1
|
Zheng HL, Zhang LK, Zheng HH, Lv CB, Xu BB, Lin GT, Chen QY, Lin JX, Zheng CH, Huang CM, Xie JW. Timing of postoperative chemotherapy and prognosis in neoadjuvant-treated gastric cancer patients: a multicenter real-world cohort study. Ann Med 2025; 57:2500690. [PMID: 40329795 PMCID: PMC12064125 DOI: 10.1080/07853890.2025.2500690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND The optimal time to chemotherapy (TTC) in locally advanced gastric cancer (LAGC) patients treated with neoadjuvant chemotherapy (NLAGC) remains unclear. METHODS Consecutive 524 patients with NLAGC between Jan. 2010 and Dec. 2022 were identified. Patients were categorized into three groups: TTC < 6w, 6w ≤ TTC ≤ 8w, and TTC > 8w. Survival analysis was conducted using the Cox proportional hazards model to assess the impact of TTC on gastric cancer-specific mortality (GCSM) and all-cause mortality (ACM). Cumulative competing risk curves were employed to evaluate the incidence of competing events. RESULTS Overall, 451 patients were included.Cumulative competing risk curves showed that the 3-year ACM and GCSM were significantly lower in the 6w ≤ TTC ≤ 8w group (ACM: 19.7% vs. 37.2% vs. 39.7%, GCSM: 19.7% vs. 35.2% vs. 38.8%) compared to the TTC < 6w and TTC > 8w groups. Compared to patients with 6w ≤ TTC ≤ 8w, those with TTC < 6w or >8w had an increased risk of GCSM (HR: 2.792 and HR: 2.343, respectively) and ACM (HR: 3.102 and HR: 2.719, respectively) after adjusting for confounders. Furthermore, 6w ≤ TTC ≤ 8w had later peak recurrence compared to TTC < 6w and TTC > 8w (Peak months: 9.7 vs. 4.3 vs. 3.1). CONCLUSION A postoperative chemotherapy timing of 6-8 weeks was associated with better survival and delayed recurrence in NLAGC patients. These findings suggest that the 6-8 week time-window should be a key timeframe for personalized postoperative adjuvant chemotherapy decisions.
Collapse
Affiliation(s)
- Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Ling-Kang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Hong-Hong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chen-Bin Lv
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Bin-Bin Xu
- Department of Digestive Endoscopy, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
2
|
Shen LL, Zheng HL, Zheng ZW, Xu BB, Xue Z, Jia-Lin, Chen QY, Xie JW, Li P, Huang CM, Lin JX, Zheng CH. Paradoxical effects of adiposity and inflammation on immunotherapy efficacy in gastric cancer: novel insights from real-world data. Gastric Cancer 2025:10.1007/s10120-025-01622-w. [PMID: 40350511 DOI: 10.1007/s10120-025-01622-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Emerging studies suggest obesity may improve PD-1/PD-L1 inhibitor efficacy, correlating with prolonged survival, known as the 'obesity paradox'. However, the impact of this paradox and obesity-related chronic inflammation on immunotherapy for advanced gastric cancer (AGC) has not received sufficient research. METHODS Between January 2018 and December 2021, patients receiving neoadjuvant therapy were categorized into two groups: combined immunotherapy (ICIs, n = 173) and neoadjuvant chemotherapy (NAC, n = 126). Visceral (VATI) and subcutaneous adipose tissue index (SATI) were obtained from pre-treatment CT images. The systemic immune-inflammation index (SII) was calculated as platelet count multiplied by the neutrophil-to-lymphocyte ratio. RESULTS The median age of patients was 64 years (IQR 56-69), with 219 (73.2%) males and 80 (26.8%) females. In the ICIs group, the VATI-High group showed significantly higher 3-year overall survival (OS) (p = 0.010) and disease-free survival (DFS) (p = 0.029). Similar results were observed in the SATI analysis (p < 0.05). Conversely, OS (p = 0.040) and DFS (p = 0.039) were significantly lower in the SII-High group. Both VATI and SATI were independent protective factors for OS and DFS, but the effect disappeared after adjustment for SII. SII was associated with poorer OS and DFS, even after adjustment for VATI and SATI. No significant differences were observed in the analysis of the NAC group. CONCLUSIONS Elevated adiposity indices (VATI/SATI) and low SII correlate with survival benefit in ICI-treated AGC patients, and importantly, this paradoxical survival benefit is dependent on SII status. In contrast, no such benefit is observed in chemotherapy-alone cohorts.
Collapse
Affiliation(s)
- Li-Li Shen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Zhi-Wei Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Bin-Bin Xu
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Department of Digestive Endoscopy, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Zhen Xue
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, 59, Fuzhou, 350001, Fujian, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
3
|
Zhong Q, Weng CM, Jiang MC, Sun YQ, Li BL, Zhao W, Zhang HX, Zhang ZQ, Ma YB, Wu SC, Ye W, Wu J, Du H, Zheng CH, Li P, Chen QY, Huang CM, Xie JW. Patterns of Survival and Recurrence in Poor Responders to Neoadjuvant Therapy for Gastric Cancer: A Real-World Multicenter Study. Ann Surg Oncol 2025:10.1245/s10434-025-17396-5. [PMID: 40329137 DOI: 10.1245/s10434-025-17396-5] [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: 11/01/2024] [Accepted: 04/13/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Neoadjuvant therapy (NAT) is increasingly used in locally advanced gastric cancer (LAGC), but a significant proportion of patients respond poorly, causing adverse outcomes. Few studies have specifically examined the prognosis of this subgroup. This study aimed to analyze survival and recurrence in poor responders to guide follow-up and treatment strategies. METHODS This multicenter retrospective study included patients with LAGC who received NAT. Tumor regression was graded following the Becker system, defining TRG 2-3 as poor response. Outcomes were assessed for overall survival (OS), recurrence-free survival (RFS), and recurrence patterns. RESULTS 648 patients were included: 341 with TRG 2 and 307 with TRG 3. In the entire cohort, the 3-year OS and RFS were 54.6% and 55.2%, respectively. Recurrence occurred in 299 patients, with the following recurrence patterns: distant metastasis (26.1%, n = 78), peritoneal metastasis (21.1%, n = 63), locoregional recurrence (18.7%, n = 56), and multiple-site recurrence (18.4%, n = 55). Liver metastasis was significantly higher in the TRG 3 group than in the TRG 2 group (14.1% versus 5.3%, P = 0.010). ypN+ was the most significant independent risk factor for recurrence (OR = 2.73, 95% CI 1.83-4.08, P < 0.001); an increasing number of positive lymph nodes led to higher 3-year cumulative mortality in patients. Despite poor response to NAT, completing over four adjuvant chemotherapy cycles was associated with improved survival outcomes. CONCLUSION Poor NAT responders in LAGC have high recurrence rates, particularly in the first year post-surgery, with ypN+ status being the strongest predictor of recurrence. Completing over four cycles of AC was associated with survival improvement in this group.
Collapse
Affiliation(s)
- Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Cai-Ming Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Mei-Chen Jiang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yu-Qin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Bao-Long Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of General Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Wei Zhao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hao-Xiang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Yu-Bin Ma
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Xining, China
| | - Shi-Chao Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Gastrointestinal Surgery Unit 2, Putian First Hospital of Fujian Medical University, Putian, China
| | - Wen Ye
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - He Du
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Xining, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
4
|
Li H, Huang J, Liu K, Liu J, Liu Q, Zhou Z, Zong Z, Mao S. ChatGPT-4o outperforms gemini advanced in assisting multidisciplinary decision-making for advanced gastric cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110096. [PMID: 40294561 DOI: 10.1016/j.ejso.2025.110096] [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: 10/09/2024] [Revised: 02/09/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND & AIMS The treatment of advanced gastric cancer (GC) requires precise and comprehensive clinical decision-making. Artificial intelligence (AI) chatbots offer potential tools to enhance multidisciplinary team (MDT) discussions. This study aims to compare the performances of ChatGPT-4o and Gemini Advanced in generating treatment recommendations for advanced GC. METHODS The study involved three steps: (1) evaluating responses to ten critical clinical questions, (2) analyzing clinical cases from MDT meetings at our institution, and (3) reviewing rare GC cases from PubMed. It included 95 advanced GC patients discussed between November 2022 and July 2024, and 14 rare cases from PubMed. Prompts designed from advanced GC cases were submitted to ChatGPT-4o and Gemini Advanced using a standardized format. Outputs were evaluated for accuracy and completeness using a structured 4-point Likert scale. Interrater reliability was calculated to ensure consistency among evaluators. RESULTS For the ten clinical questions, ChatGPT-4o achieved better performances compared to Gemini Advanced. In MDT cases, ChatGPT-4o provided more valuable recommendations in surgical suggestion, chemotherapy recommendation, and chemotherapy regimens. Subgroup analysis confirmed these findings in both routine and complex cases with high interrater reliability. ChatGPT-4o also outperformed Gemini Advanced in the analysis of rare GC cases from PubMed, showing superior accuracy with high interrater reliability. CONCLUSIONS While our findings suggest that AI chatbots can generate clinically relevant and guideline-based treatment recommendations, their use in MDT decision-making should be viewed as supportive rather than autonomous. We emphasize that while AI chatbots have potential as decision-support tools, but they should be integrated only under expert supervision in a real-world clinical context.
Collapse
Affiliation(s)
- Huizi Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Jiaobao Huang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kuntang Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jibiao Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Queling Liu
- Department of Medical Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhiyong Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhen Zong
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shengxun Mao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
5
|
Wu D, Bian L, Wang Z, Ni J, Chen Y, Zhang L, Chen X. Influence of visceral adipose tissue on the accuracy of tumor T-staging of gastric cancer in preoperative CT. Jpn J Radiol 2025; 43:656-665. [PMID: 39607533 DOI: 10.1007/s11604-024-01711-3] [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: 09/25/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
OBJECTIVES To evaluate the impact of the visceral adipose tissue (VAT) area and density on the accuracy of tumor T-staging of gastric cancer in preoperative computed tomography (CT). METHODS This study included 136 patients with gastric cancer in our research center from January 2021 to June 2022. The patients were divided into two groups based on their postoperative pathological results: accurate-staging (matched T-staging evaluated by preoperative CT and postoperative pathology) and inaccurate-staging (unmatched T-staging evaluated by preoperative CT and postoperative pathology) groups. Preoperative CT was performed to assess the VAT area and density, and logistic regression was employed to evaluate the effect of VAT on the accuracy of preoperative-CT-evaluated T-staging of patients with gastric cancer. RESULTS The accurate-staging group had a significantly higher VAT area (134.64 ± 70.55 cm2 vs 95.44 ± 66.18 cm2, P = 0.003) and significantly lower VAT density (-95.05 ± 12.28 Hounsfield Units [HU] vs - 89.68 ± 13.26 HU, P = 0.027) than the inaccurate-staging group. A low VAT area (P = 0.002) and tumor located in the upper stomach (P = 0.019) were significantly associated with and were independent risk factors for the error of CT-evaluated T-staging. Compared to a VAT area ≥ 81.04 cm2, which was used as a reference, the odds ratio (OR) of a VAT area < 81.04 cm2 for the probability of T-staging mis-assessment was 4.455 (95% confidence interval [CI]: 1.728-11.485). CONCLUSIONS A low VAT area in patients with gastric cancer had adverse effects on preoperative CT-evaluated T-staging.
Collapse
Affiliation(s)
- Danping Wu
- Department of Nuclear Medicine, Jiangyuan Hospital Affiliated to Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Linjie Bian
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Radiology, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China
| | - Zhongjuan Wang
- Department of Radiology, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China.
| | - Jianming Ni
- Department of Radiology, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China
| | - Yigang Chen
- Department of General Surgery, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China
| | - Lei Zhang
- Department of Radiology, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China
| | - Xulei Chen
- Department of Pathology, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China
| |
Collapse
|
6
|
He QC, Huang ZN, Lv CB, Wu YH, Qiu WW, Ma YB, Wu J, Zheng CY, Lin GS, Li P, Wang JB, Lin JX, Lin M, Tu RH, Zheng CH, Huang CM, Cao LL, Xie JW. Effect of Helicobacter pylori infection on survival outcomes of patients undergoing radical gastrectomy after neoadjuvant chemotherapy: a multicenter study in China. BMC Cancer 2025; 25:460. [PMID: 40082850 PMCID: PMC11907980 DOI: 10.1186/s12885-025-13840-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NAC) has been confirmed to improve the prognosis of patients with advanced gastric cancer (AGC). However, no study has investigated whether Helicobacter pylori (HP) infection affects the postoperative survival of patients who receive NAC. METHODS This retrospective cohort study included 307 patients with AGC who underwent laparoscopic radical gastrectomy after NAC at three hospitals in China between January 1, 2016, and April 31, 2020. Cox regression was used to assess prognostic factors for survival. Kaplan-Meier was used for survival analysis. RESULTS The HP + and the HP- group included 141 and 166 cases. The 3-year overall survival (OS) and disease-free survival (DFS) of the HP + group were significantly better than the HP- group (3-year OS: 75.9% vs. 60.2%, 3-year DFS: 70.2% vs. 52.3%; All P < 0.001). For the HP + group, ypTNM Stage III (HR, 4.00; 95% CI, 1.11-14.39; P = 0.034), NAC ≥ 4 cycles (HR, 0.43; 95% CI, 0.20-0.90; P = 0.026), and adjuvant chemotherapy (AC) ≥ 4 cycles (HR, 0.20; 95% CI, 0.09-0.48; P < 0.001) are independent prognostic factors for OS. In the cohort of HP + patients who received ≥ 4 cycles of NAC, the prognosis of patients who received ≥ 4 cycles of AC after surgery was better than that of patients who received < 4 cycles of AC (3-year OS: 92.5% vs 71.4%; P = 0.042). CONCLUSIONS Following NAC, HP + patients with AGC exhibit better prognosis than that of HP- counterparts. For potentially resectable HP + AGC patients, radical surgery following ≥ 4 cycles of NAC with ≥ 4 cycles of sequential AC might be recommended to improve survival.
Collapse
Affiliation(s)
- Qi-Chen He
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Chen-Bin Lv
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Yong-He Wu
- Department of Pathology, Zhangzhou Affiliated Hospital of Fujian Medical University, ZhangZhou, China
| | - Wen-Wu Qiu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Chang-Yue Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Putian University, Putian, China
| | - Guo-Sheng Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
7
|
Hu M, Zheng H, Zheng H, Xu B, Wei L, Xue Z, Shen L, Yu J, Xie R, Lin J, Zhang L, Zheng Z, Xie J, Zheng C, Huang C, Wang J, Li P. Clinical Value of Nomograms Integrating Circulating Lipid and Inflammation Risk Score in Predicting Long-Term Outcomes After Radical Gastrectomy in Gastric Cancer: A Multicenter Real-World Study. Ann Surg Oncol 2025; 32:2172-2184. [PMID: 39681718 DOI: 10.1245/s10434-024-16687-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND The clinical value of incorporating lipid and inflammatory factors to predict long-term survival in patients with gastric cancer (GC) is unreported. This study aimed to investigate the clinical value of nomograms integrating the Circulating Lipid and Inflammation Risk Score (CLIRS) for predicting the long-term outcome of patients with GC. METHODS A retrospective analysis included patients with GC who underwent radical resection at four tertiary medical centers. Patients were divided into training and validation cohorts, with least absolute shrinkage and selection operator regression selecting optimal lipid and inflammatory indicators related to GC prognosis. The CLIRS was developed from six indicators: lymphocyte, triglycerides, total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and apolipoprotein B. RESULTS Overall, 2534 patients were studied, including 1910 in the training cohort and 624 in the validation cohort. The CLIRS was an independent risk factor for overall survival (OS; hazard ratio [HR] 1.529, 95% confidence interval [CI] 1.271-1.839; p < 0.001) and disease-free survival (DFS; HR 1.511, 95% CI 1.267-1.801; p < 0.001) in GC patients. The OS nomogram (area under the receiver operating characteristic curve 0.823 vs. 0.785; p < 0.05) and DFS nomogram (AUC 0.804 vs. 0.770; p < 0.05) based on the CLIRS outperformed pTNM stage. High-risk patients had earlier and more sustained recurrence, with higher rates of local, peritoneal, and distant recurrences (p < 0.05). CONCLUSIONS The CLIRS, combining circulating lipid and inflammatory factors, is an independent prognostic factor for patients with GC. Nomograms incorporating the CLIRS are superior to pTNM stage in predicting postoperative survival and recurrence in patients with GC.
Collapse
Affiliation(s)
- Minggao Hu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Anqing 116 Hospital, China RongTong Medical, Healthcare Group Co. Ltd, Anqing, China
| | - Hualong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Honghong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Binbin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Linghua Wei
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Zhen Xue
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Lili Shen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Junhua Yu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Rongzhen Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jia Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Lingkang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Zhiwei Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Jianwei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Chaohui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Changming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China.
| | - Jiabin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China.
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China.
| |
Collapse
|
8
|
Yu J, Zheng H, Xue Z, Sun Y, Xu B, Shen L, Ma Y, Zhang L, Zheng H, Wang Y, Zheng C, Wu S, Huang C, Lin J, Zheng C. Effect of Adjuvant Chemotherapy Cycles on Patients with Node-Negative Gastric Cancer Following Neoadjuvant Chemotherapy: Multicenter Cohort Study. Ann Surg Oncol 2025; 32:2150-2160. [PMID: 39674863 DOI: 10.1245/s10434-024-16585-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: 07/10/2024] [Accepted: 11/12/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Research investigating the potential impact of postoperative adjuvant chemotherapy cycles on patients with lymph node-negative gastric cancer following neoadjuvant chemotherapy is currently sparse. This study aims to explore the effect of adjuvant chemotherapy cycles on the prognosis of this specific patient group. PATIENTS AND METHODS We analyzed clinicopathological data from patients at four institutions between 2010 and 2020. Independent risk factors associated with 3-year overall survival (OS) were identified using a Cox proportional hazards regression model. RESULTS We enrolled a total of 219 patients in this study. Patients with lymph node-negative gastric cancer who received neoadjuvant chemotherapy and underwent at least five cycles of adjuvant chemotherapy (AC ≥ 5) after surgery had a significantly higher 3-year overall survival rate of 86.8% compared with those who received fewer than five cycles (AC < 5) with a survival rate of 68.1% (P = 0.016). Multivariate analysis identified several factors, including AC ≥ 5 (HR 0.367, 95% CI 0.166-0.815, P = 0.014), ypT stage ≥ 2 (HR 2.779, 95% CI 1.199-6.438, P = 0.017), and poorly differentiated tumors (HR 2.501, 95% CI 1.385-4.517, P = 0.002), as independently associated with 3-year OS in this patient group. Stratified analysis further revealed that AC ≥ 5 significantly enhanced long-term outcomes in patients with ypT stage ≥2 (3-year OS, 82.5% vs. 62.6%, P = 0.025) and in those with poorly differentiated tumors (3-year OS, 82.6% vs. 53.3%, P = 0.021). CONCLUSION Patients who have lymph node-negative gastric cancer following neoadjuvant chemotherapy, with either ypT stage ≥ 2 or poorly differentiated gastric cancers, may experience benefits from undergoing at least five cycles of adjuvant chemotherapy.
Collapse
Affiliation(s)
- Junhua Yu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Gastrointestinal Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang Province, China
| | - Hualong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Zhen Xue
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yuqin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Binbin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Lili Shen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yubin Ma
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Lingkang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Honghong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yonghong Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Gastrointestinal Surgery, The People's Hospital of Leshan, Leshan, Sichuan Province, China
| | - Changyue Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Putian University, Putian,, Fujian Province, China
| | - Shichao Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
- Gastrointestinal Surgery Unit 2, Teaching Hospital of Putian First Hospital of Fujian Medical University, Putian, Fujian Province, China
| | - Changming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jianxian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Chaohui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
9
|
Wong E, Kamarajah SK, Dahdaleh F, Naffouje S, Kunene V, Fackrell D, Griffiths EA. Adjuvant chemotherapy for node-negative gastric adenocarcinoma after neoadjuvant chemotherapy and gastrectomy: A propensity score matched analysis study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109506. [PMID: 39756161 DOI: 10.1016/j.ejso.2024.109506] [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: 05/23/2024] [Revised: 11/10/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025]
Abstract
INTRODUCTION The long term survival of patients undergoing curative resection for gastric cancer remains poor owing to high recurrence rates. The use of adjuvant chemotherapy in node positive gastric cancer to prolong survival and prevent recurrence is widely accepted. However, the role for adjuvant chemotherapy in node negative gastric cancer is less clear, particularly in the era of neoadjuvant chemotherapy. OBJECTIVE To determine the association of adjuvant chemotherapy with survival in patients undergoing pathologically node negative gastric cancer resection, following neoadjuvant chemotherapy. METHODS We examined a national cancer database containing patients who had undergone neoadjuvant chemotherapy and pathologically node negative curative gastrectomy. We divided these patients into those who had undergone adjuvant chemotherapy versus those who had not. Using a propensity score matched analysis, we analyzed the survival of these patients between the 2 groups. RESULTS 5309 patients who had undergone curative gastrectomy were identified from the database and 806 of these patients were given adjuvant chemotherapy. Following propensity score matched analysis, patients who had been given adjuvant chemotherapy had an increased median survival of 150 vs 125 months (5-year 68 % vs 62 %, p < 0.001). CONCLUSION There is a small, but statistically significant survival benefit for adjuvant chemotherapy in patients with node negative gastric cancer who had undergone neoadjuvant chemotherapy. Further studies are required to examine the role of adjuvant chemotherapy in this subset of patients.
Collapse
Affiliation(s)
- Enoch Wong
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - Sivesh K Kamarajah
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham, UK; Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Fadi Dahdaleh
- Department of Surgical Oncology, Edward-Elmshurst Health, Naperville, IL, USA
| | - Samer Naffouje
- Department of General Surgery, Cleveland Clinic Main Campus, Cleveland, OH, USA
| | - Victoria Kunene
- Department of Oncology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - David Fackrell
- Department of Oncology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - Ewen A Griffiths
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham, UK; Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
10
|
Miciak M, Jurkiewicz K, Dzierżek P, Rudno-Rudzińska J, Kielan W. Prognostic Significance of Lymph Node Ratio (LNR) in Gastric Cancer in Predicting Postoperative Complications and Survival: A Single-Center Study. Cancers (Basel) 2025; 17:743. [PMID: 40075592 PMCID: PMC11899347 DOI: 10.3390/cancers17050743] [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: 02/04/2025] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES The Lymph Node Ratio (LNR) index is the proportion of lymph nodes with present metastases to lymph nodes removed and examined. This is an additionally established parameter for predicting the prognosis of gastric cancer patients. The most popular cancer classification, TNM, describes only the number of affected lymph nodes. It can result in a negative overestimation of the prognosis of patients with gastric cancer if the number of nodes examined is relatively limited. METHODS In this study, we retrospectively analyzed 194 patients diagnosed with gastric cancer operated on between 2017 and 2021 at the Clinical Department of Oncological Surgery, University Centre of General and Oncological Surgery of the University Clinical Hospital in Wroclaw. In total, 133 patients underwent gastrectomy with D2 lymphadenectomy and 61 remaining patients had the resection procedure abandoned due to an unresectable lesion. The LNR index was calculated based on histopathological examination, and postoperative complications were assessed using the Clavien-Dindo (C-D) scale. Statistical analysis was performed regarding the dependence of LNR on the following patient characteristics: sex, age, TNM features, tumor stage, tumor location, performed procedure, chemotherapy application, C-D complication rate, and survival rate. RESULTS The value of the LNR index significantly depends on TNM features (p < 0.05), clinical tumor stage (p < 0.05), and patient survival (p < 0.05), while no statistically significant relationship with C-D complication rate was demonstrated. CONCLUSIONS The LNR index is a relevant parameter in predicting prognosis and survival time in gastric cancer patients, but future studies on larger and differentiated groups of patients could further confirm its usefulness in the development of guidelines.
Collapse
Affiliation(s)
- Michał Miciak
- Clinical Department of Oncological Surgery, University Centre of General and Oncological Surgery, Faculty of Medicine, Wroclaw Medical University, 50-556 Wrocław, Poland; (K.J.); (J.R.-R.); (W.K.)
| | | | - Przemysław Dzierżek
- Clinical Department of Oncological Surgery, University Centre of General and Oncological Surgery, Faculty of Medicine, Wroclaw Medical University, 50-556 Wrocław, Poland; (K.J.); (J.R.-R.); (W.K.)
| | | | | |
Collapse
|
11
|
Mavani PT, Koo A, Kooby DA, Shah MM. ASO Author Reflections: Enhancing Return to Intended Oncologic Therapy in Gastric Cancer-The Role of Minimally Invasive Gastrectomy. Ann Surg Oncol 2025; 32:1257-1258. [PMID: 39556175 PMCID: PMC11710969 DOI: 10.1245/s10434-024-16517-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024]
Affiliation(s)
- Parit T Mavani
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Andee Koo
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - David A Kooby
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Mihir M Shah
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
12
|
Badheeb AM, Alyami IA, Alyami AY, Alyami M, Al Walani M, Alkarak S, Aman AA, Albaiji FM, Al Masad AG, Alyami AS, Seada IA, Abu Bakar A. Treatment Modalities and Survival Outcomes in Gastric Cancer: Insights From Najran, Saudi Arabia. Cureus 2025; 17:e79649. [PMID: 40151724 PMCID: PMC11949575 DOI: 10.7759/cureus.79649] [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] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is the fifth most prevalent cancer worldwide and the fourth leading cause of cancer-related mortality. The survival of GC patients is significantly affected by the timing of diagnosis, as late-stage detection drastically reduces survival rates. This study investigates treatment modalities, survival outcomes, and mortality factors of GC patients in Najran, Saudi Arabia. MATERIALS AND METHODS A retrospective analysis was conducted involving 121 patients diagnosed with GC treated at King Khaled Hospital in Najran from January 1, 2014, to December 31, 2022. Clinical and pathological parameters, treatment interventions, and survival outcomes were extracted and analyzed from medical records. The Kaplan-Meier method created survival curves and assessed survival probabilities over time. Univariate and multivariate Cox regression analyses identified independent factors associated with mortality risk, calculating hazard ratios (HR) and 95% confidence intervals (CI) to evaluate associations. RESULTS The median age of the patients was 64 years, with 70 (57.9%) being over 60 years old. The cohort included 86 males (71.1%) and 35 females (28.9%), resulting in a male-to-female ratio of 2.5:1. Notably, 24 patients (19.8%) were underweight, 49 patients (40.5%) had hypertension, and 33 patients (27.3%) had diabetes. Helicobacter pylori infection was present in 18 patients (14.9%). Human epidermal growth factor receptor-2 (HER2) status was negative in 38 patients (31.4%). Elevated levels of carcinoembryonic antigen (CEA ≥ 5 ng/ml) and CA19-9 (≥ 37 U/mL) were noted in 51 patients (42.1%) and 50 patients (41.3%), respectively. Surgical intervention was performed on 72 patients (59.5%), while 49 patients (40.5%) were deemed non-resectable. Among the surgical cohort, neoadjuvant therapy was administered to 36 patients (30%), while others underwent initial or staged surgeries. Pathological findings predominantly showed 95 cases (78.5%) of intestinal-type adenocarcinomas compared to 26 cases (21.5%) of diffuse type. The most common TNM (tumor, node, metastasis) stage was IV (50 patients, 41.3%), followed by stage III (25 patients, 20.7%). During a follow-up of 41.3 ± 38.8 months, 96 patients (79.3%) were alive, while 25 patients (20.7%) had died. The median survival time was 28 months (95% CI: 20-51 months). The one-year, three-year, and five-year survival rates were 68.2% (82 patients), 42.8% (52 patients), and 37.4% (45 patients), respectively. Increased mortality was significantly associated with female gender (HR: 3.12; 95% CI: 1.56-6.24; p = 0.0013), stage IV disease (HR: 1.92; 95% CI: 1.01-3.66; p = 0.0481), and elevated CEA levels (HR: 1.95; 95% CI: 1.08-3.53; p = 0.0277). In contrast, neoadjuvant chemotherapy was linked to reduced mortality (HR: 0.55; 95% CI: 0.31-0.97; p = 0.0374). CONCLUSIONS The findings of this study indicate a five-year survival rate of 37.4% (95% CI: 29.3-47.8%) among GC patients in Najran. Notably, female gender, advanced disease stage, and elevated CEA levels emerged as significant predictors of increased mortality. Conversely, the administration of neoadjuvant chemotherapy was associated with a reduced mortality risk. These results emphasize the critical need for personalized treatment strategies and robust risk assessments to improve patient outcomes, particularly high-risk ones.
Collapse
Affiliation(s)
- Ahmed M Badheeb
- Oncology, King Khalid Hospital, Najran, SAU
- Medicine, Hadhramout University, Mukalla, YEM
| | | | | | - Mohammed Alyami
- Colorectal and Oncology Surgery, King Khalid Hospital, Najran, SAU
| | - Mugahed Al Walani
- Gastroenterology and Advanced Endoscopy, King Khalid Hospital, Najran, SAU
| | | | - Abdelaziz A Aman
- Internal Medicine/Endocrine and Diabetes, King Khalid Hospital, Najran, SAU
| | | | - Ali G Al Masad
- Gastroenterology and Hepatology, King Khalid Hospital, Najran, SAU
| | | | - Islam A Seada
- Cardiothoracic Surgery, King Khalid Hospital, Najran, SAU
| | | |
Collapse
|
13
|
Gong Z, Zhou L, He Y, Zhou J, Deng Y, Huang Z, Wang W, Yang Q, Pan J, Li Y, Yuan X, Ma M. Efficacy analysis of prophylactic hyperthermic intraperitoneal chemotherapy in patients with locally advanced gastric cancer: a retrospective study. Front Oncol 2025; 14:1503045. [PMID: 39850825 PMCID: PMC11754059 DOI: 10.3389/fonc.2024.1503045] [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: 09/28/2024] [Accepted: 12/19/2024] [Indexed: 01/25/2025] Open
Abstract
Purpose This study aims to evaluate the effectiveness and safety of prophylactic hyperthermic intraperitoneal chemotherapy (P-HIPEC) in patients with locally advanced gastric cancer (AGC) after laparoscopic radical gastrectomy. Additionally, it explores how the frequency and timing of P-HIPEC influence treatment outcomes. Methods A retrospective analysis was conducted on 227 patients with locally AGC who underwent laparoscopic surgery at Maoming People's Hospital from January 2016 to December 2022. Patients were stratified into the HIPEC group (n=101) and the non-HIPEC group (n=126), based on whether they received postoperative P-HIPEC. Propensity score matching (PSM) was used to adjust for baseline characteristics, facilitating a comparative analysis of survival outcomes, postoperative complications and recurrence patterns. Cox regression analysis was performed to identify prognostic factors. Furthermore, the impact of varying P-HIPEC frequencies and initiation timings was evaluated. Results No significant differences in overall survival (OS) or postoperative complication rates were observed between the two groups in the original and PSM cohorts. But the disease-free survival (DFS) of the HIPEC group was significantly higher than that of the non-HIPEC group (HR 0.569; 95% CI 0.362-0.894; p = 0.013) in the PSM cohort, with 1-year, 3-year, and 5-year DFS rates showing notable improvement (77.9% vs. 69.7%, 60.1% vs. 43.0%, and 46.2% vs. 25.5%). The incidence of isolated peritoneal metastasis (PM) was significantly lower in the HIPEC group (5.3% vs. 17.3%, p = 0.039). Multivariate Cox regression analysis identified P-HIPEC as an independent protective factor for DFS. Further analysis indicated that neither the number of P-HIPEC sessions had a significant impact on OS (p = 0.388) or DFS (p = 0.735), nor did the timing of P-HIPEC initiation affect OS (p = 0.620) or DFS (p = 0.488). Likewise, different P-HIPEC frequencies or initiation timings had no significant impact on postoperative complication rates or recurrence patterns. Conclusion P-HIPEC effectively reduces the risk of postoperative PM and improves DFS in patients with locally AGC without increasing postoperative complications. However, it does not significantly impact OS. Additionally, variations in the frequency and timing of P-HIPEC initiation do not significantly affect survival outcomes, postoperative complications, or recurrence patterns.
Collapse
Affiliation(s)
- Zhijie Gong
- Second Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Liping Zhou
- Gastrointestinal Endoscopy Center, Maoming People’s Hospital, Maoming, China
| | - Yinghao He
- Second Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jun Zhou
- Second Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - Yanjie Deng
- The First Clinical College of Medicine, Guangdong Medical University, Zhanjiang, China
| | - Zudong Huang
- Second Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - WeiWei Wang
- Second Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - Qiangbang Yang
- Second Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - Jian Pan
- Second Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - Yingze Li
- Second Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| | - Xiaolu Yuan
- Department of Pathology, Maoming People’s Hospital, Maoming, China
| | - Minghui Ma
- Second Department of Gastrointestinal Surgery, Maoming People’s Hospital, Maoming, China
| |
Collapse
|
14
|
Li Z, Sun C, Fei H, Li Z, Zhao D, Guo C, Du C. Downstaging Effect Rather than the Full Intended Cycles of Perioperative Chemotherapy Determines the Value of Adjuvant Chemotherapy in Gastric Cancer. Ann Surg Oncol 2025; 32:342-350. [PMID: 39448412 DOI: 10.1245/s10434-024-16365-8] [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: 07/11/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Perioperative chemotherapy is the standard treatment modality for locally advanced gastric cancer. However, the efficacy and indication of adjuvant chemotherapy in patients who have already received neoadjuvant chemotherapy remain unclear. This study aims to explore the association between adjuvant chemotherapy with patient prognosis in those who have received neoadjuvant chemotherapy plus D2 gastrectomy in a real-world setting, and whether this association is affected by the duration of neoadjuvant treatment. PATIENTS AND METHODS A total of 174 patients with cT3-4N+ gastric cancer who had received neoadjuvant chemotherapy plus D2 radical gastrectomy were included in the study. Kaplan-Meier curves and log-rank tests were used to assess and compare the survival outcomes between patients who received adjuvant therapy and those who did not. RESULTS Patients who were younger age, had a lower American Society of Anesthesiologists (ASA) grade, did not experience postoperative complication, and received fewer than six cycles of neoadjuvant chemotherapy were more likely to receive adjuvant chemotherapy, rather than those with advanced ypTNM stage or poor tumor regression grade. Patients who received adjuvant therapy had a better overall survival (OS) (2-year OS rate 86.2% versus 64.1%, p = 0.002). Adjuvant therapy was associated with longer survival in patients who remained ypTNM stage III despite receiving at least six cycles of neoadjuvant chemotherapy. However, there was no significant longer survival observed in patients with ypTNM stages 0-II receiving adjuvant chemotherapy, even when they received less than six cycles of neoadjuvant chemotherapy. CONCLUSIONS Patients with locally advanced gastric cancer may still need adjuvant chemotherapy, even after receiving neoadjuvant chemotherapy. The value of adjuvant chemotherapy after neoadjuvant chemotherapy depends more on the actual downstaging effect achieved after neoadjuvant chemotherapy, rather than the completion of "full intended" cycles of perioperative treatment.
Collapse
Affiliation(s)
- Zefeng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chongyuan Sun
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - He Fei
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongbing Zhao
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Chunguang Guo
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Chunxia Du
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
15
|
Li Z, Zhang X, Sun C, Fei H, Li Z, Zhao D, Guo C, Du C. Evaluation of pathologic response and surgical safety of total neoadjuvant therapy for patients with clinical stage III gastric cancer in a real-world setting. J Gastrointest Surg 2024; 28:1597-1604. [PMID: 39019340 DOI: 10.1016/j.gassur.2024.07.008] [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: 04/29/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Perioperative chemotherapy is the standard treatment for locally advanced gastric cancer. However, the potential benefit of extending therapy before surgery remains largely unknown. In this study, we aimed to evaluate the efficacy and safety of total neoadjuvant chemotherapy, with or without immune checkpoint blockade. METHODS A cohort of 174 patients with clinical stage III gastric cancer who underwent D2 gastrectomy from October 2021 to March 2024 in the real-world setting were included in this study. Among these patients, 101 were treated with total neoadjuvant therapy (TNT) and 73 were treated with perioperative neoadjuvant therapy (PNT). We compared the pathologic complete response (pCR) rate, ypN0 rate, recurrence-free survival (RFS), overall survival (OS), and postoperative complications between the 2 groups. Multivariate logistic regression analysis was conducted to identify factors associated with pCR or ypN0. RESULTS Compared with the PNT group, the patients in the TNT group were more frequently treated with intensive chemotherapy with triplets + immunotherapy. Apart from this, there were no significant differences in baseline characteristics. There were no statistically significant differences in pCR (16.8% vs 12.3%), ypN0 (49.5% vs 38.4%), RFS, OS, and postoperative complications (27.7% vs 26.0%) between the TNT and PNT groups. Older age, diffuse type, and stable disease/progressive disease based on clinical efficacy evaluation were independently associated with non-pCR. Stable disease/progressive disease, linitis plastica, and poor differentiation were independently associated with ypN+. Neither the number of neoadjuvant therapy cycles nor the specific regimens were associated with pCR or ypN0. In the subgroup analysis of patients receiving total gastrectomy, there were still no statistically significant differences in pCR (16.7% vs 2.6%), ypN0 (43.8% vs 39.5%), and postoperative complications (45.8% vs 36.8%) between the 2 groups. CONCLUSION Although TNT did not increase the postoperative complication rate, it also did not provide any additional short-term benefits compared with PNT for clinical stage III gastric cancer.
Collapse
Affiliation(s)
- Zefeng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaojie Zhang
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chongyuan Sun
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - He Fei
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongbing Zhao
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Chunguang Guo
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxia Du
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Zheng Y, Qiu B, Liu S, Song R, Yang X, Wu L, Chen Z, Tuersun A, Yang X, Wang W, Liu Z. A transformer-based deep learning model for early prediction of lymph node metastasis in locally advanced gastric cancer after neoadjuvant chemotherapy using pretreatment CT images. EClinicalMedicine 2024; 75:102805. [PMID: 39281097 PMCID: PMC11402411 DOI: 10.1016/j.eclinm.2024.102805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/18/2024] Open
Abstract
Background Early prediction of lymph node status after neoadjuvant chemotherapy (NAC) facilitates promptly optimization of treatment strategies. This study aimed to develop and validate a deep learning network (DLN) using baseline computed tomography images to predict lymph node metastasis (LNM) after NAC in patients with locally advanced gastric cancer (LAGC). Methods A total of 1205 LAGC patients were retrospectively recruited from three hospitals between January 2013 and March 2023, constituting a training cohort, an internal validation cohort, and two external validation cohorts. A transformer-based DLN was developed using 3D tumor images to predict LNM after NAC. A clinical model was constructed through multivariate logistic regression analysis as a baseline for subsequent comparisons. The performance of the models was evaluated through discrimination, calibration, and clinical applicability. Furthermore, Kaplan-Meier survival analysis was conducted to assess overall survival (OS) of LAGC patients at two follow-up centers. Findings The DLN outperformed the clinical model and demonstrated a robust performance for predicting LNM in the training and validation cohorts, with areas under the curve (AUCs) of 0.804 (95% confidence interval [CI], 0.752-0.849), 0.748 (95% CI, 0.660-0.830), 0.788 (95% CI, 0.735-0.835), and 0.766 (95% CI, 0.717-0.814), respectively. Decision curve analysis exhibited a high net clinical benefit of the DLN. Moreover, the DLN was significantly associated with the OS of LAGC patients [Center 1: hazard ratio (HR), 1.789, P < 0.001; Center 2:HR, 1.776, P = 0.013]. Interpretation The transformer-based DLN provides early and effective prediction of LNM and survival outcomes in LAGC patients receiving NAC, with promise to guide individualized therapy. Future prospective multicenter studies are warranted to further validate our model. Funding National Natural Science Foundation of China (NO. 82373432, 82171923, 82202142), Project Funded by China Postdoctoral Science Foundation (NO. 2022M720857), Regional Innovation and Development Joint Fund of National Natural Science Foundation of China (NO. U22A20345), National Science Fund for Distinguished Young Scholars of China (NO. 81925023), Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application (NO. 2022B1212010011), High-level Hospital Construction Project (NO. DFJHBF202105), Natural Science Foundation of Guangdong Province for Distinguished Young Scholars (NO. 2024B1515020091).
Collapse
Affiliation(s)
- Yunlin Zheng
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Bingjiang Qiu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Sciences, Guangzhou, 510080, China
| | - Shunli Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong Province, 266000, China
| | - Ruirui Song
- Department of Radiology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Xianqi Yang
- Department of Gastric Surgery, and State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lei Wu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Zhihong Chen
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou, 510006, China
| | - Abudouresuli Tuersun
- Department of Radiology, The First People's Hospital of Kashi Prefecture, Kashi, 844700, China
| | - Xiaotang Yang
- Department of Radiology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Wei Wang
- Department of Gastric Surgery, and State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| |
Collapse
|
17
|
Jin L, Zhang L, Fu L, Song F, Cheng A. 18F-FDG PET/CT metabolism multi-parameter prediction of chemotherapy efficacy in locally progressive gastric cancer. Ann Nucl Med 2024; 38:475-482. [PMID: 38536655 PMCID: PMC11108936 DOI: 10.1007/s12149-024-01921-9] [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: 11/19/2023] [Accepted: 03/03/2024] [Indexed: 05/22/2024]
Abstract
PURPOSE This study aimed to use an 18F-FDG PET/CT multiparametric quantitative analysis to determine the efficacy of neoadjuvant chemotherapy in patients with locally progressive gastric cancer. MATERIALS AND METHODS We conducted a retrospective analysis of 34 patients with pathologically identified gastric cancer who received neoadjuvant chemotherapy and surgery. Chemotherapy regimens were followed and 18F-FDG PET/CT was conducted. We ascertained multiparamaters of the target lesions pre- and post-treatment and determined the ideal cutoff values for the percentage change in biomarkers. Independent factors were evaluated using binary logistic regression. A response classification system was used to explore the association between metabolic and anatomical responses and the degree of pathological remission. RESULTS Binary logistic regression analysis showed that Lauren bowel type and change in total lesion glycolysis >45.2% were risk predictors for the efficacy of neoadjuvant chemotherapy; total lesion glycolysis demonstrated the best predictive efficacy. The categorical variable system of the two-module response (metabolic and anatomical response) group had a higher predictive accuracy than that of the single-module response (metabolic or anatomical response) group. CONCLUSIONS Using 18F-FDG PET/CT multiparametric quantitative analysis, Lauren bowel type and change in total lesion glycolysis >45.2% were independent predictors of the efficacy of neoadjuvant chemotherapy in patients with gastric adenocarcinoma. Additionally, the dual-module assessment demonstrated high predictive efficacy.
Collapse
Affiliation(s)
- Luqiang Jin
- Jinzhou Medical University Postgraduate Training Base (Zhejiang Provincial People's Hospital), Hangzhou, Zhejiang, China
| | - Linghe Zhang
- Cancer Center, Department of Nuclear Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang, China
| | - Liping Fu
- Cancer Center, Department of Nuclear Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang, China
| | - Fahuan Song
- Cancer Center, Department of Nuclear Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang, China
| | - Aiping Cheng
- Cancer Center, Department of Nuclear Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang, China.
| |
Collapse
|
18
|
Bhandare MS, Gundavda KK, Yelamanchi R, Chopde A, Batra S, Kolhe M, Ramaswamy A, Ostwal V, Deodhar K, Chaudhari V, Shrikhande SV. Impact of pCR after neoadjuvant chemotherapy and radical D2 dissection in locally advanced gastric cancers: Analysis of 1001 cases. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108343. [PMID: 38640606 DOI: 10.1016/j.ejso.2024.108343] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Advances in perioperative chemotherapy have improved outcomes in patients with gastric cancers (GC). This strategy leads to tumour downstaging and may result in a pathologic complete response (pCR). The study aimed to evaluate the predictors of pCR and determine the impact of pCR on long-term survival. METHODS At the Department of Gastrointestinal and HPB Oncology at the Tata Memorial Centre, Mumbai, 1001 consecutive patients with locally advanced GCs undergoing radical resection following neoadjuvant chemotherapy from January 2005 to June 2022 were included. RESULTS At a median follow-up of 61 months, the median OS was 53 months with a 5-year OS of 46.8 %. Ninety-five patients (9.49 %) realized pCR. Non-signet and well-differentiated histology were associated with pCR. pCR was significantly associated with improved OS, 5-year OS 79.2 % vs 43.2 % (HR 0.30, p < 0.001). On multivariable analysis, the realization of pCR and completion of adjuvant chemotherapy had superior OS. Whereas, signet-ring histology, linitis-like tumours, and high lymph node ratio had adverse outcomes. CONCLUSION Tumour grade and signet-ring histology predict achievement of pCR in locally advanced GCs after neoadjuvant chemotherapy. Patients with pCR have significantly improved survival. Future neoadjuvant strategies should focus on enhancing pCR rates to improve overall outcomes.
Collapse
Affiliation(s)
- Manish S Bhandare
- Department of Gastrointestinal and Hepato-pancreato-biliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Kaival K Gundavda
- Department of Gastrointestinal and Hepato-pancreato-biliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Raghav Yelamanchi
- Department of Gastrointestinal and Hepato-pancreato-biliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Amit Chopde
- Department of Gastrointestinal and Hepato-pancreato-biliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Swati Batra
- Department of Surgical Oncology, Armed Forces Medical Services (Army Hospital, Research and Referral), Delhi, India.
| | - Manjushree Kolhe
- Department of Statistics, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Kedar Deodhar
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Vikram Chaudhari
- Department of Gastrointestinal and Hepato-pancreato-biliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Shailesh V Shrikhande
- Department of Gastrointestinal and Hepato-pancreato-biliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| |
Collapse
|
19
|
Liu B, Yin X, Cai Z, Shen C, Jiang T, Han Y, Yin Y, Zhang B. Neoadjuvant and/or adjuvant chemotherapy for gastric cancer patients with microsatellite instability or deficient mismatch repair: a systematic review and meta-analysis study protocol. BMJ Open 2024; 14:e084496. [PMID: 38670615 PMCID: PMC11057263 DOI: 10.1136/bmjopen-2024-084496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
INTRODUCTION Whether gastric cancer (GC) patients with deficient mismatch repair or microsatellite instability-high (dMMR/MSI-H) benefit from perioperative (neoadjuvant and/or adjuvant) chemotherapy is controversial. This protocol delineates the planned scope and methods for a systematic review and meta-analysis that aims to compare the efficacy of perioperative chemotherapy with surgery alone in resectable dMMR/MSI-H GC patients. METHODS AND ANALYSIS This study protocol is reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Protocols-P guideline. PubMed, Embase, Cochrane (CENTRAL), and the Web of Science databases will be searched, supplemented by a secondary screening of relevant records. Both randomised controlled trials and non-randomised studies will be included in this study. The primary and secondary outcomes under scrutiny will be overall survival, disease-free survival and progression-free survival. Two reviewers will independently screen studies, extract data and assess the risk of bias. We will analyse different treatment settings (eg, neoadjuvant or adjuvant or combined as perioperative chemotherapies) separately and conduct sensitivity analyses. ETHICS AND DISSEMINATION No ethics approval is required for this systematic review and meta-analysis, as no individual patient data will be collected. The findings of our study will be published in a peer-reviewed journal. PROSPERO REGISTRATION NUMBER CRD42023494276.
Collapse
Affiliation(s)
- Baike Liu
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaonan Yin
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhaolun Cai
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chaoyong Shen
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tianxiang Jiang
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yihui Han
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yuan Yin
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Department of Gastrointestinal Surgery, West China Xiamen Hospital of Sichuan University, Xiamen, People's Republic of China
| | - Bo Zhang
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
20
|
Zheng HL, Wei LH, Xu BB, Zheng HH, Xue Z, Chen QY, Xie JW, Zheng CH, Huang CM, Lin JX, Li P. Prognostic value of preoperative sarcopenia in gastric cancer: A 10-year follow-up study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108004. [PMID: 38330540 DOI: 10.1016/j.ejso.2024.108004] [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: 10/05/2023] [Revised: 12/26/2023] [Accepted: 02/02/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Preoperative sarcopenia is associated with prognosis in patients with gastric cancer (GC); however, studies with 10-year survival follow-up are lacking. METHODS Consecutive patients with GC who underwent radical gastrectomy between December 2009-2012 were included retrospectively. Preoperative sarcopenia was diagnosed using computed tomography skeletal muscle index. The Kaplan-Meier method estimated overall survival (OS) and relapse-free survival (RFS). Cox proportional hazard regression analysis determined the prognostic factors for OS and RFS. RESULTS In total, 781 patients with GC were included; among these, 207 (26.5%) had preoperative sarcopenia. Patients with sarcopenia had significantly lower 10-year OS and RFS than patients without sarcopenia (39.61% vs. 58.71% and 39.61% vs. 57.84%, respectively). Further, preoperative sarcopenia was an independent risk factor for 10-year OS (HR = 1.467; 95% confidence interval [CI]: 1.169-1.839) and RFS (HR = 1.450; 95% CI: 1.157-1.819). Patients with sarcopenia had a higher risk of death and recurrence in the first 10 years postoperatively than patients without sarcopenia. Additionally, the risk of death (HR = 2.62; 95% CI:1.581-4.332) and recurrence (HR = 2.34; 95% CI:1.516-3.606) was the highest in the 1st postoperative year and remained relatively stable thereafter. Further, postoperative adjuvant chemotherapy significantly improved 10-year OS (p = 0.006; HR = 0.558) and RFS (p = 0.008; HR = 0.573) in patients with TNM stage II-III GC that presented with sarcopenia. CONCLUSION Preoperative sarcopenia remained an independent risk factor for postoperative very long-term prognosis of GC. Postoperative adjuvant chemotherapy improved the long-term outcomes of stage II-III patients with sarcopenia.
Collapse
Affiliation(s)
- Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Ling-Hua Wei
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Bin-Bin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Hong-Hong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Zhen Xue
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China.
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China.
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China.
| |
Collapse
|
21
|
Zheng HL, Wang FH, Zhang LK, Li P, Zheng CH, Chen QY, Huang CM, Xie JW. Trajectories of neutrophil-to-lymphocyte ratios during neoadjuvant chemotherapy correlate with short- and long-term outcomes in gastric cancer: a group-based trajectory analysis. BMC Cancer 2024; 24:226. [PMID: 38365617 PMCID: PMC10873962 DOI: 10.1186/s12885-024-11950-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 02/04/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Systemic inflammatory factors can predict the survival prognosis of gastric cancer (GC) patients after neoadjuvant chemotherapy (NACT). However, whether longitudinal changes in systemic inflammatory factors are associated with short - and long-term outcomes has not been reported. METHODS This study is a retrospective analysis of 216 patients with advanced gastric cancer who received NACT between January 2011 and June 2019, comparing receiver operating characteristic (ROC) curves for screening suitable inflammatory markers. Group-based trajectory modeling (GBTM) was used to analyze longitudinal changes in inflammatory markers during NACT to identify different potential subgroups and to compare postoperative complications, recurrence-free survival (RFS), and overall survival (OS) among subgroups. RESULTS Ultimately, neutrophil-lymphocyte ratio (NLR) had the highest area under the curve (AUC) value in predicting prognosis was included in the GBTM analysis. Three trajectories of NLR were obtained: Stable group (SG) (n = 89), Ascent-descend group (ADG) (n = 80) and Continuous descend group (CDG) (n = 47). Compared with SG, ADG and CDG are associated with an increased risk of postoperative recurrence and death. The median time of RFS and OS of SG was longer than that of ADG and CDG (median RFS 81 vs. 44 and 22 months; median OS 69 vs. 41 and 30 months). In addition, CDG had significantly higher postoperative serious complications than SG and ADG (17 (36.2%) vs. 17 (19.1%) and 12 (15.0%); p = 0.005). CONCLUSION There were different trajectories of NLR during NACT, and these potential trajectories were significantly associated with severe postoperative complications, recurrence, and mortality in patients with GC.
Collapse
Affiliation(s)
- Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fu-Hai Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ling-Kang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China.
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China.
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China.
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
22
|
Chen M, Yu S, Chen C, Liang J, Zhou D. Development and evaluation of the Newstage system: integrating tumor regression grade and lymph node status for improved prognostication in neoadjuvant treatment of gastric cancer. World J Surg Oncol 2024; 22:16. [PMID: 38195570 PMCID: PMC10777530 DOI: 10.1186/s12957-023-03291-4] [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: 09/25/2023] [Accepted: 12/26/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND The predictive correlation of tumor depth of invasion changes after neoadjuvant therapy, and the 8th American Joint Committee on Cancer (AJCC) ypTNM system for gastric cancer may not accurately predict patient prognosis following neoadjuvant therapy. METHODS A retrospective analysis was conducted on a total of 258 patients who underwent radical surgery for gastric cancer after neoadjuvant therapy. The Newstage system was established based on tumor regression grade and pathological lymph node status. The 3-year survival rates of patients classified by the Newstage system were compared with those classified by the AJCC ypTNM system. RESULTS In a cohort of 258 patients, the 3-year overall survival rates based on the Newstage system were: (I) 94.6%, (II) 79.3%, (III) 54.5%, and (IV) 30.2%. The Newstage system exhibited a lower Akaike information criterion value (902.57 vs. 912.03). Additionally, the area under the ROC curve (0.756 vs. 0.733) and the C-index (0.731 vs. 0.718) was higher than the AJCC ypTNM system. Furthermore, a multivariate analysis indicated that the Newstage system was an independent prognostic factor (p = 0.001). CONCLUSION The Newstage system exhibits superior predictive performance in estimating survival rates for neoadjuvant therapy in gastric cancer. It also functions as an independent prognostic factor.
Collapse
Affiliation(s)
- Ming Chen
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shanshan Yu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Cheng Chen
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jinxiao Liang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Donghui Zhou
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
23
|
Liu L, Wang Y, Liu T, Rao S, Zeng M. The largest lymph node defined response to neoadjuvant chemotherapy can predict long-term prognosis in locally advanced gastric cancer. Abdom Radiol (NY) 2023; 48:3653-3660. [PMID: 37755476 DOI: 10.1007/s00261-023-04048-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND The assessment of tumor response to neoadjuvant chemotherapy (NACT) in locally advanced gastric cancer (LAGC) remain challenging. We aimed to explore the potential role of peri-NACT change of the largest lymph node (LN) and primary tumor (P-T) in the prediction of tumor response and patient overall survival (OS) in LAGC. METHODS A cohort of LAGC patients who underwent NACT followed by radical surgery from a prospective clinical trial were retrospectively analyzed. The percentage change of the largest LN and P-T from initial to post-NACT Computed Tomography (CT) were measured. Tumor response was defined by the change of LN (LN-response) and P-T (P-T-response), respectively. A multivariate Cox model was constructed to examine if P-T- and LN-determined response had significant predictive ability for OS when adjusting with other possible prognostic factors. RESULTS Of the 41 patients, 28 (68.3%) was defined as LN-responders to NACT, and 17 (41.5%) patients was defined as P-T-responders. When the cohort was stratified by LN response standard, LN-responders showed a significant longer median OS than LN-nonresponders (p = 0.031, 20.6 vs 16.6 months). When stratified by primary tumor response, no significant difference in OS was observed between P-T-responders and P-T-nonresponders (p = 0.377, 18.5 vs 19.0 months). In the multivariate analysis, number of positive LN (p = 0.004, hazard ratio [HR] = 1.284), recurrence (p = 0.024, HR =3556), LN shrinkage (p = 0.022, HR = 0.930) and LN-response (p = 0.033, HR = 0.008) were observed with independent association with OS. CONCLUSIONS Peri-NACT change of the largest LN could reflect tumor response to NACT, and LN-defined response was useful in predicting the long-term prognosis (OS) of LAGC patients who underwent NACT followed by radical surgery.
Collapse
Affiliation(s)
- Liheng Liu
- Department of Radiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China
| | - Yan Wang
- Cancer Center, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China
| | - Tianshu Liu
- Cancer Center, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China.
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China.
| | - Shengxiang Rao
- Department of Radiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China.
- Shanghai Institute of Medical Imaging, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China.
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, China
| |
Collapse
|
24
|
Takayama T, Tsuji Y. Updated Adjuvant Chemotherapy for Gastric Cancer. J Clin Med 2023; 12:6727. [PMID: 37959193 PMCID: PMC10648766 DOI: 10.3390/jcm12216727] [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: 08/25/2023] [Revised: 10/03/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Surgical resection is currently the best curative approach for gastric cancer (GC); however, the prognosis of patients with advanced GC remains poor even with curative resection. For this reason, perioperative chemotherapy has been combined with surgery to reduce the risk of postoperative recurrence. Standard perioperative chemotherapy for resectable advanced GC varies from region to region. Postoperative S-1 therapy was standardized via the ACTS-GC study in East Asia, perioperative ECF (Epirubicin + Cisplatin + Fluorouracil) was standardized via the MAGIC study in Europe, and postoperative chemoradiotherapy was standardized via the US intergroup study in North America. Since then, more intensive regimens have been developed. In recent years, perioperative therapy using novel agents, such as molecular-targeted drugs and immune checkpoint inhibitors (ICIs), has also been tested and evaluated in the three major regions (East Asia, Europe, and North America) with promising results. Perioperative chemotherapy has become an integral part of many treatment strategies and requires continued research and evaluation.
Collapse
Affiliation(s)
- Toshizo Takayama
- Department of Medical Oncology, Tonan Hospital, Sapporo 060-0004, Japan
- Department of Medical Oncology, Daido Hospital, Nagoya 457-8511, Japan
| | - Yasushi Tsuji
- Department of Medical Oncology, Tonan Hospital, Sapporo 060-0004, Japan
| |
Collapse
|
25
|
Fiflis S, Papakonstantinou M, Giakoustidis A, Christodoulidis G, Louri E, Papadopoulos VN, Giakoustidis D. Comparison between upfront surgery and neoadjuvant chemotherapy in patients with locally advanced gastric cancer: A systematic review. World J Gastrointest Surg 2023; 15:1808-1818. [PMID: 37701690 PMCID: PMC10494580 DOI: 10.4240/wjgs.v15.i8.1808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/07/2023] [Accepted: 06/27/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is a major health concern worldwide. Surgical resection and chemotherapy is the mainstay treatment for gastric carcinoma, however, the optimal approach remains unclear and should be different in each individual. Chemotherapy can be administered both pre- and postoperatively, but a multidisciplinary approach is preferred when possible. This is particularly relevant for locally advanced GC (LAGC), as neoadjuvant chemotherapy (NAT) could potentially lead to tumor downsizing thus allowing for a complete resection with curative intent. Even though the recent progress has been impressive, European and International guidelines are still controversial, thus attenuating the need for a more standardized approach in the management of locally advanced cancer. AIM To investigate the effects of NAT on the overall survival (OS), the disease-free survival (DFS), the morbidity and the mortality of patients with LAGC in comparison to upfront surgery (US). METHODS For this systematic review, a literature search was conducted between November and February 2023 in PubMed, Cochrane Library and clinicaltrials.gov for studies including patients with LAGC. Two independent reviewers conducted the research and extracted the data according to predetermined inclusion and exclusion criteria. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses was used to form the search strategy and the study protocol has been registered in the International Prospective Register of Systematic Reviews. RESULTS Eighteen studies with 4839 patients with LAGC in total were included in our systematic review. Patients were separated into two groups; one receiving NAT before the gastrectomy (NAT group) and the other undergoing upfront surgery (US group). The OS ranged from 41.6% to 74.2% in the NAT group and from 30.9% to 74% in the US group. The DFS was also longer in the NAT group and reached up to 80% in certain patients. The complications related to the chemotherapy or the surgery ranged from 6.4% to 38.1% in the NAT group and from 5% to 40.5% in the US group. Even though in most of the studies the morbidity was lower in the NAT group, a general conclusion could not be drawn as it seems to depend on multiple factors. Finally, regarding the mortality, the reported rate was higher and up to 5.3% in the US group. CONCLUSION NAT could be beneficial for patients with LAGC as it leads to better OS and DFS than the US approach with the same or even lower complication rates. However, patients with different clinicopathological features respond differently to chemotherapy, therefore currently the treatment plan should be individualized in order to achieve optimal results.
Collapse
Affiliation(s)
- Stylianos Fiflis
- A’ Department of Surgery, General Hospital Papageorgiou, Thessaloniki 56429, Greece
| | | | | | | | - Eleni Louri
- A’ Department of Surgery, General Hospital Papageorgiou, Thessaloniki 56429, Greece
| | | | | |
Collapse
|
26
|
Graziosi L, Natalizi N, Donini A. Rationale in the Use of Adjuvant Chemotherapy in pT3N0M0 Gastric Cancer Resected Patients. Comment on Chen et al. Prognostic Factors and the Role of Adjuvant Chemotherapy in Pathological Node-Negative T3 Gastric Cancer. J. Pers. Med. 2023, 13, 553. J Pers Med 2023; 13:974. [PMID: 37373963 DOI: 10.3390/jpm13060974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
I read with great interest the well-written and well-made study by Yi-Fu Chen et al. recently published in the "Journal of Personalized Medicine" [...].
Collapse
Affiliation(s)
- Luigina Graziosi
- Emergency and General Surgery, "Santa Maria della Misericordia" Hospital, University of Perugia, 06129 Perugia, Italy
| | - Nicola Natalizi
- Emergency and General Surgery, "Santa Maria della Misericordia" Hospital, University of Perugia, 06129 Perugia, Italy
| | - Annibale Donini
- Emergency and General Surgery, "Santa Maria della Misericordia" Hospital, University of Perugia, 06129 Perugia, Italy
| |
Collapse
|
27
|
Luo D, Ye Z, Zhang R, Li Q, Li X. Use of lymph node ratio to guide clinical decision-making concerning adjuvant radiotherapy in pT1-2N1 rectal cancer. Int J Colorectal Dis 2023; 38:115. [PMID: 37148381 DOI: 10.1007/s00384-023-04415-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/08/2023]
Abstract
PURPOSE Lymph node metastases are uncommon in pT1-2 rectal cancer. pT1-2N1 are often characterized with low tumor burden and intermediate prognosis. Therefore, adjuvant radiotherapy (ART) is controversial in these patients. This study aimed to investigate the value of ART in pT1-2 rectal cancer and evaluate the guiding role of lymph node ratio (LNR) for utilization of ART. METHODS pT1-2N1 rectal cancer patients who received surgery without neoadjuvant radiotherapy between 2000 and 2018 with at least 12 lymph node harvest were extracted from the Surveillance, Epidemiology and End Results (SEER) database. We used time-dependent receiver operating characteristic (ROC) analysis to determine the optimal cutoff of LNR. Kaplan-Meier methods and Cox proportional hazards regression models were performed to determine the prognostic value of ART in pT1-2N1 rectal cancer patients and subgroups stratified by LNR. RESULTS A total of 674 and 1321 patients with pT1N1 and pT2N1 rectal cancer were eligible for analysis. There was no statistical cancer-specific survival (CSS) difference in pT1N1 rectal cancer patients between receiving and not receiving ART (P = 0.464). The 5-year CSS was 89.6% and 83.2% in pT2N1 rectal cancer patients between receiving and not receiving ART, respectively (P = 0.003). A total of 7.0% was identified as the optimal cutoff value of LNR. Survival improvement offered by ART was only found in LNR ≥ 7.0% subgroup (5-year CSS: 89.5% versus 79.6%, P = 0.003) instead of LNR < 7.0% subgroup (5-year CSS: 89.9% versus 86.3%, P = 0.208). CONCLUSION ART show substantial survival benefit in pT2N1 rectal cancer patients with LNR ≥ 7.0%, warranting the conventional adoption of ART in this subgroup.
Collapse
Affiliation(s)
- Dakui Luo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China
| | - Zilan Ye
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China
| | - Ruijia Zhang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China.
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China.
| |
Collapse
|
28
|
Pradhan SP, Agarwala S, Sahoo J, Pradhan SK, Jena S, Satpathy N, Epari V. Survival of Gastric Cancer Patients at a Tertiary Care Hospital in Eastern India: A Retrospective Data Analysis. Cureus 2023; 15:e37064. [PMID: 37153312 PMCID: PMC10155757 DOI: 10.7759/cureus.37064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2023] [Indexed: 04/05/2023] Open
Abstract
Background Gastric cancer is one of the most common cancers and a leading cause of death worldwide. Most cases of gastric cancer are diagnosed at an advanced stage when no definitive treatment is available leading to an overall declined survival rate. In this study, we aimed to investigate the survival rate of gastric cancer patients admitted to our tertiary care center and determined the relationship between sociodemographic and clinicopathological characteristics with mortality. Methodology Gastric cancer patients treated between January 2019 and December 2020 were included in this retrospective study. The clinicopathological and demographic data of 275 gastric cancer patients were analyzed. The Kaplan-Meier method was used to calculate the overall survival of gastric cancer patients. The Kaplan-Meier log-rank test was used to calculate the difference. Results The mean survival of gastric cancer patients was 20.10 months (95% confidence interval = 19.20-21.03). Deaths were higher among stage III (42.6%) and stage IV (36.1%) patients compared to stage I (1.6%) and stage II (19.7%) patients. Mortality was significantly higher (70.5%) in patients without surgery. Conclusions The mean survival in our study setting is lower and is associated with the pathological stage of the disease, surgical intervention, and patients presenting with other gastrointestinal symptoms. A lower survival rate can be attributed to late diagnosis.
Collapse
|
29
|
Oganyan KA, Musaelyan AA, Lapin SV, Kupenskaya TV, Sveсhkova AA, Belyaev MA, Zakharenko AA, Orlov SV. Molecular markers as predictors of response to perioperative chemotherapy in locally advanced gastric cancer. ADVANCES IN MOLECULAR ONCOLOGY 2023. [DOI: 10.17650/2313-805x-2023-10-1-40-48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Introduction. Perioperative FLOT chemotherapy has improved prognosis in patients with locally advanced resectable gastric cancer (GC). However, in 80 % of cases, the tumor is resistant to the therapy, resulting in unnecessary toxicity and delayed surgical treatment.Aim. Evaluation of clinico-morphological patterns of microsatellite instability, HER2 gene amplification, changes in gene copy number and their relationship with the response to perioperative FLOT chemotherapy in patients with locally advanced resectable GC.Materials and methods. The retrospective study included 185 patients. All tumor samples were assessed for HER2 and microsatellite instability status. Among all cases there were 45 patients with locally advanced T2–4N1–2 M0 GC, who underwent a total or subtotal gastrectomy with D2 lymphadenectomy and perioperative chemotherapy with FLOT. Microsatellite instability detection was performed using fragment analysis, HER2 gene amplification testing – fluorescent in situ hybridization. Also 19 patients were tested for copy number changes of the FGFR1, FGFR2, KRAS, MET, EGFR, CCND1, MYC genes using Multiplex ligation-dependent probe amplification. The endpoints were progression-free survival and objective response rate.Results. Microsatellite instability was detected in 4.8 % (9/185) of GC cases. Microsatellite instability was associated with advanced age (p = 0.005), low grade of differentiation (p = 0.011), presence of tumor-infiltrating lymphocytes (p = 0.0004), and high preoperative CA 72–4 levels (p = 0.025). Prevalence of HER2 amplification was 7.5 % (14/185). It was associated with low grade of differentiation (p = 0.048) and metastasis in regional lymph nodes (p = 0.037). PFS in patients with HER2-positive (HER2 – human epidermal growth factor receptor 2) GC treated with perioperative FLOT chemotherapy (4/45) was significantly lower than in patients with HER2-negative GC: the median was 156 and 317 days, respectively (hazard ratio 0.49; 95 % confidence interval 0.16–1.47; p = 0.0006). There was no correlation between the presence of the alteration and ORR (p = 1.0). Progression-free survival in GC patients with KRAS amplification (3/19) was significantly lower comparing with patients without it: the median was 98 and 327 days, respectively (hazard ratio 0.29; 95 % confidence interval 0.07–1.19; p <0.0001). There was no association between an increase in KRAS copy number and objective response rate (p = 1.0). For microsatellite instability and other studied markers no statistically significant correlation with progression-free survival and objective response rate was found (p >0.05).Conclusion. The presence of HER2 and KRAS amplification have been shown as promising predictive markers of the treatment failure in patients treated with perioperative FLOT chemotherapy for locally advanced resectable GC.
Collapse
Affiliation(s)
- K. A. Oganyan
- Pavlov First Saint Petersburg State Medical University
| | - A. A. Musaelyan
- Pavlov First Saint Petersburg State Medical University; Research Institute of Medical Primatology
| | - S. V. Lapin
- Pavlov First Saint Petersburg State Medical University
| | | | | | - M. A. Belyaev
- Pavlov First Saint Petersburg State Medical University
| | | | - S. V. Orlov
- Pavlov First Saint Petersburg State Medical University; Research Institute of Medical Primatology
| |
Collapse
|
30
|
Zhu L, Ma M, Zhang L, Wang S, Guo Y, Ling X, Lin H, Lai N, Lin S, Du L, Dong Q. System Analysis Based on Lipid-Metabolism-Related Genes Identifies AGT as a Novel Therapy Target for Gastric Cancer with Neoadjuvant Chemotherapy. Pharmaceutics 2023; 15:810. [PMID: 36986671 PMCID: PMC10051152 DOI: 10.3390/pharmaceutics15030810] [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] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most common causes of cancer-related deaths worldwide, and chemotherapy is still a standard strategy for treating patients with advanced GC. Lipid metabolism has been reported to play an important role in the carcinogenesis and development of GC. However, the potential values of lipid-metabolism-related genes (LMRGs) concerning prognostic value and the prediction of chemotherapy responsiveness in GC remains unclear. A total of 714 stomach adenocarcinoma patients were enrolled from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database. Using univariate Cox and LASSO regression analyses, we developed a risk signature based on LMRGs that can distinguish high-GC-risk patients from low-risk patients with significant differences in overall survival. We further validated this signature prognostic value using the GEO database. The R package "pRRophetic" was applied to calculate the sensitivity of each sample from high- and low-risk groups to chemotherapy drugs. The expression of two LMRGs, AGT and ENPP7, can predict the prognosis and response to chemotherapy in GC. Furthermore, AGT significantly promoted GC growth and migration, and the downregulation of AGT enhanced the chemotherapy response of GC both in vitro and in vivo. Mechanistically, AGT induced significant levels of epithelial-mesenchymal transition (EMT) through the PI3K/AKT pathway. The PI3K/AKT pathway agonist 740 Y-P can restore the EMT of GC cells impaired by AGT knockdown and treatment with 5-fluorouracil. Our findings suggest that AGT plays a key role in the development of GC, and targeting AGT may help to improve the chemotherapy response of GC patients.
Collapse
Affiliation(s)
- Le Zhu
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Ming Ma
- Gastroenterology Department of Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Lumin Zhang
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Shun Wang
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Yu Guo
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Xinxin Ling
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Hanchao Lin
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Nannan Lai
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Shengli Lin
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University & Shanghai Collaborative Innovation Center of Endoscopy, Shanghai 200032, China
| | - Ling Du
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Qiongzhu Dong
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai 201199, China
| |
Collapse
|
31
|
Tong X, Zhi P, Lin S. Neoadjuvant Chemotherapy in Asian Patients With Locally Advanced Gastric Cancer. J Gastric Cancer 2023; 23:182-193. [PMID: 36750998 PMCID: PMC9911622 DOI: 10.5230/jgc.2023.23.e12] [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: 10/25/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 02/09/2023] Open
Abstract
Presently, surgery is the only treatment approach for gastric cancer and improving the prognosis of locally advanced gastric cancer is one of the key factors in promoting gastric cancer survival benefit. The MAGIC study was the first to demonstrate the efficacy of neoadjuvant chemotherapy (NAC) in European countries. In recent years, several clinical trials have provided evidence for the use of NAC in Asian patients with locally advanced gastric cancer. However, clinical practice guidelines vary between Asian and non-Asian populations. Optimal NAC regimens, proper target populations, and predictors of NAC outcomes in Asian patients are still under investigation. Herein, we summarized the current progress in the administration of NAC in Asian patients with gastric cancer.
Collapse
Affiliation(s)
- Xie Tong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Peng Zhi
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Shen Lin
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|