51
|
Li ZY, Wei B, Zhou YB, Li TY, Li JP, Zhou ZW, She JJ, Qin XG, Hu JK, Li YX, Qian F, Shi Y, Cui H, Tian YL, Gao GM, Gao RZ, Liang CC, Shi FY, Yu LJ, Yang K, Zhang SX, Yu PW, Zhao YL. Long-term oncological outcomes of robotic versus laparoscopic gastrectomy for gastric cancer: multicentre cohort study. Br J Surg 2024; 111:znad435. [PMID: 38215239 DOI: 10.1093/bjs/znad435] [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: 08/26/2023] [Revised: 11/23/2023] [Accepted: 12/13/2023] [Indexed: 01/14/2024]
Abstract
BACKGROUND The aim of this multicentre cohort study was to compare the long-term oncological outcomes of robotic gastrectomy (RG) and laparoscopic gastrectomy (LG) for patients with gastric cancer. METHODS Patients with gastric cancer who underwent radical gastrectomy by robotic or laparoscopic approaches from 1 March 2010 to 31 December 2018 at 10 high-volume centres in China were selected from institutional databases. Patients receiving RG were matched 1 : 1 by propensity score with patients undergoing LG. The primary outcome was 3-year disease-free survival. Secondary outcomes were overall survival and disease recurrence. RESULTS Some 2055 patients who underwent RG and 4309 patients who had LG were included. The propensity score-matched cohort comprised 2026 RGs and 2026 LGs. Median follow-up was 41 (i.q.r. 39-58) months for the RG group and 39 (38-56) months for the LG group. The 3-year disease-free survival rates were 80.8% in the RG group and 79.5% in the LG group (log rank P = 0.240; HR 0.92, 95% c.i. 0.80 to 1.06; P = 0.242). Three-year OS rates were 83.9 and 81.8% respectively (log rank P = 0.068; HR 0.87, 0.75 to 1.01; P = 0.068) and the cumulative incidence of recurrence over 3 years was 19.3% versus 20.8% (HR 0.95, 0.88 to 1.03; P = 0.219), with no difference between groups. CONCLUSION RG and LG in patients with gastric cancer are associated with comparable disease-free and overall survival.
Collapse
Affiliation(s)
- Zheng-Yan Li
- Department of General Surgery, Centre for Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bo Wei
- Department of General Surgery, Chinese PLA General Hospital First Medical Centre, Beijing, China
| | - Yan-Bing Zhou
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tai-Yuan Li
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ji-Peng Li
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhi-Wei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Jun-Jun She
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xin-Gan Qin
- Department of Gastrointestinal Gland Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jian-Kun Hu
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yong-Xiang Li
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Feng Qian
- Department of General Surgery, Centre for Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yan Shi
- Department of General Surgery, Centre for Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hao Cui
- Department of General Surgery, Chinese PLA General Hospital First Medical Centre, Beijing, China
| | - Yu-Long Tian
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Geng-Mei Gao
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rui-Zi Gao
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Cheng-Cai Liang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Fei-Yu Shi
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li-Jun Yu
- Department of Gastrointestinal Gland Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kun Yang
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shang-Xin Zhang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Pei-Wu Yu
- Department of General Surgery, Centre for Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yong-Liang Zhao
- Department of General Surgery, Centre for Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
52
|
Niu Z, Liang D, Guan C, Zheng Y, Meng C, Sun X, Liu Z. External validation of the FAMISH predicting score for early gastric cancer with endoscopic submucosal dissection. Eur J Gastroenterol Hepatol 2024; 36:26-32. [PMID: 37642661 DOI: 10.1097/meg.0000000000002635] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
OBJECTIVE To externally validate the robustness of the FAMISH predicting score designed to estimate the risk of metachronous gastric lesions (MGLs) after endoscopic submucosal dissection (ESD) for early gastric cancer (EGC). METHODS This multicenter, retrospective study included 618 patients with EGC who underwent curative ESD at two tertiary referral teaching hospitals between January 2014 and December 2019. FAMISH score was a composite indicator of age, sex, family history, corpus intestinal metaplasia, synchronous lesions, and H. pylori infection. Discrimination, calibration, and risk stratification of these scores were assessed. Associations between MGL characteristics and FAMISH scores were also explored. RESULTS After a median follow-up period of 60 months, 83 of 618 patients (13.4%) developed MGL. The discrimination ability according to the area under the curve was 0.708 (95% CI, 0.645-0.772) for predicting the 5-year MGL. The calibration results showed good consistency between the predicted and actual MGL (Hosmer-Lemeshow, P > 0.05). In terms of risk stratification, the 5-year MGL rates were 4.1% (95% CI, 1.6%-6.5%), 10.8% (95% CI, 7.2%-14.3%), and 32.1% (95% CI, 20.9%-41.7%) in the low-, intermediate-, and high-risk groups, respectively ( P < 0.001). For patients with MGL, the curative resection rate of ESD was significantly higher in the low- and intermediate-risk groups than in the high-risk group (100% vs. 80%, P = 0.037). CONCLUSION The FAMISH predicting score was externally validated and can be generalized to an independent patient population. This adjuvant tool can assist in individual clinical decision-making.
Collapse
Affiliation(s)
| | | | - Chaoyong Guan
- Department of Surgery and Anesthesiology, Xingtai Third Hospital
| | - Yang Zheng
- Department of Gastroenterology, Xingtai Third Hospital
| | | | - Xiaofang Sun
- Endoscopy Room, Xingtai People's Hospital, Xingtai, China
| | | |
Collapse
|
53
|
Zhang L, Huang L, Liu Z, Ling T. Immune Checkpoint Inhibitor Plus Chemotherapy as First-Line Treatment for Advanced Gastric or Gastroesophageal Junction Cancer: A Systematic Review and Meta-Analysis. Technol Cancer Res Treat 2024; 23:15330338241273286. [PMID: 39110075 PMCID: PMC11307348 DOI: 10.1177/15330338241273286] [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: 01/22/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
Background: Immune checkpoint inhibitor (ICI) plus chemotherapy is effective in advanced gastric or gastroesophageal junction (G/GEJ) cancer. This study aims to evaluate the clinical effect of first-line immunotherapy in combination with chemotherapy for advanced G/GEJ cancer. Methods: PubMed, Web of Science, Embase and Cochrane databases were systematically searched from the inception of the databases to December 2021. Randomized trials comparing ICI plus chemotherapy with chemotherapy in first-line treatment for advanced G/GEJ cancer were included. The outcomes were overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and adverse events (AEs). Analyses were performed in Stata 14.0 software. The study protocol was registered with PROSPERO, number CRD42022300907. Results: Five trials were included for analysis, involving 2, 814 patients. ICI plus chemotherapy can significantly improve OS (hazards ratio [HR], 0.86; 95% CI 0.78-0.94; P = .002), PFS (HR, 0.79; 95% CI 0.63-0.99; P < .001) and ORR (relative ratio [RR], 1.20; 95% CI 1.11-1.30; P < .001). In safety analyses, there were no significant differences in incidence of all AEs, treatment-related adverse event (TRAE), TRAE of grade 3 or higher, serious TRAE and TRAE leading to death between two arms (P > .05). Conclusions: ICI plus chemotherapy is more effective first-line treatment for advanced G/GEJ cancer in contrast to chemotherapy regrading to improving OS, PFS and ORR, without increasing TRAE risk. This study will redefine the role of ICI in combination with chemotherapy in the first-line setting for G/GEJ cancer, and provide reference for clinical treatment.
Collapse
Affiliation(s)
- Lianghui Zhang
- Department of oncology, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| | - Lingli Huang
- Department of Pharmacy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhixian Liu
- Department of Pharmacy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Ling
- Department of Pharmacy, Suqian First Hospital, Suqian, China
| |
Collapse
|
54
|
Chen WS, Huang ZX, Zhang HH, Chen XD, Cai YQ, Chen WJ, Zhu GB, Huang YS. Lactate Dehydrogenase and Risk of Readmission with Gastric Cancer: A Propensity Score Matching Analysis. J INVEST SURG 2023; 36:2172488. [PMID: 36775654 DOI: 10.1080/08941939.2023.2172488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/18/2023] [Indexed: 02/14/2023]
Abstract
PURPOSE Readmission is one of the measures of quality of care and potential costs. This study aimed to determine whether lactate dehydrogenase (LDH) is associated with an increased risk of 30-day readmission in gastric cancer. METHODS We performed a retrospective study of patients who underwent radical gastrectomy for gastric cancer at our institution between July 2014 and May 2018. Balanced cohorts were created by propensity score matching (PSM) with a 1:1 ratio to generate the elevated LDH (ELDH) group (n = 151) and the low LDH group (Control) (n = 302). To determine the incidence, causes, and risk factors of 30-day readmission, subgroup analyzes were performed and used to develop an efficient prediction model. RESULTS A total of 788 patients met the criteria to be included in the study. The cutoff value for serum LDH was 215.5. After PSM, a total of 302 patients were matched in pairs (ELDH group, n = 151, Control group, n = 151). ELDH levels had a higher risk of readmission (p = 0.005, Odds ratio 3.768, 95% confidence interval 1.493-9.510). The pre-match 30-day readmission rate was 7.2 percent, and common causes of post-match readmission included infection-related symptoms, gastrointestinal symptoms, and gastrointestinal bleeding. CONCLUSIONS Patients with preoperative ELDH levels, postoperative complications, and high preoperative American Society of Anesthesiologists Scores had a higher risk of readmission 30 days after surgery.
Collapse
Affiliation(s)
- Wei-Sheng Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ze-Xin Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hui-Hui Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiao-Dong Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yi-Qi Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Wen-Jing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Guan-Bao Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yun-Shi Huang
- Department of Trauma & Emergency Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
55
|
Huang J, Xu Z, Chen D, Zhou C, Shen Y. Pancancer analysis reveals the role of disulfidptosis in predicting prognosis, immune infiltration and immunotherapy response in tumors. Medicine (Baltimore) 2023; 102:e36830. [PMID: 38206694 PMCID: PMC10754585 DOI: 10.1097/md.0000000000036830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 12/08/2023] [Indexed: 01/13/2024] Open
Abstract
Disulfidptosis has been reported as a novel cell death process, suggesting a therapeutic strategy for cancer treatment. Herein, we constructed a multiomics data analysis to reveal the effects of disulfidptosis in tumors. Data for 33 kinds of tumors were downloaded from UCSC Xene, and disulfidptosis-related genes (DRGs) were selected from a previous study. After finishing processing data by the R packages, the expression and coexpression of DRGs in different tumors were assessed as well as copy number variations. The interaction network was drawn by STRING, and the activity of disulfidptosis was compared to the single-sample gene set enrichment analysis algorithm. Subsequently, the differences in DRGs for prognosis and clinicopathological features were evaluated, and the tumor immune microenvironment was assessed by the TIMER and TISCH databases. Tumor mutation burden, stem cell features and microsatellite instability were applied to predict drug resistance, and the expression of checkpoints was identified for the prediction of immunotherapy. Moreover, the TCIA, CellMiner and Enrichr databases were also utilized for selecting potential agents. Ten DRGs were differentially expressed in tumors, and the plots of coexpression and interaction revealed their correlation. Survival analysis suggested SLC7A11 as the most prognosis-related DRG with the most significant results. Additionally, the comparison also reflected the differences in DRGs in the status of pathologic lymph node metastasis for 5 types of tumors. The tumor immune microenvironment showed commonality among tumors based on immune infiltration and single-cell sequencing, and the analysis of tumor mutation burden, stemness and microsatellite instability showed a mostly positive correlation with DRGs. Moreover, referring to the prediction about clinical treatment, most DRGs can enhance sensitivity to chemotherapeutic agents but decrease the response to immune inhibitors with increasing expression. In this study, a primarily synthetic landscape of disulfidptosis in tumors was established and provided guidance for further exploration and investigation.
Collapse
Affiliation(s)
- Juntao Huang
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Ziqian Xu
- Department of Dermatology, Ningbo First Hospital, Zhejiang University, Zhejiang, China
| | - Dahua Chen
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Chongchang Zhou
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yi Shen
- Centre for Medical Research, Ningbo No.2 Hospital, Ningbo, China
- School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
56
|
Chen Q, Xiao H, Zhang L, You J, Jin Z, Zhang B. Association between adjuvant chemotherapy and survival in stage I gastric cancer patients after curative resection. Gastroenterol Rep (Oxf) 2023; 11:goad070. [PMID: 38058518 PMCID: PMC10697734 DOI: 10.1093/gastro/goad070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/04/2023] [Accepted: 11/14/2023] [Indexed: 12/08/2023] Open
Abstract
Background The efficacy of adjuvant chemotherapy (AC) on survival outcomes of patients with stage I gastric cancer (GC) after curative resection remains controversial. We aimed to determine whether these patients would benefit from AC. Methods This retrospective study included patients with pathologically confirmed stage I GC who underwent curative resection between November 2010 and December 2020. Patients were divided into AC and non-AC groups, then a 1:1 propensity score matching (PSM) analysis was performed to minimize the selection bias. Potential risk factors including age, pN stage, pT stage, lymphovascular invasion, perineural invasion, tumor size, histological type, and carcinoembryonic antigen level were used as matching covariates. The recurrence-free survival (RFS) and disease-specific survival (DSS) were compared between groups using the Kaplan-Meier method. Results A total of 902 consecutive patients were enrolled and 174 (19.3%) patients were treated with AC. PSM created 123 pairs of patients. Before PSM, patients receiving AC had lower 10-year RFS rates (90% vs 94.6%, P = 0.035) than those who did not receive AC; the two groups had similar 10-year DSS rates (93.8% vs 95.0%, P = 0.240). After PSM, there were no statistical differences in the 10-year RFS (90.9% vs 93.0%, P = 0.507) or DSS rates (93.5% vs 93.6%, P = 0.811) between the two groups. Similar results were found in the stage IA and IB subgroups. Moreover, these findings were not affected by AC cycles. Conclusions The addition of AC could not provide survival benefits for patients with stage I GC after surgery and follow-up is thus recommended. However, large-scale randomized clinical trials are required.
Collapse
Affiliation(s)
- Qiuying Chen
- Department of Radiology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, P. R. China
- Graduate College, Jinan University, Guangzhou, Guangdong, P. R. China
| | - Hua Xiao
- Department of Hepatobiliary and Intestinal Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
| | - Lu Zhang
- Department of Radiology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, P. R. China
- Graduate College, Jinan University, Guangzhou, Guangdong, P. R. China
| | - Jingjing You
- Department of Radiology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, P. R. China
- Graduate College, Jinan University, Guangzhou, Guangdong, P. R. China
| | - Zhe Jin
- Department of Radiology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, P. R. China
- Graduate College, Jinan University, Guangzhou, Guangdong, P. R. China
| | - Bin Zhang
- Department of Radiology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, P. R. China
- Graduate College, Jinan University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
57
|
Liu P, Ding P, Wu H, Wu J, Yang P, Tian Y, Guo H, Zhao Q. Prediction of occult peritoneal metastases or positive cytology using CT in gastric cancer. Eur Radiol 2023; 33:9275-9285. [PMID: 37414883 DOI: 10.1007/s00330-023-09854-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 07/08/2023]
Abstract
OBJECTIVE Accurate prediction of preoperative occult peritoneal metastasis (OPM) is critical to selecting appropriate therapeutic regimen for gastric cancer (GC). Considering the clinical practicability, we develop and validate a visible nomogram that integrates the CT images and clinicopathological parameters for the individual preoperative prediction of OPM in GC. METHODS This retrospective study included 520 patients who underwent staged laparoscopic exploration or peritoneal lavage cytology (PLC) examination. Univariate and multivariate logistic regression results were used to screen model predictors and construct nomograms of OPM risk. The performance of the model was detected by using ROC, accuracy, and C-index. The bootstrap resampling method was considered internal validation of the model. The Delong test was used to evaluate the difference in AUC between the two models. RESULTS Grade 2 mural stratification, tumor thickness, and the Lauren classification diffuse were significant predictors of OPM (p < 0.05). The nomogram of these three factors (compared with the original model) showed a higher predictive effect (p < 0.001). The area under the curve (AUC) of the model was 0.830 (95% CI 0.788-0.873), and the internally validated AUC of 1000 bootstrap samples was 0.826 (95% CI 0.756-0.870). The sensitivity, specificity, and accuracy were 76.0%, 78.8%, and 78.3%, respectively. CONCLUSIONS CT phenotype-based nomogram demonstrates favorable discrimination and calibration, and it can be conveniently used for preoperative individual risk rating of OPM in GC. CLINICAL RELEVANCE STATEMENT In this study, the preoperative OPM prediction model based on CT images (mural stratification, tumor thickness) combined with pathological parameters (the Lauren classification) showed excellent predictive ability in GC, and it is also suitable for clinicians to use rather than limited to professional radiologists. KEY POINTS • Nomogram based on CT image analysis can effectively predict occult peritoneal metastasis in gastric cancer (training area under the curve (AUC) = 0.830 and bootstrap AUC = 0.826). • Nomogram model combined with CT features performed better than the original model (established using only clinicopathological parameters) in differentiating occult peritoneal metastasis of gastric cancer.
Collapse
Affiliation(s)
- Pengpeng Liu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Ping'an Ding
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Haotian Wu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Jiaxiang Wu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Peigang Yang
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Yuan Tian
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Honghai Guo
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Qun Zhao
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China.
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China.
| |
Collapse
|
58
|
Rezaul Islam M, Rauf A, Akash S, Kumer A, Hussain MS, Akter S, Gupta JK, Thameemul Ansari L, Mahfoj Islam Raj MM, Bin Emran T, Aljohani AS, Abdulmonem WA, Thiruvengadam R, Thiruvengadam M. Recent perspective on the potential role of phytocompounds in the prevention of gastric cancer. Process Biochem 2023; 135:83-101. [DOI: 10.1016/j.procbio.2023.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
|
59
|
dos Santos SN, Junior DSG, Pereira JPM, Iadocicco NM, Silva AH, do Nascimento T, Dias LAP, de Oliveira Silva FR, Ricci-Junior E, Santos-Oliveira R, Bernardes ES. Development of glycan-targeted nanoparticles as a novel therapeutic opportunity for gastric cancer treatment. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00161-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
AbstractChemotherapy resistance remains a major cause of therapeutic failure in gastric cancer. The combination of genetic material such as interference RNAs (iRNAs) to silence cancer-associated genes with chemotherapeutics has become a novel approach for cancer treatment. However, finding the right target genes and developing non-toxic, highly selective nanocarrier systems remains a challenge. Here we developed a novel sialyl-Tn-targeted polylactic acid—didodecyldimethylammonium bromide nanoparticle (PLA-DDAB) nanoparticles (NPs) loaded with dsRNA targeting ST6GalNac-I and/or galectin-3 genes. Using single photon emission computed tomography (SPECT), we have demonstrated that 99mtechnetium radiolabeled sialyl-Tn-targeted nanoparticles can reach the tumor site and downregulate ST6GalNAc-I and galectin-3 RNA expression levels when injected intravenously. Furthermore, using an in vivo gastric tumor model, these nanoparticles increased the effectiveness of 5-FU in reducing tumor growth. Our findings indicate that cancer-associated glycan-targeted NPs loaded with dsRNA targeting ST6GalNAc-I and/or galectin-3 in combination with standard chemotherapy, have the potential to become a novel therapeutic tool for gastric cancer.
Collapse
|
60
|
Yin Q, Zhang G, Qie P, Han S, Liu L. Total laparoscopic total gastrectomy and distal esophagectomy combined with reconstruction by transhiatal esophagojejunal Roux-en-y mediastinal anastomosis for Siewert II AEG. J Cardiothorac Surg 2023; 18:339. [PMID: 37990247 PMCID: PMC10664637 DOI: 10.1186/s13019-023-02453-5] [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: 05/21/2022] [Accepted: 11/08/2023] [Indexed: 11/23/2023] Open
Abstract
PURPOSE The optimal procedure is still controversial about Siewert type II AEG, We are attempt to explore the efficacy and feasibility of total laparoscopic total gastrectomy and distal esophagectomy combined with reconstruction by transhiatal esophagojejunal Roux-en-y mediastinal anastomosis for Siewert type II AEG. METHOD Data of patients with Siewert type II AEG who received total laparoscopic total gastrectomy and distal esophagectomy combined with reconstruction by transhiatal esophagojejunal Roux-en-y mediastinal anastomosis in the Hebei General Hospital were collected from October 2020 to October 2021, The operation time, surgical blood loss, the number of dissected lymph nodes, duration of drainage tube, the length of stay in ICU, the resume oral feeding time, the length of postoperative hospital stay, postoperative complications and other related indicators of the patients were collected to evaluate the safety and feasibility of this operation. RESULT A total of 17 patients received total laparoscopic total gastrectomy and distal esophagectomy combined with reconstruction by transhiatal esophagojejunal Roux-en-y mediastinal anastomosisin the treatment of Siewert type II AEG were analyzed in our research. The mean operation time was 253 ± 24.8 min (196-347 min); The median surgical blood loss was 250 ml (20-2400 ml); The average number of dissected lymph nodes were 28 ± 4.6 (17-36); The median duration of drainage tube was 5 days (3-7days); The median length of stay in ICU was 18 h(10-34 h); The median time of resume oral feeding was 6 days (5-7days); The median postoperative hospital stay was 11 days (8-15 days). Among the all enrolled patients, one patient underwent the conversion to laparotomy due to the massive intraoperative bleeding, one patient developed anastomotic stenosis at jejunum side-to-side anastomosis on the first month after surgery, there was no case of death during the operation and postoperative anastomotic fistula. All patients achieved R0 resection with an average distance of 6 cm (4-8.5 cm) from the upper margin of the tumor to the resection margin. CONCLUSION The operation of total laparoscopic total gastric and distal esophagectomy combined with reconstruction by transhiatal esophagojejunal Roux-en-y mediastinal anastomosis is technically feasible and sufficiently safe in the treatment of Seiwert type II AEG from our primary clinical experience. This procedure could be one of the alternatives for the radical treatment of Siewert type II AEG.
Collapse
Affiliation(s)
- Qifan Yin
- Thoracic Surgery, Hebei General Hospital, No 348,West He-Ping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China
| | - Guibin Zhang
- Thoracic Surgery, Hebei General Hospital, No 348,West He-Ping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China
| | - Peng Qie
- Thoracic Surgery, Hebei General Hospital, No 348,West He-Ping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China
| | - Shaohui Han
- Thoracic Surgery, Hebei General Hospital, No 348,West He-Ping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China
| | - Lijun Liu
- Thoracic Surgery, Hebei General Hospital, No 348,West He-Ping Road, Xinhua District, Shijiazhuang, 050000, Hebei Province, China.
| |
Collapse
|
61
|
Yu CD, Zhang K. Efficacy and safety of laparoscopic vs open gastrectomy after neoadjuvant therapy for locally advanced gastric cancer. World J Clin Cases 2023; 11:7795-7805. [DOI: 10.12998/wjcc.v11.i32.7795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/10/2023] [Accepted: 10/30/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Laparoscopic gastrectomy (LG) is widely accepted as a minimally invasive approach for the treatment of early gastric cancer. However, its role in locally advanced gastric cancer (LAGC) after neoadjuvant therapy (NAT) remains controversial. This study aimed to compare the efficacy and safety of LG vs open gastrectomy (OG) after NAT for the treatment of LAGC.
AIM To compare the efficacy and safety of LG vs OG after NAT for LAGC.
METHODS We conducted a prospective study of 76 patients with LAGC who underwent NAT followed by LG (n = 38) or OG (n = 38) between 2021 and 2023. The primary endpoint was overall survival (OS), and the secondary endpoints were disease-free survival (DFS), surgical complications, and quality of life (QOL).
RESULTS The two groups had comparable baseline characteristics, with a median follow-up period of 24 mo. The 3-year OS rates in the LG and OG groups were 68.4% and 60.5%, respectively (P = 0.42). The 3-year DFS rates in the LG and OG groups were 57.9% and 50.0%, respectively (P = 0.51). The LG group had significantly less blood loss (P < 0.001), a shorter hospital stay (P < 0.001), and a lower incidence of surgical site infection (P = 0.04) than the OG group. There were no significant differences in other surgical complications between the groups, including anastomotic leakage, intra-abdominal abscess, or wound dehiscence. The LG group had significantly better QOL scores than the OG group regarding physical functioning, role functioning, global health status, fatigue, pain, appetite loss, and body image at 6 months postoperatively (P < 0.05).
CONCLUSION LG after NAT is a viable and safe alternative to OG for the treatment of LAGC, with similar survival outcomes and superior short-term recovery and QOL. LG patients had less blood loss, shorter hospitalizations, and a lower incidence of surgical site infections than OG patients. Moreover, the LG group had better QOL scores in multiple domains 6 mo postoperatively. Therefore, LG should be considered a valid option for patients with LAGC who undergo NAT, particularly for those who prioritize postoperative recovery and QOL.
Collapse
Affiliation(s)
- Chang-Da Yu
- Department of General Surgery, Jiujiang First People’s Hospital, Jiujiang 332000, Jiangxi Province, China
| | - Ke Zhang
- Department of General Surgery, Jiujiang First People’s Hospital, Jiujiang 332000, Jiangxi Province, China
| |
Collapse
|
62
|
Koo JH, Lee M, Kim EH, Oh HJ, Lim JS, Hyung WJ, Yoon HI, Jung I, Chung YE. Harmful effect of repetitive intravenous iodinated contrast media administration on the long-term renal function of patients with early gastric cancer. Sci Rep 2023; 13:19448. [PMID: 37945805 PMCID: PMC10636198 DOI: 10.1038/s41598-023-46773-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023] Open
Abstract
This retrospective study investigated whether repetitive exposure to intravenous iodinated contrast media (ICM) affects long-term renal function in patients who undergo curative surgery for early gastric cancer (EGC) collected from the Korean Health Insurance and Review Assessment (HIRA) database. Patients diagnosed with gastric cancer between January 2010 and December 2013 underwent regular computed tomography (CT) scans to monitor for extragastric recurrence. Patients who already had chronic kidney disease (CKD) before cancer diagnosis or had undergone chemotherapy or repeated surgery were excluded. A nested case-control study design was chosen to analyze the effect of repetitive ICM exposure to long-term renal function by comparing patients who developed CKD 2 years after cancer diagnosis and patients who did not. Among 59,971 patients collected according to inclusion and exclusion criteria, 1021 were diagnosed with CKD 2 years after cancer diagnosis. Using 1:5 matching after adjusting for age, sex and date of cancer diagnosis, 5097 control patients were matched to 1021 CKD patients. Conditional logistic regression showed that the number of CTs taken using ICM slightly increased the odds of CKD (odds ratio, 1.080; 95% confidence interval (CI): 1.059, 1.100; P < 0.0001). Thus, the administration of ICM might contribute to chronic renal function impairment.
Collapse
Affiliation(s)
- Ja Ho Koo
- Department of Radiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Myeongjee Lee
- Biostatistics Collaboration Unit, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Hwa Kim
- Biostatistics Collaboration Unit, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyung Jung Oh
- Department of Nephrology, Sheikh Khalifa Specialty Hospital, Ras Al-Khaimah, United Arab Emirates
| | - Joon Seok Lim
- Department of Radiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Woo Jin Hyung
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hong In Yoon
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, South Korea
| | - Inkyung Jung
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea.
| | - Yong Eun Chung
- Department of Radiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
63
|
Guo J, Xiong Z, Yin S, Wen Y, Jin L, Wang C, Chen H, Luo D, Deng Z, Huang D, Li X, Yi B, Mao C, Lian L. Elderly patients with stage II gastric cancer do not benefit from adjuvant chemotherapy. World J Surg Oncol 2023; 21:319. [PMID: 37821872 PMCID: PMC10566074 DOI: 10.1186/s12957-023-03185-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/17/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND With the aging of the population, the burden of elderly gastric cancer (EGC) increases worldwide. However, there is no consensus on the definition of EGC and the efficacy of adjuvant chemotherapy in patients with stage II EGC. Here, we investigated the effectiveness of adjuvant chemotherapy in defined EGC patients. METHODS We enrolled 5762 gastric cancer patients of three independent cohorts from the Sixth Affiliated Hospital of Sun Yat-sen University (local), the Surveillance, Epidemiology, and End Results (SEER), and the Asian Cancer Research Group (ACRG). The optimal age cutoff for EGC was determined using the K-adaptive partitioning algorithm. The defined EGC group and the efficacy of adjuvant chemotherapy for them were confirmed by Cox regression and Kaplan-Meier survival analyses. Furthermore, gene set variation analyses (GSVA) were performed to reveal pathway enrichment between groups. RESULTS The optimal age partition value for EGC patients was 75. In the local, SEER, and ACRG cohorts, the EGC group exhibited significantly worse overall survival and cancer-specific survival than the non-EGC group (P < 0.05) and was an independent risk factor. Stratified analyses based on chemotherapy showed that EGC patients derived little benefit from adjuvant chemotherapy. Furthermore, GSVA analysis revealed the activation of DNA repair-related pathways and downregulation of the p53 pathway, which may partially contribute to the observed findings. CONCLUSION In this retrospective, international multi-center study, 75 years old was identified as the optimal age cutoff for EGC definition, and adjuvant chemotherapy proved to be unbeneficial for stage II EGC patients.
Collapse
Affiliation(s)
- Jianping Guo
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhizhong Xiong
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shi Yin
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yue'e Wen
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Longyang Jin
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Caiqin Wang
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Huaxian Chen
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dandong Luo
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zijian Deng
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dayin Huang
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xianzhe Li
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Biying Yi
- Follow-up office of the Sixth Affiliated Hospital, The Sixth Affiliated Hospital, Sun Yat-sen University, Guanzhou, China
| | - Chaobin Mao
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lei Lian
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
64
|
Su P, Jiang L, Zhang Y, Yu T, Huang H, Chen M, Cao C, Kang W, Liu Y, Yu J. Perioperative chemotherapy versus adjuvant chemotherapy treatment for resectable locally advanced gastric cancer: a retrospective cohort study. Eur J Med Res 2023; 28:409. [PMID: 37814327 PMCID: PMC10563233 DOI: 10.1186/s40001-023-01400-3] [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: 05/20/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NAC) is increasingly used in locally advanced gastric cancer (LAGC), but the clinical safety and efficacy are still controversial. This study aims to compare perioperative chemotherapy (PEC) with adjuvant chemotherapy (AC) for resectable LAGC. METHODS Patients who underwent D2 gastrectomy for resectable LAGC were retrospectively reviewed, and divided into NSA group (NAC plus surgery and AC) and SA group (surgery followed by AC). The baseline characteristics and perioperative data were compared. Survival analysis was based on Kaplan-Meier method. Multivariate analyses for prognostic factors were based on the Cox regression. RESULTS A total of 450 patients were eligible for this study. 218 patients received NAC plus surgery and AC, while 232 upfront surgery followed by AC. The baseline characteristics were comparable between the two groups. NSA group showed significant superiority in R0 resection rate (P = 0.014), excised tumor size (P = 0.038), and tumor downstage (all P < 0.001). NAC did not affect postoperative complications or AC-related grade 3/4 adverse events. Patients in NSA group achieved significantly longer OS (P = 0.021) and DFS (P = 0.002). The Cox regression model showed that NAC was independently associated with better OS (HR 0.245, P = 0.039) and DFS (HR 0.591, P = 0.031). CONCLUSIONS Compared with SA, the administration of NSA was considered safe and feasible for achieving higher R0 resection rate without increasing the postoperative complications or AC-related grade 3/4 adverse events, and NAC was independently associated with better OS and DFS for resectable LAGC.
Collapse
Affiliation(s)
- Pengfei Su
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lin Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yingjing Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tian Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hongyun Huang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Moxi Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Can Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Weiming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yuqin Liu
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Jianchun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
65
|
Zhou X, Meng F, Xiao L, Shen H. CYP19A1 promotes gastric cancer as part of a lipid metabolism-related gene signature related to the response of immunotherapy and prognosis. BMC Med Genomics 2023; 16:228. [PMID: 37784135 PMCID: PMC10544546 DOI: 10.1186/s12920-023-01664-y] [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: 05/19/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Increasing evidence suggests that the metabolism of lipids plays a crucial role in the progression of gastric cancer. However, the expression of lipid metabolism-related genes (LMGs) still does not serve as a prognostic biomarker in gastric cancer. METHODS We obtained transcriptome data for 751 LMGs and divided STAD patients into two subtypes based on differences in LMGs expression. Then, we analyzed genetic changes in two subtypes as well as immune features to determine their differences. We also constructed a prognostic risk model related to LMGs for individualized comprehensive evaluations. RESULTS In this study, two lipid metabolic (LM) subtypes were identified anchored in the expression profiles of LMGs. Clinical information, genomic alterations, immune features, and immunotherapy response varied significantly between the two LM subtypes. A risk model based on LMGs was also developed to assess prognosis and distinguish patients with high risk from those at low risk. The prognosis differed significantly between the two risk groups of patients. In STAD patients, the risk score was strongly correlated with genomic alterations and immune profile scores. Also, the risk score was an excellent predictor of immune checkpoint inhibitors (ICIs) response. Anchored in preliminary results derived from the aforementioned bioinformatic analysis, we chose CYP19A1 as our target gene and the expression of CYP19A1 was verified in several common gastric cancer cell lines. Then, we carried out the Western blotting, CCK-8 assay, colony formation assay, wound healing assay, and transwell assay to explore the effects of CYP19A1 on malignant biological behavior, and positive consequences were obtained. CONCLUSIONS In this study, STAD patients were divided into two subtypes based on LMGs expression. It is possible to assess the prognosis of a patient and the response to immunotherapy using the established prognostic risk model. A series of basic laboratory experiments also verified the functional role of CYP19A1 in gastric cancer.
Collapse
Affiliation(s)
- Xinyi Zhou
- Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, NanjingJiangsu Province, 210019, China
| | - Fanyu Meng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Linmei Xiao
- Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, NanjingJiangsu Province, 210019, China
| | - Hua Shen
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
66
|
Meng F, Yang Y, Wang X, Cai F, Liang H, Zhang R, Deng J. Imaging lymph node regression can predict the pathological tumor regression grade in gastric cancer after neoadjuvant treatment. Surgery 2023; 174:836-843. [PMID: 37562986 DOI: 10.1016/j.surg.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 05/24/2023] [Accepted: 07/08/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Neoadjuvant chemotherapy has become the standard treatment for locally advanced gastric cancer. The tumor regression grade system is an effective and widely used tool for the evaluation of treatment response to neoadjuvant chemotherapy. However, whether tumor regression grade could be predicted using clinical characteristics is uncertain. METHODS A total of 287 locally advanced gastric cancer patients from 2014 to 2021 were retrospectively included. According to the College of American Pathologists' tumor regression grade system, patients were classified into response group (tumor regression grade 0-1) and non-response group (tumor regression grade 2-3). Associations between clinical characteristics and neoadjuvant chemotherapy response were performed by the logistic regression model. The Kaplan-Meier method was used to estimate the survival. A prediction scoring system was constructed based on the β coefficients of multivariate analysis. The receiver operating characteristic curve and decision curve analysis were used to evaluate the performance of the predictive scoring system. RESULTS Survival analysis showed that patients with tumor regression grades 0 to 1 had significantly better disease-free survival and overall survival than the tumor regression grades 2 to 3. Tumor differentiation, ycT stage, immunotherapy, and lymph node regression were independent predictors of pathological response to neoadjuvant chemotherapy. We further developed a scoring system to predict the tumor regression grade. The receiver operating characteristic and decision curve analysis showed good predictive performance of the scoring system. CONCLUSION Lymph node regression could be used as a predictor for pathological response. We developed a scoring system to predict the treatment response of patients with gastric cancer receiving neoadjuvant chemotherapy. The scoring system based on the predictors could provide guidance for making clinical decisions.
Collapse
Affiliation(s)
- Feiyu Meng
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P. R. China
| | - Yang Yang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, P. R. China
| | - Xinyu Wang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P. R. China
| | - Fenglin Cai
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P. R. China
| | - Han Liang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P. R. China
| | - Rupeng Zhang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P. R. China
| | - Jingyu Deng
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P. R. China.
| |
Collapse
|
67
|
Staderini F, Barbato G, Bottari A, Russo E, Fortuna L, Giudici F, Coratti F, Stacchini L, Indennitate G, Cianchi F. Effects of the learning curve on operative time and lymph node harvesting during robotic gastrectomy. Int J Med Robot 2023; 19:e2522. [PMID: 37125483 DOI: 10.1002/rcs.2522] [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: 11/16/2022] [Revised: 04/11/2023] [Accepted: 04/16/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Gastric cancer is the fifth most frequent cancer globally. The introduction of minimally invasive surgery for gastric cancer aimed at reducing post-operative morbidity and hospital length of stay. Although the role of laparoscopic gastrectomy has been established, robotic gastric surgery has only recently gained popularity. The purpose of this study was to evaluate, with a multidimensional analysis, the learning curve of a single surgeon with extensive experience in laparoscopic gastrectomy. METHODS We prospectively collected data from 104 gastric cancer patients who underwent surgery with a robotic approach from June 2015 to June 2019 by a single surgeon. We performed 21 total gastrectomies (TGs) and 83 subtotal gastrectomies (STGs). A D2 lymphadenectomy was performed in all the patients. Proximal and distal resection margins were tumoour-free in all patients. There were no intraoperative complications, and no conversions occurred. RESULTS The plateau of the learning curve based on harvesting lymph nodes and operative time was not reached for TG. The learning curve of operative time for STG could be divided into three different phases: an early or learning phase from 1 to 27 cases, an intermediate or proficiency phase from 28 to 48 cases, and a late or mastery phase from 49 to 83 cases. The learning curve for harvesting lymph nodes was achieved after 41 cases in the STG group. CONCLUSION This study shows that robotic gastrectomy is a complex procedure with a significant multiphasic learning curve. Nevertheless, the robotic learning curve seems to be more rapid than that of conventional laparoscopy. Most importantly, our results suggest that the robotic technique can provide oncological adequacy in terms of lymph node harvesting even in the very first phase of the learning curve.
Collapse
Affiliation(s)
- Fabio Staderini
- Digestive Surgery Unit, Department of Clinical and Experimental Medicine, Careggi University Hospital, Florence, Italy
| | - Giuseppe Barbato
- Digestive Surgery Unit, Department of Clinical and Experimental Medicine, Careggi University Hospital, Florence, Italy
| | - Andrea Bottari
- Digestive Surgery Unit, Department of Clinical and Experimental Medicine, Careggi University Hospital, Florence, Italy
| | - Edda Russo
- Digestive Surgery Unit, Department of Clinical and Experimental Medicine, Careggi University Hospital, Florence, Italy
| | - Laura Fortuna
- Digestive Surgery Unit, Department of Clinical and Experimental Medicine, Careggi University Hospital, Florence, Italy
| | - Francesco Giudici
- Digestive Surgery Unit, Department of Clinical and Experimental Medicine, Careggi University Hospital, Florence, Italy
| | - Francesco Coratti
- Digestive Surgery Unit, Department of Clinical and Experimental Medicine, Careggi University Hospital, Florence, Italy
| | - Lorenzo Stacchini
- Department of Health Science, University of Florence, Florence, Italy
| | | | - Fabio Cianchi
- Digestive Surgery Unit, Department of Clinical and Experimental Medicine, Careggi University Hospital, Florence, Italy
| |
Collapse
|
68
|
Zheng L, Gan LH, Yao L, Li B, Huang YQ, Zhang FB, Kuang MQ, Fang N. Serum basic fibroblast growth factor and interleukin-1β predict the effect of first-line chemotherapy in patients with advanced gastric cancer. World J Clin Cases 2023; 11:6083-6090. [PMID: 37731570 PMCID: PMC10507556 DOI: 10.12998/wjcc.v11.i26.6083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/12/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND The incidence and mortality rates of gastric cancer in China are the second-highest in the world, and most patients with gastric cancer lose their chance of surgery by the time of their diagnosis. AIM To explore the predictive potential of serum basic fibroblast growth factor and interleukin-1β levels for the effect of first-line chemotherapy in patients with advanced gastric cancer. METHODS From the gastric cancer patients admitted to our hospital from May 2019 to April 2023, 84 patients were selected and randomly and equally assigned to the experimental or control group. The FLOT group received the FLOT chemotherapy regimen (composed of oxaliplatin + calcium folinate + fluorouracil + paclitaxel), while the SOX group received the SOX chemotherapy regimen (composed of oxaliplatin + tiga capsules). The clinical efficacy, tumor marker levels, adverse reactions, and survival rates of the two groups were compared 7 days after the end of the relevant treatments. RESULTS The target effective rate of the FLOT group was 54.76%, which was much higher than that of the SOX group (33.33%; P < 0.05). After treatment, both the groups demonstrated lower levels of cancer antigen (CEA), carbohydrate antigen 199 (CA199), and peptide tissue antigen (TPS). For several patients before treatment (P < 0.05). Third and fourth grades. In terms of adverse reactions, the level of white blood cells in both the groups was lower. Moreover, the incidence of hand-foot skin reactions in these two study groups was lower (P < 0.05), while those of peripheral neuritis, vomiting, diarrhea, and abnormal liver function were significant (P < 0.05). No statistically significant difference was noted between the two groups (P < 0.05). The 1-year survival rate was higher in the FLOT group (P < 0.05). CONCLUSION The FLOT regimen was effective in reducing the serum CEA, CA199, and TPS levels as well as in improving the 1-year survival rate of patients with good tolerability, making it worthy of clinical promotion and application.
Collapse
Affiliation(s)
- Li Zheng
- Department of Gastroenterology, The First Hospital of Nanchang, Nanchang 330008, Jiangxi Province, China
| | - Li-Hong Gan
- Department of Gastroenterology, The First Hospital of Nanchang, Nanchang 330008, Jiangxi Province, China
| | - Ling Yao
- Department of Gastroenterology, The First Hospital of Nanchang, Nanchang 330008, Jiangxi Province, China
| | - Bin Li
- Department of Gastroenterology, The First Hospital of Nanchang, Nanchang 330008, Jiangxi Province, China
| | - Ya-Qin Huang
- Department of Gastroenterology, The First Hospital of Nanchang, Nanchang 330008, Jiangxi Province, China
| | - Fu-Bao Zhang
- Department of Stomatology, The First Hospital of Nanchang, Nanchang 330008, Jiangxi Province, China
| | - Meng-Qi Kuang
- Department of Gastroenterology, The First Hospital of Nanchang, Nanchang 330008, Jiangxi Province, China
| | - Nian Fang
- Department of Gastroenterology, The First Hospital of Nanchang, Nanchang 330008, Jiangxi Province, China
| |
Collapse
|
69
|
Niu S, Liu Y, Li D, Sheng Y, Zhang Y, Li Z, Zhao S, Wang T. Effect of indocyanine green near-infrared light imaging technique guided lymph node dissection on short-term clinical efficacy of minimally invasive radical gastric cancer surgery: a meta-analysis. Front Oncol 2023; 13:1257585. [PMID: 37766867 PMCID: PMC10520705 DOI: 10.3389/fonc.2023.1257585] [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: 07/12/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Objective In recent years, the utilization of indocyanine green near-infrared (ICG NIR) light imaging-guided lymph node dissection in the context of minimally invasive radical gastric cancer has emerged as a novel avenue for investigation. The objective of this study was to assess the influence of employing this technique for guiding lymph node dissection on the short-term clinical outcomes of minimally invasive radical gastric cancer surgery. Methods The present study conducted a comprehensive search for short-term clinical outcomes, comparing the group undergoing ICG NIR light imaging-guided lymph node dissection with the control group, by thoroughly examining relevant literature from the inception to July 2023 in renowned databases such as PubMed, Embase, Web of Science, and Cochrane Library. The primary endpoints encompassed postoperative complications, including abdominal infection, abdominal bleeding, pneumonia, anastomotic fistula, and overall incidence of complications (defined as any morbidity categorized as Clavien-Dindo class I or higher within 30 days post-surgery or during hospitalization). Additionally, secondary outcome measures consisted of the time interval until the initiation of postoperative gas and food intake, as well as various other parameters, namely postoperative hospital stay, operative time, intraoperative blood loss, total number of harvested lymph nodes, and the number of harvested metastatic lymph nodes. To ensure methodological rigor, the Cochrane Collaboration Risk of Bias Tool and the Newcastle-Ottawa Scale (NOS) were employed to assess the quality of the included studies, while statistical analyses were performed using Review Manager 5.4 software and Stata, version 12.0 software. Results A total of 19 studies including 3103 patients were ultimately included (n=1276 in the ICG group and n=1827 in the non-ICG group). In this meta-analysis, the application of ICG near-infrared light imaging in minimally invasive radical gastric cancer surgery effectively improved the occurrence of postoperative Clavien-Dindo grade II or higher complications in patients (RR=0.72, 95% CI 0.52 to 1.00) with a statistically significant P=0.05; in reducing intraoperative blood loss and shortening While reducing intraoperative blood loss and shortening postoperative hospital stay, it could ensure the thoroughness of lymph node dissection in minimally invasive radical gastric cancer surgery (MD=5.575, 95% CI 3.677-7.473) with significant effect size (Z=5.76, p<0.00001). Conclusion The utilization of indocyanine green near-infrared light imaging technology in the context of minimally invasive radical gastric cancer surgery demonstrates notable efficacy in mitigating the occurrence of postoperative complications surpassing Clavien-Dindo grade II, while concurrently augmenting both the overall quantity of lymph node dissections and the identification of positive lymph nodes, all the while ensuring the preservation of surgical safety. Furthermore, the implementation of this technique proves particularly advantageous in the realm of robotic-assisted radical gastric cancer surgery, thus bearing significance for enhancing the short-term prognostic outcomes of patients.
Collapse
Affiliation(s)
- Sen Niu
- Department of General Surgery, Wuxi People’s Hospital Affiliated To Nanjing Medical University, Wuxi, China
| | - Yuan Liu
- Department of General Surgery, Wuxi People’s Hospital Affiliated To Nanjing Medical University, Wuxi, China
| | - Da Li
- Department of General Surgery, Wuxi People’s Hospital Affiliated To Nanjing Medical University, Wuxi, China
| | - Yufan Sheng
- Department of General Surgery, Wuxi People’s Hospital Affiliated To Nanjing Medical University, Wuxi, China
| | - Ye Zhang
- Department of General Surgery, Wuxi People’s Hospital Affiliated To Nanjing Medical University, Wuxi, China
| | - Zengyao Li
- Department of General Surgery, Wuxi People’s Hospital Affiliated To Nanjing Medical University, Wuxi, China
| | - Songyun Zhao
- Department of Neurosurgery, Wuxi People’s Hospital Affiliated To Nanjing Medical University, Wuxi, China
| | - Tong Wang
- Department of General Surgery, Wuxi People’s Hospital Affiliated To Nanjing Medical University, Wuxi, China
| |
Collapse
|
70
|
van Hootegem SJM, de Pasqual CA, Eyck BM, Mostert B, Bradshaw A, Phillips AW, Lagarde SM, Wijnhoven BPL. Clinical impact of routine response assessment after preoperative chemotherapy in patients with gastric cancer. BJS Open 2023; 7:zrad093. [PMID: 37738366 PMCID: PMC10516457 DOI: 10.1093/bjsopen/zrad093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 09/24/2023] Open
Affiliation(s)
| | - Carlo A de Pasqual
- Department of Surgery, Erasmus MC University, Rotterdam, The Netherlands
- General and Upper GI Surgery Division, University Hospital of Verona, Verona, Italy
| | - Ben M Eyck
- Department of Surgery, Erasmus MC University, Rotterdam, The Netherlands
| | - Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Centre, Rotterdam, The Netherlands
| | - Alexander Bradshaw
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle-upon-Tyne, UK
| | - Alexander W Phillips
- Northern Oesophagogastric Unit, Royal Victoria Infirmary, Newcastle-Upon-Tyne, UK
- School of Medical Education, Newcastle University, Newcastle-upon-Tyne, UK
| | - Sjoerd M Lagarde
- Department of Surgery, Erasmus MC University, Rotterdam, The Netherlands
| | - Bas P L Wijnhoven
- Department of Surgery, Erasmus MC University, Rotterdam, The Netherlands
| |
Collapse
|
71
|
Jeong JY, Seo SH, Kim KH, An MS, Baik H, Kang SH, Oh SH. Doxifluridine versus Tegafur/Gimeracil/Oteracil (S-1) as adjuvant chemotherapy for patients with gastric cancer after gastrectomy: A propensity score-matched analysis. Asian J Surg 2023; 46:3656-3662. [PMID: 37002050 DOI: 10.1016/j.asjsur.2023.03.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
INTRODUCTION Doxifluridine (DF), an oral 5-FU prodrug, has been used for various solid cancers due to its efficacy and low toxicity. We aim to evaluate the effect of DF as adjuvant monotherapy in advanced gastric cancer. METHODS We retrospectively reviewed the clinical data of 263 patients with advanced gastric cancer who underwent curative gastrectomy between January 2010 and December 2013 at our institute. Since previous randomized control trials have confirmed the efficacy of S-1 as adjuvant chemotherapy in advanced gastric cancer, we analyzed the oncologic effect and patient compliance of the DF group compared to the S-1 group. After propensity score matching, 48 patients were included in each group. RESULTS There was no significant difference in 5-year overall survival (OS) and 5-year disease-free survival (DFS) between DF and S-1 groups (5-year OS; 77.1% vs 75.0%; p = 0.729, 5-year DFS; 76.6% vs 73.9%; p = 0.748). The completion rates of the DF and S-1 groups were 60.4% and 72.9%, respectively (p = 0.194). The mean relative dose intensity of the DF and S-1 groups were 76.2% and 84.2%, respectively (p = 0.195). After multivariate analysis, the chemotherapy regimen was not a risk factor for OS and DFS, whereas relative dose intensity and pathologic stage were independent prognostic factors. CONCLUSION There was no significant difference in the oncologic effect and patient compliance between DF and S-1 groups. DF could be an alternative option for adjuvant chemotherapy in advanced gastric cancer. In addition, we confirmed that relative dose intensity is an important independent prognostic factor for survival.
Collapse
Affiliation(s)
- Ji Yoon Jeong
- Department of Surgery, Busan Paik Hospital, Inje University, Republic of Korea
| | - Sang Hyuk Seo
- Department of Surgery, Busan Paik Hospital, Inje University, Republic of Korea
| | - Kwang Hee Kim
- Department of Surgery, Busan Paik Hospital, Inje University, Republic of Korea.
| | - Min Sung An
- Department of Surgery, Busan Paik Hospital, Inje University, Republic of Korea
| | - HyungJoo Baik
- Department of Surgery, Busan Paik Hospital, Inje University, Republic of Korea
| | - Sang Hyun Kang
- Department of Surgery, Busan Paik Hospital, Inje University, Republic of Korea
| | - Sang Hoon Oh
- Department of Surgery, Busan Paik Hospital, Inje University, Republic of Korea
| |
Collapse
|
72
|
Fernström A, Kokkola A, Korpela A, Puolakkainen P, Louhimo J. Separating lymph node stations by the surgeon from the gastric cancer specimen improves the quality of nodal status evaluation. World J Surg Oncol 2023; 21:265. [PMID: 37626384 PMCID: PMC10463918 DOI: 10.1186/s12957-023-03146-y] [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: 05/30/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND In gastric cancer (GC), the pN-stage is an important prognostic factor influencing treatment. Along with the depth of invasion of the tumor, the presence of nodal metastases is one of the most important prognostic factors guiding treatment strategies in gastric cancer. Examining a small number of lymph nodes may lead to understaging of the disease; hence, it is essential for the nodal status to be precisely assessed. In this study, we explored whether dissecting lymph node stations into separate samples by the surgeon from the gastric cancer surgical specimen affects the quality of nodal status evaluation and patient outcome. METHODS The clinical data of 130 GC patients treated at the Helsinki University Hospital between 2016 and 2019 was reviewed. The performed operations included 59 total and 71 subtotal gastrectomies. The processing of the surgical specimen before the pathological examination was assessed from the operation records and pathology reports. The association of the number of examined lymph nodes with other variables was assessed, and multivariate survival analysis was performed to explore the independent prognostic factors in disease-specific survival. RESULTS Dissecting lymph node stations into separate specimens before pathological evaluation yielded a significantly greater number of examined lymph nodes compared with a specimen without intervention (median 34.5 vs 21.0, p < 0.001). The pT-stage, the pN-stage, and the extent of lymphadenectomy were identified as independent prognostic factors, whereas dissecting the specimen's lymph node stations did not associate with survival. CONCLUSIONS Dissecting lymph node stations into separate specimens results in a greater number of examined lymph nodes, which has the potential to lead to a more reliable pN-stage assessment.
Collapse
Affiliation(s)
- Aleksi Fernström
- Abdominal Center, Department of Surgery, Helsinki University Hospital and University of Helsinki, Meilahti Hospital, PO Box 440, Stenbäckinkatu 9A, Helsinki, 00029 HUS, Finland
| | - Arto Kokkola
- Abdominal Center, Department of Surgery, Helsinki University Hospital and University of Helsinki, Meilahti Hospital, PO Box 440, Stenbäckinkatu 9A, Helsinki, 00029 HUS, Finland
| | - Akseli Korpela
- Abdominal Center, Department of Surgery, Helsinki University Hospital and University of Helsinki, Meilahti Hospital, PO Box 440, Stenbäckinkatu 9A, Helsinki, 00029 HUS, Finland
| | - Pauli Puolakkainen
- Abdominal Center, Department of Surgery, Helsinki University Hospital and University of Helsinki, Meilahti Hospital, PO Box 440, Stenbäckinkatu 9A, Helsinki, 00029 HUS, Finland
| | - Johanna Louhimo
- Abdominal Center, Department of Surgery, Helsinki University Hospital and University of Helsinki, Meilahti Hospital, PO Box 440, Stenbäckinkatu 9A, Helsinki, 00029 HUS, Finland.
| |
Collapse
|
73
|
Zhao R, Jiang Y, Zhang J, Huang Y, Xiong C, Zhao Z, Huang T, Liu W, Zhou N, Li Z, Luo X, Tang Y. Development and validation of a novel necroptosis-related gene signature for predicting prognosis and therapeutic response in Ewing sarcoma. Front Med (Lausanne) 2023; 10:1239487. [PMID: 37663658 PMCID: PMC10470467 DOI: 10.3389/fmed.2023.1239487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
Ewing sarcoma (ES) is the second most common malignant bone tumor in children and has a poor prognosis due to early metastasis and easy recurrence. Necroptosis is a newly discovered cell death method, and its critical role in tumor immunity and therapy has attracted widespread attention. Thus, the emergence of necroptosis may provide bright prospects for the treatment of ES and deserves our further study. Here, based on the random forest algorithm, we identified 6 key necroptosis-related genes (NRGs) and used them to construct an NRG signature with excellent predictive performance. Subsequent analysis showed that NRGs were closely associated with ES tumor immunity, and the signature was also good at predicting immunotherapy and chemotherapy response. Next, a comprehensive analysis of key genes showed that RIPK1, JAK1, and CHMP7 were potential therapeutic targets. The Cancer Dependency Map (DepMap) results showed that CHMP7 is associated with ES cell growth, and the Gene Set Cancer Analysis (GSCALite) results revealed that the JAK1 mutation frequency was the highest. The expression of 3 genes was all negatively correlated with methylation and positively with copy number variation (CNV). Finally, an accurate nomogram was constructed with this signature and clinical traits. In short, this study constructed an accurate prognostic signature and identified 3 novel therapeutic targets against ES.
Collapse
Affiliation(s)
- Runhan Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yu Jiang
- School of Public Health, Chongqing Medical University, Chongqing, China
| | - Jun Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Chuang Xiong
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Zenghui Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Tianji Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Wei Liu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Nian Zhou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Zefang Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Orthopedics, Qianjiang Central Hospital of Chongqing, Chongqing, China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yongli Tang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
74
|
Lei X, Wang Y, Shan F, Li S, Jia Y, Miao R, Xue K, Li Z, Ji J, Li Z. Efficacy and safety of preoperative immunotherapy alone followed by surgery in the treatment of advanced gastric cancer with MSI-H/dMMR or EBV-positive. J Chin Med Assoc 2023; 86:717-724. [PMID: 37273199 DOI: 10.1097/jcma.0000000000000944] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND At present, there is no consensus on whether preoperative immunotherapy (PIT) without chemotherapy followed by surgery could benefit patients with advanced gastric cancer (AGC). Here, we report a six-case series study to describe the safety and efficacy of PIT plus gastrectomy in patients with AGC. METHODS This study involved six patients with AGC who received PIT and surgery at our center between January 2019 and July 2021. Demographic characteristics, preoperative gastroscope biopsy pathology, surgical tissue pathology, radicalness of tumor resection, surgical safety, and recovery parameters were reported. RESULTS Six patients, including four patients with Epstein-Barr virus (EBV)-positive gastric cancer (GC) and two patients with microsatellite instability-high (MSI-H)/expression deficiency of mismatch repair (dMMR) protein GC, were enrolled in this study. Four patients experienced immunotherapy-related adverse events (irAEs), without severe adverse events (SAEs). Five patients underwent R0 resection, and one patient underwent palliative gastrectomy due to liver and hilar lymph node metastasis. Pathologic responses from the surgical tissue were observed in all patients, including two pathological complete response (pCR). No operative complications or postoperative deaths occurred. Three patients (50%) experienced mild or moderate postoperative complications without severe postoperative complications. All six patients eventually recovered and were discharged. CONCLUSION This study indicated that PIT was effective and tolerant in some patients with MSI-H/dMMR and/or EBV-positive AGC. PIT followed by gastrectomy might be an alternative treatment option for these selected patients.
Collapse
Affiliation(s)
- Xiaokang Lei
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yinkui Wang
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Fei Shan
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Shuangxi Li
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yongning Jia
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Rulin Miao
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Kan Xue
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhemin Li
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiafu Ji
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Ziyu Li
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
75
|
Dong X, Fan J, Xie W, Wu X, Wei J, He Z, Wang W, Wang X, Shen P, Bei Y. Efficacy evaluation of chimeric antigen receptor-modified human peritoneal macrophages in the treatment of gastric cancer. Br J Cancer 2023; 129:551-562. [PMID: 37386139 PMCID: PMC10403530 DOI: 10.1038/s41416-023-02319-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/06/2023] [Accepted: 06/12/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Gastric cancer is one of the most common cancers. Peritoneal carcinomatosis (PC) appears to be the most common pattern of recurrence, and more than half of the GC patients eventually die from PC. Novel strategies for the management of patients with PC are urgently needed. Recently, rapid progress has been made in adoptive transfer therapy by using macrophages as the effector cells due to their capabilities of phagocytosis, antigen presentation, and high penetration. Here, we generated a novel macrophage-based therapy and investigated anti-tumoral effects on GC and potential toxicity. METHODS We developed a novel Chimeric Antigen Receptor-Macrophage (CAR-M) based on genetically modifying human peritoneal macrophages (PMs), expressing a HER2-FcεR1γ-CAR (HF-CAR). We tested HF-CAR macrophages in a variety of GC models in vitro and in vivo. RESULTS HF-CAR-PMs specifically targeted HER2-expressed GC, and harboured the FcεR1γ moieties to trigger engulfment. Intraperitoneal administration of HF-CAR-PMs significantly facilitated the HER2-positive tumour regression in PC mouse model and prolonged the overall survival rate. In addition, the combined use of oxaliplatin and HF-CAR-PMs exhibited significantly augment anti-tumour activity and survival benefit. CONCLUSIONS HF-CAR-PMs could represent an exciting therapeutic option for patients with HER2-positive GC cancer, which should be tested in carefully designed clinical trials.
Collapse
Affiliation(s)
- Xuhui Dong
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing University, 210008, Nanjing, PR China
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Jiqiang Fan
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing University, 210008, Nanjing, PR China
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Wangxu Xie
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing University, 210008, Nanjing, PR China
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Xiang Wu
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Jia Wei
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing University, 210008, Nanjing, PR China
| | - Zhonglei He
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin, Ireland
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin, Ireland
| | - Xueting Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing University, 210008, Nanjing, PR China
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Pingping Shen
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing University, 210008, Nanjing, PR China.
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 210023, Nanjing, China.
- Shenzhen Research Institute of Nanjing University, 518000, Shenzhen, China.
| | - Yuncheng Bei
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing University, 210008, Nanjing, PR China.
| |
Collapse
|
76
|
Tang XH, Wu XL, Gan XJ, Wang YD, Jia FZ, Wang YX, Zhang Y, Gao XY, Li ZY. Using Normalized Carcinoembryonic Antigen and Carbohydrate Antigen 19 to Predict and Monitor the Efficacy of Neoadjuvant Chemotherapy in Locally Advanced Gastric Cancer. Int J Mol Sci 2023; 24:12192. [PMID: 37569566 PMCID: PMC10418931 DOI: 10.3390/ijms241512192] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) are established prognostic biomarkers for patients with gastric cancer. However, their potential as predictive markers for neoadjuvant chemotherapy (NACT) efficacy has not been fully elucidated. METHODS We conducted a retrospective analysis to determine values of CEA and CA19-9 prior to NACT (pre-NACT) and after NACT (post-NACT) in 399 patients with locally advanced gastric cancer (LAGC) who received intended NACT and surgery. RESULTS Among the 399 patients who underwent NACT plus surgery, 132 patients (33.1%) had elevated pre-NACT CEA/CA19-9 values. Furthermore, either pre-NACT or post-NACT CEA /CA19-9 levels were significantly associated with prognosis (p = 0.0023) compared to patients with non-elevated levels. Moreover, among the patients, a significant proportion (73/132, 55.3%) achieved normalized CEA/CA19-9 following NACT, which is a strong marker of a favorable treatment response and survival benefits. In addition, the patients with normalized CEA/CA19-9 also had a prolonged survival compared to those who underwent surgery first (p = 0.0140), which may be attributed to the clearance of micro-metastatic foci. Additionally, the magnitude of CEA/CA19-9 changes did not exhibit a statistically significant prognostic value. CONCLUSIONS Normalization of CEA/CA19-9 is a strong biomarker for the effectiveness of treatment, and can thus be exploited to prolong the long-term survival of patients with LAGC.
Collapse
Affiliation(s)
- Xiao-Huan Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gas-Trointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing 100142, China; (X.-H.T.); (X.-L.W.)
| | - Xiao-Long Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gas-Trointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing 100142, China; (X.-H.T.); (X.-L.W.)
| | - Xue-Jun Gan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gas-Trointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing 100142, China; (X.-H.T.); (X.-L.W.)
| | - Yi-Ding Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gas-Trointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing 100142, China; (X.-H.T.); (X.-L.W.)
| | - Fang-Zhou Jia
- Biological Sample Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yi-Xue Wang
- Biological Sample Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gas-Trointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing 100142, China; (X.-H.T.); (X.-L.W.)
| | - Xiang-Yu Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gas-Trointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing 100142, China; (X.-H.T.); (X.-L.W.)
| | - Zi-Yu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gas-Trointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing 100142, China; (X.-H.T.); (X.-L.W.)
| |
Collapse
|
77
|
Tang R, Chen GF, Jin K, Zhang GQ, Wu JJ, Han SG, Li B, Chao M. Efficacy of continuous gastric artery infusion chemotherapy in relieving digestive obstruction in advanced gastric cancer. World J Gastrointest Oncol 2023; 15:1283-1294. [PMID: 37546554 PMCID: PMC10401462 DOI: 10.4251/wjgo.v15.i7.1283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/17/2023] [Accepted: 06/11/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Obstruction or fullness after feeding is common in gastric cancer (GC) patients, affecting their nutritional status and quality of life. Patients with digestive obstruction are generally in a more advanced stage. Existing methods, including palliative gastrectomy, gastrojejunostomy, endoluminal stent, jejunal nutrition tube and intravenous chemotherapy, have limitations in treating these symptoms.
AIM To analyze the efficacy of continuous gastric artery infusion chemotherapy (cGAIC) in relieving digestive obstruction in patients with advanced GC.
METHODS This study was a retrospective study. Twenty-nine patients with digestive obstruction of advanced GC who underwent at least one cycle of treatment were reviewed at The Second Affiliated Hospital of Zhejiang University School of Medicine. The oxaliplatin-based intra-arterial infusion regimen was applied in all patients. Mild systemic chemotherapy was used in combination with local treatment. The clinical response was evaluated by contrast-enhanced computed tomography using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Digestive tract symptoms and toxic effects were analyzed regularly. A comparison of the Karnofsky Performance Status (KPS) score and Stooler’s Dysphagia Score before and after therapy was made. Univariate survival analysis and multivariate survival analysis were also performed to explore the key factors affecting patient survival.
RESULTS All patients finished cGAIC successfully without microcatheter displacement, as confirmed by arteriography. The median follow-up time was 24 mo (95%CI: 20.24-27.76 mo). The overall response rate was 89.7% after cGAIC according to the RECIST criteria. The postoperative Stooler’s Dysphagia Score was significantly improved. Twenty-two (75.9%) of the 29 patients experienced relief of digestive obstruction after the first two cycles, and 13 (44.8%) initially unresectable patients were then considered radically resectable. The median overall survival time (mOS) was 16 mo (95%CI: 9.32-22.68 mo). Patients who received radical surgery had a significantly longer mOS than other patients (P value < 0.001). Multivariate Cox regression analysis indicated that radical resection after cGAIC, intravenous chemotherapy after cGAIC, and immunotherapy after cGAIC were independent predictors of mOS. None of the patients stopped treatment because of adverse events.
CONCLUSION cGAIC was effective and safe in relieving digestive obstruction in advanced GC, and it could improve surgical conversion possibility and survival time.
Collapse
Affiliation(s)
- Rui Tang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Guo-Feng Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Kai Jin
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Guang-Qiang Zhang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Jian-Jun Wu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Shu-Gao Han
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Bin Li
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Ming Chao
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
78
|
Liang C, Liang Y, Ou B, Yuan L, Yuan S. Clinicopathological and prognostic features of Borrmann type IV gastric cancer versus other Borrmann types: A unique role of signet ring cell carcinoma. Saudi J Gastroenterol 2023; 29:240-250. [PMID: 37470667 PMCID: PMC10445496 DOI: 10.4103/sjg.sjg_469_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/17/2023] [Accepted: 05/25/2023] [Indexed: 06/21/2023] Open
Abstract
Background Evidence specifically comparing the clinicopathology of Borrmann type IV (B-IV) gastric cancer with that of other Borrmann types is insufficient. Methods A total of 3130 patients with advanced gastric cancer who underwent gastrectomy from January 2001 to September 2017 were enrolled in the analysis. Logistic regression and survival analysis methodology were used to investigate factors associated with peritoneal metastasis and overall survival (OS). Results Of the total cohort, 264 (8.43%) patients were B-IV type, 1752 (55.97%) were small-size other Borrmann types, and 1114 (35.59%) were large-size other Borrmann types. Signet ring cell carcinoma (SRC) was more common in B-IV types than in other Borrmann types (33.71% vs 11.42% vs 12.66%, P < 0.001). In B-IV gastric cancers, SRC was significantly associated with peritoneal metastasis (HR = 1.898, 95% CI = 1.112 ~ 3.241, P = 0.019) and poorer OS (HR = 1.492, 95% CI = 1.088 ~ 2.045, P = 0.013) in multivariable analysis. Furthermore, stratified analysis revealed that SRC had worse survival than adenocarcinoma in the B-IV subgroups, with locally advanced stages (stages II ~ III) or negative surgical margins (all P < 0.05). In contrast, SRC failed to be significantly associated with peritoneal metastasis and poor OS in other Borrmann types (all P > 0.05). Conclusion SRC was more common in B-IV gastric cancer than in other Borrmann types. It was significantly associated with peritoneal metastasis and poorer OS in the B-IV type but not in other Borrmann types. As a unique prognostic factor for B-IV gastric cancer, SRC might help evaluate risk stratification and optimize treatment for this entity, especially for patients with locally advanced stages or R0 resection.
Collapse
Affiliation(s)
- Chengcai Liang
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yao Liang
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Biyi Ou
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Lei Yuan
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shuqiang Yuan
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
79
|
Picchetto A, Cinelli L, Bannone E, Baiocchi GL, Morales-Conde S, Casali L, Spinoglio G, Franzini C, Santi C, D'Ambrosio G, Copaescu C, Rollo A, Balla A, Lepiane P, Paganini AM, Detullio P, Quaresima S, Pesce A, Luciano T, Bianchi G, Marescaux J, Diana M. Fluorescence-based sentinel lymph node mapping and lymphography evaluation: results from the IHU-IRCAD-EAES EURO-FIGS registry. Surg Endosc 2023; 37:5472-5481. [PMID: 37043006 DOI: 10.1007/s00464-023-10043-8] [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: 12/17/2022] [Accepted: 03/25/2023] [Indexed: 04/13/2023]
Abstract
BACKGROUND The identification of metastatic lymph nodes is one of the most important prognostic factors in gastrointestinal (GI) cancers. Near-infrared fluorescence (NIRF) imaging has been successfully used in GI tumors to detect the lymphatic pathway and the sentinel lymph node (SLN), facilitating fluorescence image-guided surgery (FIGS) with the purpose to achieve a correct nodal staging. The aim of this study was to analyze the current results of NIRF SLN navigation and lymphography through data collected in the EURO-FIGS registry. METHODS Prospectively collected data regarding patients and ICG-guided lymphadenectomies were analyzed. Additional analyses were performed to identify predictors of metastatic SLN and determinants of fluorescence positivity and nodal metastases outside the boundaries of standard lymphadenectomies. RESULTS Overall, 188 patients were included by 18 surgeons from 10 different centers. Colorectal cancer was the most reported pathology (77.7%), followed by gastric (19.1%) and esophageal tumors (3.2%). ICG was injected with higher doses (p < 0.001) via extraparietal side (63.3%), and with higher volumes (p < 0.001) via endoluminal side (36.7%). Overall, NIRF SLN navigation was positive in 75.5% of all cases and 95.5% of positive SLNs were retrieved, with a metastatic rate of 14.7%. NIRF identification of lymph nodes outside standard lymphatic stations occurred in 52.1% of all cases, 43.8% of which were positive for metastatic involvement. Positive NIRF SLN identification was an independent predictor of metastasis outside standard lymphatic stations (OR = 4.392, p = 0.029), while BMI independently predicted metastasis in retrieved SLNs (OR = 1.187, p = 0.013). Lower doses of ICG were protective against NIRF identification outside standard of care lymphadenectomy (OR = 0.596, p = 0.006), while higher volumes of ICG were predictive of metastatic involvement outside standard of care lymphadenectomy (OR = 1.597, p = 0.001). CONCLUSIONS SLN mapping helps identifying potentially metastatic lymph nodes outside the boundaries of standard lymphadenectomies. The EURO-FIGS registry is a valuable tool to share and analyze European surgeons' practices.
Collapse
Affiliation(s)
- Andrea Picchetto
- Department of General Surgery and Surgical Specialties "Paride Stefanini", Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy.
| | - Lorenzo Cinelli
- Research Institute Against Digestive Cancer (IRCAD), Strasbourg, France
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Bannone
- Research Institute Against Digestive Cancer (IRCAD), Strasbourg, France
- Department of Surgery, Istituto Fondazione Poliambulanza, Brescia, Italy
- Department of Pancreatic Surgery, Verona University, Verona, Italy
| | - Gian Luca Baiocchi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Salvador Morales-Conde
- Unit of Innovation in Minimally Invasive Surgery, Department of General Surgery, University Hospital Virgen del Rocío, University of Sevilla, Seville, Spain
- General and Digestive Unit, Hospital Quironsalud Sagrado Corazon, Seville, Spain
| | | | | | | | | | - Giancarlo D'Ambrosio
- Department of General Surgery and Surgical Specialties "Paride Stefanini", Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | | | | | - Andrea Balla
- UOC of General and Minimally Invasive Surgery, Hospital "San Paolo", Largo Donatori del Sangue 1, 00053, Rome, Civitavecchia, Italy
| | - Pasquale Lepiane
- UOC of General and Minimally Invasive Surgery, Hospital "San Paolo", Largo Donatori del Sangue 1, 00053, Rome, Civitavecchia, Italy
| | - Alessandro M Paganini
- Department of General Surgery and Surgical Specialties "Paride Stefanini", Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | | | - Silvia Quaresima
- Department of General Surgery and Surgical Specialties "Paride Stefanini", Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | | | | | | | - Jacques Marescaux
- Research Institute Against Digestive Cancer (IRCAD), Strasbourg, France
| | - Michele Diana
- Research Institute Against Digestive Cancer (IRCAD), Strasbourg, France
- University of Strasbourg, Strasbourg, France
- Department of General, Digestive, and Endocrine Surgery, University Hospital of Strasbourg, Strasbourg, France
- ICube Laboratory, Photonics for Health, University of Strasbourg, Strasbourg, France
| |
Collapse
|
80
|
Nie H, Wang H, Zhang M, Ning Y, Chen X, Zhang Z, Hu X, Zhao Q, Chen P, Fang J, Wang F. Comprehensive analysis of cuproptosis-related genes in prognosis, tumor microenvironment infiltration, and immunotherapy response in gastric cancer. J Cancer Res Clin Oncol 2023; 149:5453-5468. [PMID: 36462036 DOI: 10.1007/s00432-022-04474-4] [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: 08/31/2022] [Accepted: 11/09/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUNDS Cuproptosis is the most recently identified copper-dependent cell death form that influences tricarboxylic acid (TCA) cycle. However, the relationship between cuproptosis and clinical prognosis, tumor microenvironment infiltration (TME), and response to immunotherapy remains unclear. METHODS Single-sample gene-set enrichment analysis (ssGSEA) was employed to construct cuproptosisScore (cpS) and 1378 gastric cancer (GC) patients from five independent public datasets were classified into high- or low-cpS groups according to the median of cpS. Then the impacts of cuproptosis on tumor microenvironment infiltration (TME), biological function, response to immunotherapy, and clinical prognosis of GC were evaluated. RiskScore and nomogram were constructed using Lasso Cox regression algorithm to validate its predictive capability in GC patients. RESULTS Compared to patients with high cpS, patients with low cpS exhibited poorer prognosis, higher TNM stage, and stronger stromal activation. Meanwhile, the analysis of response to immunotherapy confirmed patients with high cpS could better benefit from immunotherapy and had a better susceptibility to chemotherapeutic drugs. Then, 9 prognosis-related signatures were collected based on differentially expressed genes (DEGs) of cpS groups. Finally, a riskScore model was constructed using the multivariate Cox (multi-Cox) regression coefficients of prognosis-related signatures and had an excellent capability of predicting 1-, 3-, and 5-year survival in GC patients. CONCLUSIONS This study revealed the role of curproptosis in TME, response to immunotherapy, and clinical prognosis in GC, which highlighted the significant clinical implications of curproptosis and provided novel ideas for the therapeutic application of cuproptosis in GC.
Collapse
Affiliation(s)
- Haihang Nie
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center and Key Lab of Intestinal and Colorectal Diseases of Hubei Province, Wuhan, 430071, China
| | - Haizhou Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center and Key Lab of Intestinal and Colorectal Diseases of Hubei Province, Wuhan, 430071, China
| | - Meng Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center and Key Lab of Intestinal and Colorectal Diseases of Hubei Province, Wuhan, 430071, China
| | - Yumei Ning
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center and Key Lab of Intestinal and Colorectal Diseases of Hubei Province, Wuhan, 430071, China
| | - Xiaojia Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center and Key Lab of Intestinal and Colorectal Diseases of Hubei Province, Wuhan, 430071, China
| | - Zhang Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center and Key Lab of Intestinal and Colorectal Diseases of Hubei Province, Wuhan, 430071, China
| | - Xinyi Hu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center and Key Lab of Intestinal and Colorectal Diseases of Hubei Province, Wuhan, 430071, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center and Key Lab of Intestinal and Colorectal Diseases of Hubei Province, Wuhan, 430071, China
| | - Pengfei Chen
- Department of Gastroenterology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China.
| | - Jun Fang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Clinical Center and Key Lab of Intestinal and Colorectal Diseases of Hubei Province, Wuhan, 430071, China.
- Department of Gastroenterology, Renmin Hospital of Huangmei County, Huanggang, 435500, China.
| | - Fan Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Clinical Center and Key Lab of Intestinal and Colorectal Diseases of Hubei Province, Wuhan, 430071, China.
| |
Collapse
|
81
|
Wan D, He L, Guo C, Zhong Z, Yan X, Cao J, Xu Q, Zhang H, Duan B. m6A-related lncRNAs predict prognosis and indicate cell cycle in gastric cancer. Front Genet 2023; 14:1140218. [PMID: 37408779 PMCID: PMC10319253 DOI: 10.3389/fgene.2023.1140218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/11/2023] [Indexed: 07/07/2023] Open
Abstract
Background: N6-methyladenosine (m6A) modification is a common epigenetic methylation modification of RNA, which plays an important role in gastric carcinogenesis and progression by regulating long non-coding RNA (lncRNA). This study is aimed to investigate the potential prognostic signatures of m6A -related lncRNAs in STAD. Methods: The m6A-related lncRNAs with the most significant impact on gastric cancer prognosis in the TCGA database were identified by bioinformatics and machine learning methods. The m6A-related lncRNA prognostic model (m6A-LPS) and nomogram was constructed by Cox regression analysis with the minimum absolute contraction and selection operator (LASSO) algorithm. The functional enrichment analysis of m6A-related lncRNAs was also investigated. The miRTarBase, miRDB and TargetScan databases were utilized to establish a prognosis-related network of competing endogenous RNA (ceRNA) by bioinformatics methods. The correlation of AL391152.1 expressions and cell cycle were experimentally testified by qRT-PCR and flow cytometry. Results: In total, 697 lncRNAs that were identified as m6A-related lncRNAs in GC samples. The survival analysis showed that 18 lncRNAs demonstrated prognostic values. A risk model with 11 lncRNAs was established by Lasso Cox regression, and can predict the prognosis of GC patients. Cox regression analysis and ROC curve indicated that this lncRNA prediction model was an independent risk factor for survival rates. Functional enrichment analysis and ceRNA network revealed that the nomogram was notably associated with cell cycle. qRT-PCR and flow cytometry revealed that downregulation of GC m6A-related lncRNA AL391152.1 could decrease cyclins expression in SGC7901 cells. Conclusion: A m6A-related lncRNAs prognostic model was established in this study, which can be applied to predict prognosis and cell cycle in gastric cancer.
Collapse
Affiliation(s)
- Dong Wan
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lingnan He
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cheng Guo
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zishao Zhong
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Xiaohan Yan
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia Cao
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qinwei Xu
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haibin Zhang
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bensong Duan
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
82
|
Li W, Shao X, Ma F, Wang B, Xue L, Hu H, Kang W, Xiong J, Tian Y. Total Lymph Node Yield Is Associated with Prolonged Survival after Neoadjuvant Chemotherapy in ypN0 Gastric Cancer Patients. Oncol Res Treat 2023; 46:287-295. [PMID: 37302386 DOI: 10.1159/000531436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Lymph node status after neoadjuvant chemotherapy (NAC) plays the main role in predicting the survival of gastric cancer (GC) patients who underwent curative gastrectomy after NAC. NAC can reduce the number of involved lymph nodes. However, it is unknown whether other variables are associated with the survival outcomes for ypN0 GC patients. It is unknown whether lymph node yield (LNY) has prognostic value in ypN0 GC patients treated with NAC plus surgery. METHODS In this retrospective study, we reviewed the data of patients treated with NAC plus gastrectomy and identified those with ypN0 disease. The LNY cut-off was calculated using the X-tile program to determine the greatest actuarial survival difference. Patients were categorized into the downstaged N0 (cN+/ypN0) and natural N0 (cN0/ypN0) groups based on nodal status. Multivariate analysis was used to identify the prognostic factors and the association between LNY and prognosis. RESULTS A total of 211 GC patients with ypN0 status were included. The optimal LNY cut-off was 23. Kaplan-Meier analysis revealed no significant difference in overall survival between the natural and downstaged N0 groups, while ypN0 GC patients with an LNY of ≥24 had significantly longer overall survival than those with an LNY of ≤23. Univariate analysis identified that LNY, cT stage, tumor location, ypT stage, perineural invasion, lymphovascular invasion, tumor size, Mandard tumor regression grade, and extent of gastrectomy were significantly associated with overall survival. Multivariate analysis confirmed that perineural invasion (hazard ratio, 4.246; p < 0.001), lymphovascular invasion (hazard ratio, 2.694; p = 0.048), and an LNY of ≥24 (hazard ratio, 0.394; p = 0.011) were independent prognostic factors. CONCLUSIONS Patients with natural and downstaged ypN0 GC had similar overall survival after NAC. LNY was an independent prognostic factor in these patients, and an LNY of ≥24 predicted prolonged overall survival.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Fuhai Ma
- Department of Surgical Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Bingzhi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Xue
- Department of Pathology, 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
| | - Wenzhe Kang
- 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
| | - Jianping Xiong
- 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
| | - 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
|
83
|
Zhang Y, Tao K, Yu J, Chen C, Zheng Q, Lei S, Zhong X, Liu L, Wang W, Wang Q, Li E, Luo Y, Zhang G, Feng X, Li Y, Wang J. Evaluation of early liquid drinking after radical gastrectomy in gastric cancer: a Chinese multicenter propensity score matching analysis. Gastroenterol Rep (Oxf) 2023; 11:goad029. [PMID: 37304556 PMCID: PMC10250079 DOI: 10.1093/gastro/goad029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 04/14/2023] [Indexed: 06/13/2023] Open
Abstract
Background Enhanced recovery after surgery is used in gastrointestinal surgery. This study aimed to access the effects of early liquid drinking (ELD) on gastrointestinal function recovery in patients with gastric cancer (GC) who underwent radical gastrectomy, as high-quality evidence on the outcomes of ELD after gastrectomy is currently lacking. Methods Clinicopathological data of patients with GC from 11 centers were retrospectively analysed. Clinical outcomes were investigated in 555 patients, including 225 who started drinking liquid within 48 h (ELD group) of surgery and 330 who started drinking liquid after flatus resumption (traditional liquid drinking [TLD] group). Propensity score matching (PSM) analysis was performed using a match ratio of 1:1 and 201 patients were selected from each group for the analysis. Primary outcome was time to first passage of flatus. Secondary outcomes included time to first defecation, post-operative hospitalization days, occurrence of short-term post-operative complications, and hospitalization costs. Results After PSM, baseline characteristics were not significantly different between the two groups. The time to first flatus (2.72 ± 1.08 vs 3.36 ± 1.39 days), first defecation (4.34 ± 1.85 vs 4.77 ± 1.61 days), and post-operative hospital stay (8.27 ± 4.02 vs 12.94 ± 4.43 days) were shorter in the ELD group than in the TLD group (all P < 0.05). The ELD group had lower hospitalization costs than the TLD group ([7.83 ± 2.44 vs 8.78 ± 3.41] × 104 RMB, P = 0.041). No significant differences were observed in the incidence of post-operative complications. Conclusions Compared with TLD, post-operative ELD could promote rapid recovery of gastrointestinal function and reduce hospitalization costs; moreover, ELD does not increase the risk of post-operative complications.
Collapse
Affiliation(s)
| | | | | | - Chao Chen
- Department of Gastrointestinal Surgery, Huizhou Municipal Central Hospital, Huizhou, Guangdong, P. R. China
| | - Quan Zheng
- Department of Gastrointestinal Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, P. R. China
| | - Sanlin Lei
- Department of Gastrointestinal Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, P. R. China
| | - Xiaogang Zhong
- Department of Surgical Treatment of Gastrointestinal Hernia and Fistula, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi, P. R. China
| | - Lixin Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Wei Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Qiang Wang
- Department of Gastrointestinal Surgery, Guangzhou First People’s Hospital, Guangzhou, Guangdong, P. R. China
| | - En Li
- Department of Gastrointestinal Surgery, Meizhou People’s Hospital, Meizhou, Guangdong, P. R. China
| | - Yuwen Luo
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Guanrong Zhang
- lnformation and Statistics Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Xingyu Feng
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P. R. China
- Shantou University Medical College, Shantou, Guangdong, P. R. China
| | - Yong Li
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P. R. China
- Shantou University Medical College, Shantou, Guangdong, P. R. China
| | - Junjiang Wang
- Corresponding author. Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, No. 106 Zhongshan Second Street, Yuexiu District, Guangzhou 510080, Guangdong, P. R. China. Tel: +86-20-83827812;
| |
Collapse
|
84
|
McDonald HG, Harper MM, Hill K, Gao A, Solomon AL, Bailey CJ, Lin M, Barry-Hundeyin M, Cavnar MJ, Mardini SH, Pandalai PJ, Patel RA, Kolesar JM, Rueckert JA, Hookey L, Ropeleski M, Merchant SJ, Kim J, Gao M. Creation of EGD-Derived Gastric Cancer Organoids to Predict Treatment Responses. Cancers (Basel) 2023; 15:3036. [PMID: 37296998 PMCID: PMC10252567 DOI: 10.3390/cancers15113036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Gastric adenocarcinoma (GAd) is the third leading cause of cancer-related deaths worldwide. Most patients require perioperative chemotherapy, yet methods to accurately predict responses to therapy are lacking. Thus, patients may be unnecessarily exposed to considerable toxicities. Here, we present a novel methodology using patient-derived organoids (PDOs) that rapidly and accurately predicts the chemotherapy efficacy for GAd patients. Methods:Endoscopic GAd biopsies were obtained from 19 patients, shipped overnight, and PDOs were developed within 24 h. Drug sensitivity testing was performed on PDO single-cells with current standard-of-care systemic GAd regimens and cell viability was measured. Whole exome sequencing was used to confirm the consistency of tumor-related gene mutations and copy number alterations between primary tumors, PDOs, and PDO single-cells. Results:Overall, 15 of 19 biopsies (79%) were appropriate for PDO creation and single-cell expansion within 24 h of specimen collection and overnight shipment. With our PDO single-cell technique, PDOs (53%) were successfully developed. Subsequently, two PDO lines were subjected to drug sensitivity testing within 12 days from initial biopsy procurement. Drug sensitivity assays revealed unique treatment response profiles for combination drug regimens in both of the two unique PDOs, which corresponded with the clinical response. Conclusions:The successful creation of PDOs within 24 h of endoscopic biopsy and rapid drug testing within 2 weeks demonstrate the feasibility of our novel approach for future applications in clinical decision making. This proof of concept sets the foundation for future clinical trials using PDOs to predict clinical responses to GAd therapies.
Collapse
Affiliation(s)
- Hannah G. McDonald
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Megan M. Harper
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Kristen Hill
- College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Anqi Gao
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Angelica L. Solomon
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Charles J. Bailey
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Miranda Lin
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Mautin Barry-Hundeyin
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Michael J. Cavnar
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Samuel H. Mardini
- Division of Gastroenterology, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Prakash J. Pandalai
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Reema A. Patel
- Division of Medical Oncology, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jill M. Kolesar
- College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Justin A. Rueckert
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Lawrence Hookey
- Division of Gastroenterology, Department of Internal Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Mark Ropeleski
- Division of Gastroenterology, Department of Internal Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Shaila J. Merchant
- Division of General Surgery and Surgical Oncology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Joseph Kim
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| | - Mei Gao
- Division of Surgical Oncology, Department of General Surgery, University of Kentucky, Lexington, KY 40536, USA; (H.G.M.); (M.G.)
| |
Collapse
|
85
|
Gao Z, Chen X, Bai D, Fahmy L, Qin X, Peng Y, Ren M, Tian Y, Hu J. A Novel Intraoperative Leak Test Procedure (GAM Procedure) to Prevent Postoperative Anastomotic Leakage in Gastric Cancer Patients Who Underwent Gastrectomy. Surg Laparosc Endosc Percutan Tech 2023; 33:224-230. [PMID: 37172005 DOI: 10.1097/sle.0000000000001171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 10/21/2022] [Indexed: 05/14/2023]
Abstract
BACKGROUND Anastomotic leakage following a radical gastrectomy is a serious complication of gastric cancer and esophagogastric junction cancer. The benefit of intraoperative leak testing for the prevention of postoperative anastomotic leakage has been controversial. We introduce a new procedure, which combines the techniques of gastroscopy, air, and methylene blue (GAM) for intraoperative leakage testing. Our objective was to evaluate the efficacy and safety of the GAM procedure for intraoperative leak testing and to compare the surgical complications of gastric cancer patients who underwent gastrectomy with and without intraoperative leak testing using the GAM procedure. MATERIALS AND METHODS A total of 210 patients who underwent radical gastrectomy for gastric cancer were included. Patients were divided into 2 groups: the intraoperative leak testing group using the GAM procedure (IOLT), and the group for which no intraoperative leak testing was done (NIOLT). Clinical and pathologic characteristics, the incidence of postoperative anastomotic leakage, and other surgical complications were compared between the 2 groups. RESULTS There were 82 patients in the IOLT group and 82 patients in the NIOLT group after propensity score matching. In the IOLT group, 4 (4.9%) patients were found to have anastomotic discontinuity during the operation; we repaired these anastomotic discontinuities intraoperatively. The incidence of postoperative anastomotic leakage was higher in the NIOLT group compared with the IOLT group, 6 (7.3%) versus 0 (0%), respectively ( P =0.01). The average time of the GAM procedure was 4.99±1.75 minutes. The surgical time was prolonged by 30 minutes in the IOLT group compared with the NIOLT group, 302.2±79.9 versus 272.1±85.2, respectively ( P =0.02). The length of hospital stay, 15.80±4.55 versus 17.00±6.20 ( P =0.16) was reduced in the IOLT group compared with the NIOLT group. The logistic regression model suggested that IOLT, sex, age, Eastern Cooperative Oncology Group, cT stage, tumor diameter, pT stage, pN stage, and Lauren classification were not risk factors for postoperative complication. CONCLUSIONS The GAM procedure of intraoperative leakage testing can effectively reduce the incidence of postoperative anastomotic leakage in gastric cancer patients undergoing gastrectomy.
Collapse
Affiliation(s)
- Zhenguo Gao
- Department of General Surgery, The Affiliated Nanchong Central Hospital of North Sichuan Medical College (University), Nanchong
| | - Xin Chen
- Department of General Surgery, The First People's Hospital Shuangliu District, Chengdu[West China (Airport) Hospital Sichuan University], Chengdu, Sichuan, China
| | - Dan Bai
- Department of General Surgery, The Affiliated Nanchong Central Hospital of North Sichuan Medical College (University), Nanchong
| | - Lara Fahmy
- Department of Radiology, Wayne State University, Detroit, MI
| | - Xiangzhi Qin
- Department of General Surgery, The Affiliated Nanchong Central Hospital of North Sichuan Medical College (University), Nanchong
| | - Yong Peng
- Department of General Surgery, The Affiliated Nanchong Central Hospital of North Sichuan Medical College (University), Nanchong
| | - Mingyang Ren
- Department of General Surgery, The Affiliated Nanchong Central Hospital of North Sichuan Medical College (University), Nanchong
| | - Yunhong Tian
- Department of General Surgery, The Affiliated Nanchong Central Hospital of North Sichuan Medical College (University), Nanchong
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI
| |
Collapse
|
86
|
Cheng R, Peng Y, Sun X, Zhang S, Li P. Circulating Tumor Cells as Diagnostic Markers of Early Gastric Cancer and Gastric Precancerous Lesions. Oncology 2023; 101:512-519. [PMID: 37263263 DOI: 10.1159/000531323] [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: 11/28/2022] [Accepted: 04/21/2023] [Indexed: 06/03/2023]
Abstract
INTRODUCTION Circulating tumor cells (CTCs) may be potential diagnostic biomarkers of various malignancies including gastric cancer. This study aimed to evaluate whether CTCs could be used to facilitate the diagnosis of early gastric cancer (EGC) or precancerous gastric lesions. METHODS The diagnostic study included consecutive patients with EGC, gastric precancerous lesions, or fundic gland polyps admitted to the Gastroenterology Department, Beijing Friendship Hospital Affiliated to Capital Medical University (National Center for Digestive Diseases) between October 2016 and January 2018. RESULTS A total of 92 patients were enrolled, including 57 patients with EGC, 14 patients with gastric precancerous lesions, and 21 patients with fundic gland polyps (control group). CTCs were detected in 47.89% (34/71) of patients with EGC/gastric precancerous lesions and 4.76% (1/21) of patients with fundic gland polyps (p < 0.001). CTC detection distinguished EGC/precancerous lesions from fundic gland polyps with an area under the receiver operating characteristic curve of 0.740 (95% confidence interval, 0.640-0.840; p = 0.001), a sensitivity of 49.10%, a specificity of 95.00%, a positive predictive value of 97.00%, and a negative predictive value of 64.90%. CONCLUSIONS Peripheral blood CTCs are more common in patients with EGC or gastric precancerous lesions than in patients with fundic gland polyps. Measurement of CTCs may be a useful tool to aid in the diagnosis of EGC and precancerous lesions.
Collapse
Affiliation(s)
- Rui Cheng
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yang Peng
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Gastroenterology, Suining First People's Hospital, Suining, China
| | - Xiujing Sun
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
87
|
Miró M, Vives R, Farran L, Secanella L, Varela M, Baixeras N, Estremiana F, Bettonica C, Aranda H, Galán M. Utility of Molecular Analysis of Peritoneal Fluid in Staging Laparoscopy of Advanced Esophagogastric Junction and Gastric Cancer Prior to Neoadjuvant Treatment. J Gastrointest Cancer 2023; 54:651-661. [PMID: 35881277 DOI: 10.1007/s12029-022-00846-8] [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: 06/20/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Molecular analysis of peritoneal fluid in staging laparoscopy of gastric cancer is performed to improve the detection of free intraperitoneal tumor cells. Nevertheless, its significance is controversial, especially in patients with negative cytology but positive molecular analysis. The aim of this study was to analyze the sensitivity of molecular analysis and its prognostic value. METHODS A retrospective analysis from April 2011 to October 2019 was performed. Cytology (Cyt) and molecular analysis were analyzed by real-time reverse transcriptase polymerase chain reaction (RT-PCR) of the carcinoembryonic antigen (CEA) and cytokeratin 20 (CK20) tumor makers. RESULTS During the study period, 138 staging laparoscopies were performed. Macroscopic carcinomatosis was found in 12.3%. Of the remaining 87.7%, 9.9% were Cyt + and 11.6% were Cyt- RT-PCR + . Of the latter, 9 responded to chemotherapy and underwent radical surgery. The sensitivity of cytology and molecular analysis was 0.70 and 0.76, respectively (p = 0.67). The 2-year overall survival (OS) of Cyt- RT-PCR + vs. Cyt + was similar (p = 0.1). The 2-year OS of Cyt-RT-PCR + subgroup who underwent radical surgery vs. Cyt-RT-PCR- patients was similar (p = 0.69), but disease-free survival was shorter in the first group (p = 0.005). CONCLUSION Our results show that the sensitivity of molecular analysis is similar to that of cytology. The prognostic value of positive molecular analysis was similar to positive cytology in terms of 2-year overall survival, except in the subgroup of operated patients in whom the overall survival was similar to that of those with a negative molecular analysis, albeit with a shorter disease-free survival.
Collapse
Affiliation(s)
- M Miró
- Unit of Esophagogastric Surgery, General and Digestive Surgery Service, Bellvitge University Hospital, St Feixa Llarga s/n, l'Hospitalet del Llobregat, Barcelona, 08907, Spain.
| | - R Vives
- Unit of Esophagogastric Surgery, General and Digestive Surgery Service, Bellvitge University Hospital, St Feixa Llarga s/n, l'Hospitalet del Llobregat, Barcelona, 08907, Spain
| | - L Farran
- Unit of Esophagogastric Surgery, General and Digestive Surgery Service, Bellvitge University Hospital, St Feixa Llarga s/n, l'Hospitalet del Llobregat, Barcelona, 08907, Spain
| | - L Secanella
- General and Digestive Surgery Service, Bellvitge University Hospital, L'Hospitalet del Llobregat, Barcelona, Spain
| | - M Varela
- Pathology Department, Bellvitge University Hospital, L'Hospitalet del Llobregat, Barcelona, Spain
| | - N Baixeras
- Pathology Department, Bellvitge University Hospital, L'Hospitalet del Llobregat, Barcelona, Spain
| | - F Estremiana
- Unit of Esophagogastric Surgery, General and Digestive Surgery Service, Bellvitge University Hospital, St Feixa Llarga s/n, l'Hospitalet del Llobregat, Barcelona, 08907, Spain
| | - C Bettonica
- Unit of Esophagogastric Surgery, General and Digestive Surgery Service, Bellvitge University Hospital, St Feixa Llarga s/n, l'Hospitalet del Llobregat, Barcelona, 08907, Spain
| | - H Aranda
- Unit of Esophagogastric Surgery, General and Digestive Surgery Service, Bellvitge University Hospital, St Feixa Llarga s/n, l'Hospitalet del Llobregat, Barcelona, 08907, Spain
| | - M Galán
- Medical Oncology Service, Institut Catala d'Oncologia, Gran Via de L'Hospitalet 199-203, L'Hospitalet del Llobregat, Barcelona, 08908, Spain
| |
Collapse
|
88
|
Carmo Bastos ML, Silva-Silva JV, Neves Cruz J, Palheta da Silva AR, Bentaberry-Rosa AA, da Costa Ramos G, de Sousa Siqueira JE, Coelho-Ferreira MR, Percário S, Santana Barbosa Marinho P, Marinho AMDR, de Oliveira Bahia M, Dolabela MF. Alkaloid from Geissospermum sericeum Benth. & Hook.f. ex Miers (Apocynaceae) Induce Apoptosis by Caspase Pathway in Human Gastric Cancer Cells. Pharmaceuticals (Basel) 2023; 16:ph16050765. [PMID: 37242548 DOI: 10.3390/ph16050765] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Gastric cancer is among the major causes of death from neoplasia leading causes of death worldwide, with high incidence rates and problems related to its treatment. Here, we outline how Geissospermum sericeum exerts antitumor activity on the ACP02 cell line (human gastric adenocarcinoma) and the mechanism of cell death. The ethanol extract and fractions, neutral fraction and alkaloid fraction, were characterized by thin-layer chromatography and HPLC-DAD, yielding an alkaloid (geissoschizoline N4-methylchlorine) identified by NMR. The cytotoxicity activity of the samples (ethanol extract, neutral fraction, alkaloid fraction, and geissoschizoline N4-methylchlorine) in HepG2 and VERO cells was determined by MTT. The ACP02 cell line was used to assess the anticancer potential. Cell death was quantified with the fluorescent dyes Hoechst 33342, propidium iodide, and fluorescein diacetate. The geissoschizoline N4-methylchlorine was evaluated in silico against caspase 3 and 8. In the antitumor evaluation, there was observed a more significant inhibitory effect of the alkaloid fraction (IC50 18.29 µg/mL) and the geissoschizoline N4-methylchlorine (IC50 12.06 µg/mL). However, geissoschizoline N4-methylchlorine showed lower cytotoxicity in the VERO (CC50 476.0 µg/mL) and HepG2 (CC50 503.5 µg/mL) cell lines, with high selectivity against ACP02 cells (SI 39.47 and 41.75, respectively). The alkaloid fraction showed more significant apoptosis and necrosis in 24 h and 48 h, with increased necrosis in higher concentrations and increased exposure time. For the alkaloid, apoptosis and necrosis were concentration- and time-dependent, with a lower necrosis rate. Molecular modeling studies demonstrated that geissoschizoline N4-methylchlorine could occupy the active site of caspases 3 and 8 energetically favorably. The results showed that fractionation contributed to the activity with pronounced selectivity for ACP02 cells, and geissoschizoline N4-methylchlor is a promising candidate for caspase inhibitors of apoptosis in gastric cancer. Thus, this study provides a scientific basis for the biological functions of Geissospermum sericeum, as well as demonstrates the potential of the geissoschizoline N4-methylchlorine in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Mirian Letícia Carmo Bastos
- Post-Graduate Program in Biodiversity and Biotechnology, Federal University of Pará, Belém 66075-110, PA, Brazil
- Post-Graduate Program in Pharmaceutical Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - João Victor Silva-Silva
- Laboratory of Medicinal and Computational Chemistry, Institute of Physics of São Carlos, University of São Paulo, São Carlos 13563-120, SP, Brazil
| | - Jorddy Neves Cruz
- Post-Graduate Program in Pharmaceutical Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
| | | | | | - Gisele da Costa Ramos
- Post-Graduate Program in Chemistry, Federal University of Pará, Belém 66075-110, PA, Brazil
| | | | - Márlia Regina Coelho-Ferreira
- Emílio Goeldi Paraense Museum, Coordination of Botany, Ministry of Science, Technology, Innovation and Communications, Belém 66077-830, PA, Brazil
| | - Sandro Percário
- Post-Graduate Program in Biodiversity and Biotechnology, Federal University of Pará, Belém 66075-110, PA, Brazil
| | | | | | - Marcelo de Oliveira Bahia
- Laboratory of Human Cytogenetic, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - Maria Fâni Dolabela
- Post-Graduate Program in Biodiversity and Biotechnology, Federal University of Pará, Belém 66075-110, PA, Brazil
- Post-Graduate Program in Pharmaceutical Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
- Faculty of Pharmacy, Federal University of Pará, Belém 66075-110, PA, Brazil
- Post-Graduate Program in Pharmaceutical Innovation, Federal University of Pará, Belém 66075-110, PA, Brazil
| |
Collapse
|
89
|
Liu W, Huo G, Chen P. Efficacy of PD-1/PD-L1 inhibitors in advanced gastroesophageal cancer based on characteristics: a meta-analysis. Immunotherapy 2023. [PMID: 37190983 DOI: 10.2217/imt-2022-0305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
Objective: Evaluate the potency of anti-PD-1/PD-L1 antibodies in advanced gastroesophageal cancer patients with different clinical features. Methods: Randomized, controlled trials comparing anti-PD-1/PD-L1 antibodies with chemotherapy in individuals with gastroesophageal cancer were retrieved. Results: 15 trials involving 9194 individuals were included. PD-1/PD-L1 inhibitors significantly improved overall survival (OS) but not progression-free survival. Significantly improved OS was observed in PD-L1 combined positive score ≥1, primary esophageal cancer, primary gastric cancer and Asian patients. Subgroup analysis revealed significant OS benefit achieved for esophageal squamous cell carcinoma, but not for esophageal adenocarcinoma. Conclusion: PD-1/PD-L1 inhibitors improved OS in advanced gastroesophageal carcinoma, especially in patients with esophageal cancer. Race, primary tumor sites and PD-L1 combined positive score can be used to predict the potency of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention & Therapy of Tianjin; Tianjin's Clinical Research Center for Cancer; Tianjin, 300060, China
| | - Gengwei Huo
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention & Therapy of Tianjin; Tianjin's Clinical Research Center for Cancer; Tianjin, 300060, China
| | - Peng Chen
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention & Therapy of Tianjin; Tianjin's Clinical Research Center for Cancer; Tianjin, 300060, China
| |
Collapse
|
90
|
Škapars R, Gašenko E, Broza YY, Sīviņš A, Poļaka I, Bogdanova I, Pčolkins A, Veliks V, Folkmanis V, Lesčinska A, Liepniece-Karele I, Haick H, Rumba-Rozenfelde I, Leja M. Breath Volatile Organic Compounds in Surveillance of Gastric Cancer Patients following Radical Surgical Management. Diagnostics (Basel) 2023; 13:diagnostics13101670. [PMID: 37238155 DOI: 10.3390/diagnostics13101670] [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: 03/24/2023] [Revised: 04/20/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
As of today, there is a lack of a perfect non-invasive test for the surveillance of patients for potential relapse following curative treatment. Breath volatile organic compounds (VOCs) have been demonstrated to be an accurate diagnostic tool for gastric cancer (GC) detection; here, we aimed to prove the yield of the markers in surveillance, i.e., following curative surgical management. Patients were sampled in regular intervals before and within 3 years following curative surgery for GC; gas chromatography-mass spectrometry (GC-MS) and nanosensor technologies were used for the VOC assessment. GC-MS measurements revealed a single VOC (14b-Pregnane) that significantly decreased at 12 months, and three VOCs (Isochiapin B, Dotriacontane, Threitol, 2-O-octyl-) that decreased at 18 months following surgery. The nanomaterial-based sensors S9 and S14 revealed changes in the breath VOC content 9 months after surgery. Our study results confirm the cancer origin of the particular VOCs, as well as suggest the value of breath VOC testing for cancer patient surveillance, either during the treatment phase or thereafter, for potential relapse.
Collapse
Affiliation(s)
- Roberts Škapars
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Evita Gašenko
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Yoav Y Broza
- Department of Chemical Engineering and Russel Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Armands Sīviņš
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Inese Poļaka
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Inga Bogdanova
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Andrejs Pčolkins
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Viktors Veliks
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Valdis Folkmanis
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Anna Lesčinska
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Inta Liepniece-Karele
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Hossam Haick
- Department of Chemical Engineering and Russel Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Ingrīda Rumba-Rozenfelde
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Mārcis Leja
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| |
Collapse
|
91
|
Guo X, Gao Y, Song Q, Wei J, Wu J, Dong J, Chen L, Xu S, Wu D, Yang X, Chen L, Li X, Ji G, Lv X, Wei B. Early assessment of circulating exosomal lncRNA-GC1 for monitoring neoadjuvant chemotherapy response in gastric cancer. Int J Surg 2023; 109:1094-1104. [PMID: 37222716 PMCID: PMC10389467 DOI: 10.1097/js9.0000000000000249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/22/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND The timing of surgery for patients with gastric cancer (GC) who undergo neoadjuvant chemotherapy (neoCT) was mainly guided by serial radiologic imaging. However, an earlier assessment was indispensable to avoid delayed treatment for nonresponders and excessive toxicity for responders. Our previous study has identified circulating extracellular vesicles-derived lncRNA-GC1 as a biomarker for early detection and monitoring progression of GC. However, the potential role of neoCT remains poorly understood. METHODS In this explorative biomarker analysis, we conducted a multi-cohort study to examine longitudinal levels of circulating extracellular vesicles-derived lncRNA-GC1 in 798 patients enrolled in the RESONANCE study (NCT01583361). Both circulating extracellular vesicles-derived lncRNA-GC1 and traditional gastrointestinal biomarkers were assessed at defined time nodes. Computed tomography (CT) scans were performed before treatment and 8-10 weeks and assessed based on the RECIST criteria. RESULTS Circulating extracellular vesicles-derived lncRNA-GC1 could be detected in 96.3% of patients at baseline, and significant reductions were observed before cycle 2 (P<0.0001). Levels of circulating extracellular vesicles-derived lncRNA-GC1 showed a stronger correlation with tumor burden and exhibited earlier dynamic changes than the traditional gastrointestinal biomarkers during the first cycle of neoCT. Strong agreement was observed between circulating extracellular vesicles-derived lncRNA-GC1 response (reduction >50%) and radiographic response (Cohen's κ, 0.704). Importantly, circulating extracellular vesicles-derived lncRNA-GC1 maintained predictive value in two external cohorts. Patients with circulating extracellular vesicles-derived lncRNA-GC1 response showed superior disease-free survival [hazard ratio (HR), 0.6238; 95% CI, 0.4095-0.9501; P=0.0118] and overall survival (HR, 0.6131; 95% CI, 0.4016-0.9358; P=0.0090). CONCLUSION Circulating extracellular vesicles-derived lncRNA-GC1 is an early marker of neoCT efficacy and predicts superior survival in GC patients treated with neoCT.
Collapse
Affiliation(s)
- Xin Guo
- Department of Digestive Surgery
- Department of Endoscopic Surgery, Air Force 986th Hospital, Fourth Military Medical University, Xian
- Department of General Surgery, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Yunge Gao
- Department of Gynecology and Obstetrics
| | - Qiying Song
- Department of General Surgery, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | | | | | - Jian Dong
- Department of Gynecology and Obstetrics
| | | | - Shenhui Xu
- Department of Pathology, Xijing Hospital
| | - Di Wu
- Department of General Surgery, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | | | - Lubin Chen
- Department of Digestive Surgery
- Department of Endoscopic Surgery, Air Force 986th Hospital, Fourth Military Medical University, Xian
| | | | - Gang Ji
- Department of Digestive Surgery
| | | | - Bo Wei
- Department of General Surgery, Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
92
|
Gao Y, Xi H, Mattsson F, Liang W, Xie SH, Chen L, Lagergren J. Surgical starting time of the day and survival in gastric cancer. Sci Rep 2023; 13:6955. [PMID: 37117226 PMCID: PMC10147916 DOI: 10.1038/s41598-023-33692-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 04/17/2023] [Indexed: 04/30/2023] Open
Abstract
Previous studies indicate differences in short-term postoperative outcomes depending on the surgical starting time of the day, but long-term data are lacking. The aim of this study was to clarify if surgical starting time of the day influences long-term survival in gastric cancer patients. This cohort study consecutively included 2728 patients who underwent curatively intended gastrectomy for gastric cancer in 2011-2015 at a high-volume hospital in China, with follow-up until June 2019. Cox regression provided hazard ratios (HRs) with 95% confidence intervals (CIs) for 3-year all-cause mortality, adjusted for age, sex, health insurance, pathological tumor stage, surgical approach, neoadjuvant therapy, and weekday of surgery. Compared with patients with early starting time of gastrectomy (08:00-09:29), the point estimates for 3-year all-cause mortality were modestly increased in patients with a starting time in the middle of day (09:30-13:29; HR 1.15, 95% CI 0.97 to 1.37) and later (13:30-21:25; HR 1.10, 0.91 to 1.32). The corresponding HRs were increased particularly in patients who underwent laparoscopic gastrectomy (HR 1.54, 1.10 to 2.14 and HR 1.59, 1.12 to 2.25, respectively) and in those with stage II tumors (HR 1.74, 1.11 to 2.73 and HR 1.60, 1.00 to 2.58, respectively). Our study indicated that in patients who underwent laparoscopic gastrectomy and in those who with stage II tumors, starting surgery in the early morning might be associated with better long-term survival.
Collapse
Affiliation(s)
- Yunhe Gao
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, People's Republic of China
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Retzius väg 13 a, 171 77, Stockholm, Sweden
| | - Hongqing Xi
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, People's Republic of China
| | - Fredrik Mattsson
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Retzius väg 13 a, 171 77, Stockholm, Sweden
| | - Wenquan Liang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, People's Republic of China
| | - Shao-Hua Xie
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Retzius väg 13 a, 171 77, Stockholm, Sweden.
- Institute of Population Medicine and School of Public Health, Fujian Medical University, Fuzhou, People's Republic of China.
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China.
| | - Lin Chen
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, People's Republic of China.
| | - Jesper Lagergren
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Retzius väg 13 a, 171 77, Stockholm, Sweden
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| |
Collapse
|
93
|
Zhang B, Li Y, Zhu X, Chen Z, Huang X, Gong T, Zheng W, Bi Z, Zhu C, Qian J, Li X, Jin C. OncoVee™-MiniPDX-guided anticancer treatment for HER2-negative intermediate-advanced gastric cancer patients: a single-arm, open-label phase I clinical study. Discov Oncol 2023; 14:46. [PMID: 37093368 PMCID: PMC10126180 DOI: 10.1007/s12672-023-00661-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 04/17/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Chemotherapy is the main treatment strategy for patients with advanced HER2-negative gastric cancer (GC); yet, many patients do not respond well to treatment. This study evaluated the sensitivity of a mini patient-derived xenograft (MiniPDX) animal model in patients with HER2-negative intermediate-advanced GC. METHODS In this single-arm, open-label clinical study, we consecutively recruited patients with HER2-negative advanced or recurrent GC from September 2018 to July 2021. Tumor tissues were subjected to MiniPDX drug sensitivity tests for screening individualized anti-tumor drugs; appropriate drug types or combinations were selected based on drug screening results. The primary endpoints were progression-free survival (PFS) and safety, and the secondary endpoints were overall survival (OS) and objective response rate (ORR). RESULTS A total of 17 patients were screened, and 14 eligible patients were included.The median follow-up time was 9 (2-34) months. The median PFS time was 14.1 (2-34) months, the median OS time was 16.9 (2-34) months, ORR was 42.9% (6/14), and DCR was 92.9% (13/14). The most common treatment-related adverse events (TRAE) were fatigue (14 (100%)), anorexia (13 (93%)) and insomnia (12 (86%)), and the most common grade 3 or worse TRAE was fatigue (6 (43%)), and anorexia (6 (43%)). The occurrence rate of myelosuppression, nausea and vomiting, abnormal liver enzymes, and other grade 3-4 chemotherapy adverse reactions were relatively low, and no grade 5 treatment-related adverse events occurred. CONCLUSION Screening HER2-negative medium-advanced GC/GJC chemotherapy regimens and targeted drugs based on MiniPDX animal models showed good tumor activity and safety.
Collapse
Affiliation(s)
- Baonan Zhang
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China
| | - Yuzhen Li
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China
| | - Xiaodan Zhu
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China
| | - Zhe Chen
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China
| | - Xiaona Huang
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China
| | - Tingjie Gong
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China
| | - Weiwang Zheng
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China
| | - Zhenle Bi
- Department of Medical, Co. Ltd. Shanghai, Shanghai LIDE Biotech, China
| | - Chenyang Zhu
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China
| | - Jingyi Qian
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China
| | - Xiaoqiang Li
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China
| | - Chunhui Jin
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, 8 West Zhongnan Road, Wuxi, 214071, China.
| |
Collapse
|
94
|
Cai W, Wang M, Wang CY, Zhao CY, Zhang XY, Zhou Q, Zhao WJ, Yang F, Zhang CL, Yang AJ, Dong JF, Li M. Extracellular vesicles, hyperadhesive von willebrand factor, and outcomes of gastric cancer: a clinical observational study. Med Oncol 2023; 40:140. [PMID: 37031314 DOI: 10.1007/s12032-023-01950-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/12/2023] [Indexed: 04/10/2023]
Abstract
Von Willebrand factor (VWF) is an adhesive ligand critical for maintaining hemostasis. However, it has also been increasingly recognized for its role in cancer development because it has been shown to mediate the adhesion of cancer cells to endothelial cells, promote the epithelial-mesenchymal transition, and enhance angiogenesis. We have previously shown that gastric cancer cells synthesize VWF, which mediates the interaction between the cancer and endothelial cells to promote cancer growth. Here, we report results from a clinical observational study that demonstrate the association of VWF in plasma and on the surface of extracellular vesicles (EVs) with the pathological characteristics of gastric cancer. We found that patients with gastric cancer had elevated and intrinsically hyperadhesive VWF in their peripheral blood samples. VWF was detected on the surface of EVs from cancer cells, platelets, and endothelial cells. Higher levels of these VWF-bound EVs were associated with cancer aggression and poor clinical outcomes for patients. These findings suggest that VWF+ EVs from different cell types serve collectively as a new class of biomarkers for the outcome assessment of gastric cancer patients.
Collapse
Affiliation(s)
- Wei Cai
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
- Gansu Provincial Hospital, Lanzhou, China
| | - Min Wang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
- School of Basic Medical Sciences, Institute of Integrated Traditional Chinese and Western Medicine, Lanzhou University, Lanzhou, China
| | - Chen-Yu Wang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Chan-Yuan Zhao
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Xiao-Yu Zhang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Quan Zhou
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Wen-Jie Zhao
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Feng Yang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Chen-Li Zhang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Ai-Jun Yang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China.
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, WA, USA.
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Min Li
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China.
| |
Collapse
|
95
|
Ogata T, Narita Y, Wainberg ZA, Van Cutsem E, Yamaguchi K, Piao Y, Zhao Y, Peterson PM, Wijayawardana SR, Abada P, Chatterjee A, Muro K. Exploratory Analysis of Patients With Gastric/Gastroesophageal Junction Adenocarcinoma With or Without Liver Metastasis From the Phase 3 RAINBOW Study. J Gastric Cancer 2023; 23:289-302. [PMID: 37129153 PMCID: PMC10154140 DOI: 10.5230/jgc.2023.23.e15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 12/19/2022] [Accepted: 12/25/2022] [Indexed: 05/03/2023] Open
Abstract
PURPOSE Liver metastasis (LM) is reported in approximately 40% of patients with advanced/metastatic gastric/gastroesophageal junction adenocarcinoma (metastatic esophagogastric adenocarcinoma; mGEA) and is associated with a worse prognosis. This post-hoc analysis from the RAINBOW trial reported the efficacy, safety, and biomarker outcomes of ramucirumab and paclitaxel combination treatment (RAM+PAC) in patients with (LM+) and without (LM-) LM at baseline. MATERIALS AND METHODS Patients (n=665) were randomly assigned on a 1:1 basis to receive either RAM+PAC (LM+: 150, LM-: 180) or placebo and paclitaxel (PL+PAC) (LM+: 138, LM-: 197). The overall survival (OS) and progression-free survival (PFS) were evaluated using stratified Kaplan-Meier and Cox regression models. The correlation of dichotomized biomarkers (VEGF-C, D; VEGFR-1,2) with efficacy in the LM+ versus LM- subgroups was analyzed using the Cox regression model with reported interaction P-values. RESULTS The presence of LM was associated with earlier progression than those without LM, particularly in patients receiving PL+PAC (hazard ratio [HR], 1.68). RAM+PAC treatment improved OS and PFS irrespective of LM status but showed greater improvement in LM+ than that in LM- (OS HR, 0.71 [LM+] vs. 0.88 [LM-]; PFS HR, 0.47 [LM+] vs. 0.76 [LM-]). Treatment-emergent adverse events were similar between patients with and without LM. No predictive relationship was observed between biomarker levels (VEGF-C, D; VEGFR-1,2) and efficacy outcome (OS, PFS) (all interaction P-values >0.05). CONCLUSIONS RAM provided a significant benefit, irrespective of LM status; however, its effect was numerically stronger in patients with LM. Therefore, RAM+PAC is a clinically meaningful therapeutic option for patients with mGEA and LM. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01170663.
Collapse
Affiliation(s)
| | | | - Zev A Wainberg
- University of California Los Angeles, Los Angeles, CA, United States
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg/Leuven & Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Kensei Yamaguchi
- Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | - Yumin Zhao
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | | | - Paolo Abada
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | - Kei Muro
- Aichi Cancer Center Hospital, Nagoya, Japan.
| |
Collapse
|
96
|
Guan X, Lu N, Zhang J. Computed Tomography-Based Deep Learning Nomogram Can Accurately Predict Lymph Node Metastasis in Gastric Cancer. Dig Dis Sci 2023; 68:1473-1481. [PMID: 35909203 PMCID: PMC10102043 DOI: 10.1007/s10620-022-07640-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/18/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND Computed tomography is the most commonly used imaging modality for preoperative assessment of lymph node status, but the reported accuracy is unsatisfactory. AIMS To evaluate and verify the predictive performance of computed tomography deep learning on the presurgical evaluation of lymph node metastasis in patients with gastric cancer. METHODS 347 patients were retrospectively selected (training cohort: 242, test cohort: 105). The enhanced computed tomography arterial phase images of gastric cancer were used for lesion segmentation, radiomics and deep learning feature extraction. Three methods were used for feature selection. Support vector machine (SVM) or random forest (RF) was used to build models. The classification performance of the models was evaluated using the area under the receiver operating characteristic curve (AUC). We also established a nomogram that included clinical predictors. RESULTS The model based on ResNet50-RF showed favorable classification performance and was verified in the test cohort (AUC = 0.9803). The nomogram based on deep learning feature scores and the lymph node status reported by computed tomography showed excellent discrimination. AUC of 0.9978 was achieved in the training cohort and verified in the test cohort (AUC = 0.9914). Decision analysis curve showed the value of nomogram in clinical application. CONCLUSION The computed tomography-based deep learning nomogram can accurately and effectively evaluate lymph node metastasis in patients with gastric cancer before surgery.
Collapse
Affiliation(s)
- Xiao Guan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No. 121, Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| | - Na Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No. 121, Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| | - Jianping Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No. 121, Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| |
Collapse
|
97
|
Li X, Chen Z, Zhang Y, Zhang H, Niu H, Zheng C, Jing X, Qiao H, Wang G, Yang W. Effect of multimodal chemotherapy on survival of gastric cancer with liver metastasis – a population based analysis. Front Oncol 2023; 13:1064790. [PMID: 37007120 PMCID: PMC10061116 DOI: 10.3389/fonc.2023.1064790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
ObjectivesLimited efforts have been made to evaluate the effect of multimodal chemotherapy on the survival of gastric cancer patients with liver metastases (LMGC). This study aimed to identify prognostic factors in LMGC patients and the superiority of multimodal chemotherapy with respect to overall survival (OS) in these patients.MethodsWe conducted a retrospective cohort study of 1298 patients with M1 stage disease between January 2012 and December 2020. The effects of clinicopathological variables and preoperative chemotherapy (PECT), postoperative chemotherapy (POCT), and palliative chemotherapy on survival in patients with liver metastases (LM group) and non-liver metastases (non-LM group) were compared.ResultsOf the 1298 patients analysed, 546 (42.06%) were in the LM group and 752 (57.94%) were in the non-LM group. The median (interquartile range) age was 60 (51–66) years. The 1-year, 3-year and 5-year overall survival (OS) rates in the LM group were 29.3%, 13.9%, and 9.2%, respectively, and those in the non-LM group were. 38.2%, 17.4%, and 10.0%, respectively (P < 0.05, > 0.05, and > 0.05, respectively.) The Cox proportional hazards model revealed that palliative chemotherapy was a significant independent prognostic factor in both the LM and non-LM groups. Age ≥55 years, N stage, and Lauren classification were also independent predictors of OS in the LM group (P < 0.05). Palliative chemotherapy and POCT were associated with improved OS compared with PECT in the LM group (26.3% vs. 36.4% vs. 25.0%, P < 0.001).ConclusionLMGC patients had a worse prognosis than non- LMGC. Number of metastatic sites more than 1, liver and other metastatic sites, no CT treatment and HER2-negative had a poor prognosis. LMGC patient may benefit more from palliative chemotherapy and POCT than from PECT. Further well-designed, prospective studies are needed to validate these findings.
Collapse
Affiliation(s)
- Xinghui Li
- Cancer Institute of the General Hospital, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zhiqiang Chen
- Department of Radiology, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Yue Zhang
- Cancer Institute of the General Hospital, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Hong Zhang
- Cancer Institute of the General Hospital, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Haiyan Niu
- Department of Pathology, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Cheng Zheng
- Cancer Institute of the General Hospital, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xiaoying Jing
- Cancer Institute of the General Hospital, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Hui Qiao
- Cancer Institute of the General Hospital, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Guanhua Wang
- Cancer Institute of the General Hospital, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
- *Correspondence: Guanhua Wang, ; Wenjun Yang,
| | - Wenjun Yang
- Cancer Institute of the General Hospital, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Radiology, The First Affiliated Hospital, Hainan Medical University, Haikou, China
- Department of Pathology, The First Affiliated Hospital, Hainan Medical University, Haikou, China
- *Correspondence: Guanhua Wang, ; Wenjun Yang,
| |
Collapse
|
98
|
Yagi S, Ida S, Namikawa K, Hayami M, Makuuchi R, Kumagai K, Ohashi M, Sano T, Nunobe S. Clinical outcomes of palliative treatment for gastric bleeding from incurable gastric cancer. Surg Today 2023; 53:360-368. [PMID: 35932300 DOI: 10.1007/s00595-022-02567-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/13/2022] [Indexed: 10/16/2022]
Abstract
PURPOSE Deciding palliative treatment for gastric bleeding from incurable gastric cancer (IGC) is worrying considering different patient situations and the lack of comprehensive assessment of palliative treatment. We evaluated the clinical outcomes and prognostic factors after palliative treatment for gastric bleeding from IGC. METHODS We enrolled 48 consecutive patients with gastric bleeding from IGC who underwent palliative surgery (PS) or palliative radiotherapy (PRT). RESULTS Of the 48 patients, 23 underwent PS and 25 received PRT. More patients who had an Eastern Cooperative Oncology Group-Performance Status (ECOG-PS) ≥ 2 or who received chemotherapy underwent PRT than underwent PS. Severe complications were observed in 2 (8.6%) patients after PS. After PRT, 22 patients achieved hemostasis (88%), but rebleeding was found in 10 (40%). Chemotherapy was introduced after palliative treatment for 21 (91.3%) patients in the PS group and 17 (68%) patients in the PRT group. The median survival time (MST) of patients with and without chemotherapy after PS was 12.5 and 3.1 months, respectively (p ≤ 0.001), while the MST of patients with and without chemotherapy after PRT was 6.5 and 1.6 months (p < 0.001). Multivariate analyses identified ECOG-PS, tumor size, and chemotherapy after palliative treatment as independent risk factors. CONCLUSIONS Palliative treatment strategies for gastric bleeding should be determined with consideration of the general condition, previous chemotherapy, and chemotherapy after palliative treatment.
Collapse
Affiliation(s)
- Shusuke Yagi
- Department of Gastroenterology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan.,Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Satoshi Ida
- Department of Gastroenterology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ken Namikawa
- Department of Gastroenterology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masaru Hayami
- Department of Gastroenterology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Rie Makuuchi
- Department of Gastroenterology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Koshi Kumagai
- Department of Gastroenterology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Manabu Ohashi
- Department of Gastroenterology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Sano
- Department of Gastroenterology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Souya Nunobe
- Department of Gastroenterology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
99
|
Chen VW, Portuondo JI, Cooper Z, Massarweh NN. Variation in hospital utilization of palliative interventions for patients with advanced gastrointestinal cancer near end of life. J Surg Oncol 2023; 127:741-751. [PMID: 36514285 DOI: 10.1002/jso.27177] [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: 09/08/2022] [Revised: 10/31/2022] [Accepted: 12/04/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Patients with advanced gastrointestinal (GI) cancer often undergo noncurative interventions with palliative intent to relieve high symptom burden near end of life. Hospital-level variation in intervention utilization remains unclear. METHODS National cohort study of 142 304 patients with stage III or IV GI cancer within the National Cancer Database (2004-2014) who died within 1-year of diagnosis. Hospitals were stratified by palliative intervention utilization (surgery, chemotherapy, radiation, pain management). Multivariable, multinomial regression evaluated the association between patient/hospital factors and palliative intervention utilization. RESULTS Across 1322 hospitals, median hospital palliative intervention utilization was 12.0% [interquartile range: 0.0%-26.1%]. Utilization increased over time in all but lowest utilizing hospitals. Relative to lowest utilizing hospitals, factors associated with a lower likelihood of care at highest utilizing hospitals included: race (White [ref]; Black-Relative Risk Ratio [RRR] 0.81, 95% confidence interval [0.77-0.85]) and lower income (RRR 0.81 [0.78-0.84]). Factors associated with a higher likelihood included: lower education level (RRR 1.62 [1.55-1.69]) and hospital type (community program [ref]; comprehensive community-RRR 1.33 [1.26-1.41]; academic-RRR 1.88 [1.77-1.99]; integrated network-RRR 1.79 [1.66-1.93]). CONCLUSION Hospital variation in palliative intervention use is substantial and potentially associated with sociodemographic and hospital characteristics. Future work can examine how differences in hospital care processes translate to quantity/quality of life for cancer patients.
Collapse
Affiliation(s)
- Vivi W Chen
- Center for Innovations in Quality, Effectiveness, and Safety, Michael E DeBakey VA Medical Center, Houston, Texas, USA.,Michael E DeBakey Department of Surgery at Baylor College of Medicine, Houston, Texas, USA
| | - Jorge I Portuondo
- Michael E DeBakey Department of Surgery at Baylor College of Medicine, Houston, Texas, USA
| | - Zara Cooper
- Center for Surgery and Public Health, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Nader N Massarweh
- Surgical and Perioperative Care, Atlanta VA Health Care System, Decatur, Georgia, USA.,Department of Surgery, Division of Surgical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
100
|
Matsui R, Rifu K, Watanabe J, Inaki N, Fukunaga T. Impact of malnutrition as defined by the GLIM criteria on treatment outcomes in patients with cancer: A systematic review and meta-analysis. Clin Nutr 2023; 42:615-624. [PMID: 36931162 DOI: 10.1016/j.clnu.2023.02.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 02/01/2023] [Accepted: 02/20/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND & AIMS Malnutrition has been reported to lead to poor postoperative outcomes. The Global Leadership Initiative on Malnutrition (GLIM) criteria were published in 2019 as a global consensus on the criteria for diagnosing malnutrition. However, the relationship between GLIM-defined malnutrition and treatment outcomes in patients with cancer has not been fully investigated. Therefore, this study aimed to clarify the impact of GLIM-defined malnutrition on the treatment outcomes of patients with cancer. METHODS We searched MEDLINE, the Cochrane Central Register of Controlled Trials, EMBASE, the World Health Organization International Clinical Trials Platform Search Portal, and ClinicalTrials.gov and identified observational studies published from inception to January 17, 2022. We conducted a systematic review and random-effects meta-analysis studies that included patients with cancer aged >18 years who received any kind of treatment and whose nutritional status was assessed using GLIM criteria. We independently assessed the risk of bias and quality of evidence using Quality In Prognosis Studies and Grading of Recommendations, Assessment, Development, and Evaluation approach. The primary outcomes were overall survival (OS) and postoperative complications. Hazard ratios and 95% confidence intervals (CIs) for OS and relative risk ratios and 95% CIs for postoperative complications were pooled. The protocol was published by PROSPERO (CRD42022304004). RESULTS Of 67 studies after screening, ten studies (n = 11,700) reported the impact of GLIM-defined malnutrition on postoperative outcomes. Compared with no malnutrition, GLIM-defined malnutrition may worsen OS (hazard ratio, 1.56; 95% CI, 1.38-1.75; I2 = 37%) and increase postoperative complications (relative risk ratio, 1.82; 95% CI, 1.28-2.60; I2 = 87%). The risk of bias in each study was either moderate or high. The certainty of the evidence was low because of publication bias and a moderate or high risk of bias. CONCLUSIONS GLIM-defined malnutrition may worsen OS and increase the risk of postoperative complications in patients with cancer undergoing treatment. Further studies are needed to confirm these findings and mitigate this risk.
Collapse
Affiliation(s)
- Ryota Matsui
- Department of Surgery, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu City, Chiba 279-0021, Japan; Department of Upper Gastrointestinal Surgery, Juntendo University Hospital, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan; Department of Gastroenterological Surgery, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
| | - Kazuma Rifu
- Department of Surgery, Division of Gastroenterological, General and Transplant Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke City, Tochigi 329-0498, Japan.
| | - Jun Watanabe
- Department of Surgery, Division of Gastroenterological, General and Transplant Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke City, Tochigi 329-0498, Japan; Center for Community Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke City, Tochigi 329-0498, Japan.
| | - Noriyuki Inaki
- Department of Upper Gastrointestinal Surgery, Juntendo University Hospital, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan; Department of Gastrointestinal Surgery/Breast Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa City, Ishikawa 920-8641, Japan.
| | - Tetsu Fukunaga
- Department of Upper Gastrointestinal Surgery, Juntendo University Hospital, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan.
| |
Collapse
|