative conditions, R0 resection rate, blood transfusion, intraoperative blood loss, number of lymph nodes dissected, proximal margin, and distal margin were comparable between LG group and open OG group. For postoperative short-term clinical outcomes, LG has significantly less postoperative complications (OR = 0.65, 95%CI: 0.42-1.00, p = 0.05) and shorter postoperative time to first aerofluxus (WMD = -0.57d, 95%CI: -0.89-0.25, p = 0.0004) than OG, and anastomotic leakage, pulmonary infection, pleural effusion, surgical site infection, thrombosis, intestinal obstruction, peritoneal effusion or abscess formation, postoperative time to first defecation, postoperative time to first liquid diet, and postoperative length of stay were comparable between the two groups. For postoperative survival outcomes, there were no significant differences in disease-free survival (DFS) and overall survival (OS) between the two groups. CONCLUSION The available evidences indicated that LG is an effective and feasible technology for the treatment of AGC patients treated with NAC, and LG patients have much less postoperative complications and faster bowel function recovery than OG patients. SYSTEMATIC REVIEW REGISTRATION PROSPERO database (identifier, CRD42021228845).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jian-Kun Hu
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
43
|
Aiolfi A, Lombardo F, Matsushima K, Sozzi A, Cavalli M, Panizzo V, Bonitta G, Bona D. Systematic review and updated network meta-analysis of randomized controlled trials comparing open, laparoscopic-assisted, and robotic distal gastrectomy for early and locally advanced gastric cancer. Surgery 2021; 170:942-951. [PMID: 34023140 DOI: 10.1016/j.surg.2021.04.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/15/2021] [Accepted: 04/11/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND The role of minimally invasive surgery for the treatment of early and locally advanced gastric cancer remains controversial. The purpose of this study was to perform a comprehensive evaluation of major surgical approaches for operable distal gastric cancer. METHODS Systematic review and network meta-analyses of randomized controlled trials were performed to compare open distal gastrectomy, laparoscopic-assisted distal gastrectomy, and robotic distal gastrectomy. Risk ratio, weighted mean difference, and 95% credible intervals were used as pooled effect size measures. RESULTS Seventeen randomized controlled trials (5,909 patients) were included. Overall, 2,776 (46.8%) underwent open distal gastrectomy, 2,964 (50.1%) laparoscopic-assisted distal gastrectomy, and 141 (3.1%) robotic distal gastrectomy. Among these 3 groups, there were no significant differences in 30-day mortality, anastomotic leak, and overall complications. Compared to open distal gastrectomy, laparoscopic-assisted distal gastrectomy was associated with significantly reduced intraoperative blood loss, early postoperative pain, time to first flatus, and hospital length of stay. Similarly, robotic distal gastrectomy was associated with significantly reduced blood loss and time to first flatus compared to open distal gastrectomy. No differences were found in the total number of harvested lymph nodes, tumor-free resection margins, 5-year overall, and disease-free survival. The subgroup analysis in locally advanced gastric cancer showed trends toward reduced blood loss, time to first flatus, and hospital length of stay with minimally invasive approaches but similar overall and disease-free survival. CONCLUSION Laparoscopic-assisted distal gastrectomy and robotic distal gastrectomy performed by well-trained experienced surgeons, even in the setting of locally advanced gastric cancer, seem associated with improved short-term outcomes with similar overall and disease-free survival compared with open distal gastrectomy.
Collapse
Affiliation(s)
- Alberto Aiolfi
- Department of Biomedical Science for Health, University of Milan, Istitituto Clinico Sant'Ambrogio, Milan, Italy.
| | - Francesca Lombardo
- Department of Biomedical Science for Health, University of Milan, Istitituto Clinico Sant'Ambrogio, Milan, Italy
| | - Kazuhide Matsushima
- Division of Acute Care Surgery, LAC+USC Medical Center, University of Southern California, Los Angeles, CA
| | - Andrea Sozzi
- Department of Biomedical Science for Health, University of Milan, Istitituto Clinico Sant'Ambrogio, Milan, Italy
| | - Marta Cavalli
- Department of Biomedical Science for Health, University of Milan, Istitituto Clinico Sant'Ambrogio, Milan, Italy
| | - Valerio Panizzo
- Department of Biomedical Science for Health, University of Milan, Istitituto Clinico Sant'Ambrogio, Milan, Italy
| | - Gianluca Bonitta
- Department of Biomedical Science for Health, University of Milan, Istitituto Clinico Sant'Ambrogio, Milan, Italy
| | - Davide Bona
- Department of Biomedical Science for Health, University of Milan, Istitituto Clinico Sant'Ambrogio, Milan, Italy
| |
Collapse
|
44
|
Laparoscopic Gastrectomy with D2 Lymphadenectomy for pT1a Adenocarcinoma: Case Report and Literature Review. ARS MEDICA TOMITANA 2021. [DOI: 10.2478/arsm-2020-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Gastric cancer is one of the most common malignancies, associated with a high mortality and morbidity rate, especially in the elderly, the fourth leading cause of cancer mortality in Europe. Although the rate of diagnosis has declined in recent years, the prognosis of the disease and the mortality rate is still quite high. Laparoscopic surgery, accompanied by lymphadenectomy, is a curative method of treatment, used more and more frequently, especially in Eastern countries, both in the incipient disease and in advanced stages of the disease, which has many advantages over classical surgery.
We report the case of an 86-year-old man, who presented for marked physical asthenia, weight loss and severe anemia, in whom we performed laparoscopic distal radical gastrectomy with D2 lymphadenectomy and Roux-en-Y reconstruction. Histopathological examination established the diagnosis of low-grade / well-differentiated tubular adenocarcinoma G1 (WHO) / intestinal carcinoma (Lauren), with invasion in the lamina propria, developed on an intestinal-type adenoma. 21 lymph nodes were taken without tumor invasion.
This case highlights the complexity of laparoscopic intervention, with the proven advantages for the patient, like reduced surgical trauma and pain, with the decrease of postoperative complications, but also the difficulty of the surgical technique which requires advanced laparoscopic skills. Thus, in addition to its proven efficacy in cases of early gastric cancer, the technique can be extended to advanced malignancies, without affecting the oncological safety limit.
Collapse
|
45
|
Impact of Lymph Nodes Examined on Survival in ypN0 Gastric Cancer Patients: a Population-Based Study. J Gastrointest Surg 2021; 25:919-925. [PMID: 32318943 DOI: 10.1007/s11605-020-04579-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/23/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND Our aim was to evaluate the prognostic value of the number of lymph nodes examined (eLNs) on survival in ypN0 gastric cancer (GC) patients, and further to define the optimal number of lymph nodes needed to be examined during radical gastrectomy of ypN0 GC patients. METHODS A total of 1127 ypN0 GC patients during 2004-2015 from the Surveillance, Epidemiology, and End Results (SEER) database were included. The number of eLNs cutoff points that determined the greatest actuarial survival difference was calculated by the X-tile program. Univariate and multivariate analyses were performed to assess the impact of eLNs on overall survival (OS). RESULTS The optimal number of eLNs thresholds was determined to be 15 for ypN0 GC patients. Kaplan-Meier analysis revealed that ypN0 GC patients with ≥ 16 eLNs had a significantly better OS than those with ≤ 15 eLNs (5-year OS; 60.8 vs 45.4%, P < 0.001). Similarly, multivariate Cox analysis revealed that ypN0 GC patients with ≥ 16 eLNs experienced a significantly lower hazard of death than those with ≤ 15 eLNs (adjusted HR; 0.73, 95% CI, 0.60-0.90, P = 0.003). CONCLUSIONS The number of eLNs was an independent predictor of survival for ypN0 GC patients. A minimum of 15 eLNs is recommended as the cutoff point for the evaluation of the quality of postoperative or prognostic stratification in ypN0 GC patients.
Collapse
|
46
|
The development and external validation of a nomogram predicting overall survival of gastric cancer patients with inadequate lymph nodes based on an international database. Int J Clin Oncol 2021; 26:867-874. [PMID: 33788042 DOI: 10.1007/s10147-021-01875-2] [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: 05/28/2020] [Accepted: 01/12/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Inadequate sampling of lymph nodes could lead to stage migration and indicate a poor prognosis for gastric cancer after curative surgery. Some emerging novel predictors and the application of a nomogram could increase the accuracy of survival prediction. METHODS An international database regarding gastric cancer was employed as the primary cohort. The patients with inadequate (< 30) lymph nodes (LN) were analyzed by Cox proportional hazards regression. Based on the selected model, a nomogram was plotted and calibrated against an external validation database. RESULTS A total of 1109 patients were included in the primary cohort, and there were 6584 patients in the validation cohort. There were significant differences regarding the clinical characteristics between the two cohorts. The model containing age, T stages, N stages, metastatic lymph nodes (mLN), and the number of total LN retrieved (TLN) showed superiority over the conventional TNM stages. Harrell's concordance index of the nomogram and TNM stages was 0.744 and 0.717, respectively. The external validation demonstrated a good concordance with the nomogram-predicted survival. CONCLUSIONS The nomogram including age, T stages, N stages, mLN, and TLN had a better accuracy than the conventional TNM staging system in predicting overall survival for gastric cancer patients with inadequate (< 30) LN.
Collapse
|
47
|
Zhu K, Jin H, Li Z, Gao Y, Zhang Q, Liu X, Yu J. The Prognostic Value of Lymph Node Ratio after Neoadjuvant Chemotherapy in Patients with Locally Advanced Gastric Adenocarcinoma. J Gastric Cancer 2021; 21:49-62. [PMID: 33854813 PMCID: PMC8020003 DOI: 10.5230/jgc.2021.21.e5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 02/21/2021] [Accepted: 02/21/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose This study aimed to investigate the prognostic value of lymph node ratio (LNR) in patients with locally advanced gastric cancer who received neoadjuvant chemotherapy. Materials and Methods We retrospectively enrolled gastric cancer patients treated with neoadjuvant chemotherapy and curative surgery at the First Affiliated Hospital of Zhejiang University from 2004 to 2015 as the study cohort. Patients with the same inclusion criteria treated in 2016–2017 were enrolled as the validation cohort. Kaplan-Meier curves were assessed using the log-rank test to analyze the differences in overall survival (OS). Multivariate survival analysis was performed using the Cox proportional hazards model. The areas under the receiver operating characteristic curve of ypN and LNR categories for predicting the actual 3-year OS were compared. Results A total of 265 patients were included in the proposal cohort. The median number of retrieved lymph nodes (rLNs) was 32. The number of positive lymph nodes (pLNs) increased as rLN increased (P=0.037), but the LNR remained relatively constant (P=0.462). The LNR was categorized into 4 groups according to the prognosis: ypNr0, node-negative with rLN>25; ypNr1, node-negative with rLN≤25 or 0<LNR≤0.1; ypNr2, 0.1<LNR≤0.3; and ypNr3, LNR>0.3. In the validation cohort of 43 enrolled patients, there was a clear distinction in OS that significantly (P<0.001) varied depending on the LNR values and LNR was the only independent prognostic factor in multivariate analysis (P<0.001). Conclusions LNR was an independent prognostic factor for survival of patients with gastric cancer after preoperative chemotherapy and might be an alternative predictor for ypN stage.
Collapse
Affiliation(s)
- Kankai Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Hailong Jin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijian Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Gao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Qing Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaosun Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Jiren Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
48
|
Zhao RS, Liu YN, Dai WG, Chen SL, Ye JN, Zhai ET, Cai SR, Chen JH. A Substage Increase in The AJCC Classification System Improves Prognostic Prediction in Stage III Gastric Cancer With Insufficient Lymph Nodes Removed. Front Oncol 2021; 11:624413. [PMID: 33763360 PMCID: PMC7982898 DOI: 10.3389/fonc.2021.624413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/11/2021] [Indexed: 01/03/2023] Open
Abstract
Background The impact of lymph nodes (LNs) removed on the survivals of patients with stage III gastric cancer, especially on that of those who undergo the adjuvant chemotherapy as a compensation for a possibly insufficient lymphadenectomy, is still unclear. Methods Consecutive patients (n = 488) with stage III gastric cancer under R0 curative resection followed by adjuvant chemotherapy were analyzed. The overall survival (OS) was compared between patients with insufficient LNs removed (ILNr, <16 LNs) and sufficient LNs removed (SLNr, ≥16 LNs). Performance of the prediction systems was evaluated using the Likelihood ratio χ2 test, Akaike information criterion (AIC), Harrell’s concordance index (C-index), and area under the receiver operating characteristic curves (AUC). Results The OS of patients were significantly longer in those with SLNr relative to those with ILNr (for stage IIIA, 68.2 vs. 43.2 months, P = 0.042; for stage IIIB, 43.7 vs. 24.9 months, P < 0.001; for stage IIIC, 23.9 vs. 8.3 months, P < 0.001; and for total stage III, 37.7 vs. 21.7 months, P < 0.001). However, the OS were similar between stage IIIA patients with ILNr and stage IIIB patients with SLNr (P = 0.928), between IIIB patients with ILNr and IIIC patients with SLNr (P = 0.962), and IIIC patients with ILNr and stage IV (P = 0.668), respectively. A substage increase in the AJCC classification system, from IIIA to IIIB, from IIIB to IIIC, and from IIIC to IV in patients with ILNr, enhanced the accuracy of prognostic prediction in patients with stage III gastric cancer compared to the current TNM system (Likelihood ratio χ2, 188.6 vs. 184.8; AIC, 4336.4 vs. 4340.6; C-index, 0.695 vs. 0.679, P = 0.002). The ROC curves revealed that the performance of prognostic prediction was better in the new prediction system (AUC = 0.699) compared with the current TNM system (AUC = 0.676). Conclusions ILNr (LNs <16) impairs the long-term outcomes of stage III gastric cancer underwent adjuvant chemotherapy. The status of LNs removal adds values to the current TNM system in prognostic prediction of stage III gastric cancer.
Collapse
Affiliation(s)
- Ri-Sheng Zhao
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Yi-Nan Liu
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Wei-Gang Dai
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Si-Le Chen
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Jin-Ning Ye
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Er-Tao Zhai
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Shi-Rong Cai
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Jian-Hui Chen
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
49
|
Identifying Outlier Hospitals in Gastric Cancer Lymph Node Yield Using the National Cancer Database. J Surg Res 2021; 261:196-204. [PMID: 33450628 DOI: 10.1016/j.jss.2020.11.046] [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: 06/30/2020] [Revised: 10/06/2020] [Accepted: 11/01/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lymph node (LN) yield is a key quality indicator that is associated with improved staging in surgically resected gastric cancer. The National Comprehensive Cancer Network recommends a yield of ≥15 LNs for proper staging, yet most facilities in the United States fail to achieve this number. The present study aimed to identify factors that could affect LN yield on a facility level and identify outlier hospitals. METHODS This was a retrospective review of adults (aged ≥18 y) with gastric cancer (Tumor-Node-Metastasis Stages I-III) who underwent gastrectomy. Data were analyzed from the National Cancer Database (2004-2016). Multivariate analysis identified patient and tumor characteristics, whereas an observed-to-expected ratio of identified outlier hospitals. Facility factors were compared between high and low outliers. RESULTS A total of 26,590 patients were included in this study. Of these patients, only 50.3% had an LN yield ≥15. The multivariate model of patient and tumor characteristics demonstrated a concordance index was 0.684. A total of 1245 facilities were included. There were 198 low outlier LN yield hospitals and 135 high outlier LN yield hospitals (observed-to-expected ratio of 0.42 ± 0.24 versus 1.38 ± 0.19, P < 0.0001). There was a difference in facility type between low and high outliers (P < 0.0001). High LN yield hospitals had a larger surgical volume than low LN yield hospitals (median 8.4 [4.9, 13.5] versus 3.5 [2.4, 5.2]; P < 0.0001). CONCLUSIONS Nearly half of the population exhibited low compliance to National Comprehensive Cancer Network recommendations. Facility-level disparities exist as high yearly surgical volume and academic facility status distinguished high-performing outlier hospitals.
Collapse
|
50
|
Chen X, Feng X, Wang M, Yao X. Laparoscopic versus open distal gastrectomy for advanced gastric cancer: A meta-analysis of randomized controlled trials and high-quality nonrandomized comparative studies. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2020; 46:1998-2010. [PMID: 32758382 DOI: 10.1016/j.ejso.2020.06.046] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/01/2020] [Accepted: 06/29/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Controversy persists about the effects of laparoscopic distal gastrectomy (LDG) versus open distal gastrectomy (ODG) on short-term surgical outcomes and long-term survival within the field of advanced gastric cancer (AGC). METHODS Studies published from January 1994 to February 2020 that compare LDG and ODG for AGC were identified. All randomized controlled trials (RCTs) were included. The selection of high-quality nonrandomized comparative studies (NRCTs) was based on a validated tool (Methodological Index for Nonrandomized Studies, MINORS). The short- and long-term outcomes of both procedures were compared. RESULTS Overall, 30 studies were included in this meta-analysis, which comprised of 8 RCTs and 22 NRCTs involving 16,029 patients (7864 LDGs, 8165 ODGs). The recurrence, 3-year disease-free survival (DFS), 3-year overall survival (OS), and 5-year OS rates for LDG and ODG were comparable. LDG was associated with a lower postoperative complication rate (OR 0.79; P < 0.00001), lower estimated volume of blood loss (WMD -102.21 mL; P < 0.00001), shorter postoperative hospital stay (WMD -1.96 days; P < 0.0001), shorter time to first flatus (WMD -0.54 day; P = 0.0007) and shorter time to first liquid diet (WMD -0.66 day; P = 0.001). The number of lymph nodes retrieved, mortality, intraoperative complications, intraoperative blood transfusion, and time to ambulation were similar. However, LDG was associated with a longer surgical time (WMD 33.57 min; P < 0.00001). CONCLUSIONS LDG with D2 lymphadenectomy is a safe and effective technique for patients with AGC when performed by experienced surgeons at high-volume specialized centers.
Collapse
Affiliation(s)
- Xin Chen
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong Province, PR China; Shantou University Medical College, Shantou, 515041, Guangdong Province, PR China
| | - Xingyu Feng
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong Province, PR China
| | - Muqing Wang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong Province, PR China; School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, PR China
| | - Xueqing Yao
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong Province, PR China; Shantou University Medical College, Shantou, 515041, Guangdong Province, PR China; School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, PR China.
| |
Collapse
|