1
|
Stroobant EE, Kong SH, Bencivenga M, Kinoshita T, Kim TH, Sano T, de Manzoni G, Yang HK, Kitagawa Y, Strong VE. Korea, Japan, Europe, and the United States: Why are guidelines for gastric cancer different? Gastric Cancer 2025; 28:559-568. [PMID: 40240698 DOI: 10.1007/s10120-025-01613-x] [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: 11/27/2024] [Accepted: 03/29/2025] [Indexed: 04/18/2025]
Abstract
As a global health concern, gastric cancer management has been systematized by individual countries and regions into regimented guidelines. To explore international differences, we examined the guidelines of Korea, Japan, Europe, and the United States. Guidelines are created by experts in the field, focusing on evidence-based recommendations to standardize and improve patient care, but the methodology for guideline creation, incorporation of new innovations, and review differs significantly. National and regional differences within the guidelines are apparent, stemming from various factors including local incidence, stage, presentation, patient preferences, and governmental influences. Differences include the use of neoadjuvant chemotherapy, criteria for endoscopic resection, and extent of lymphadenectomy. Nonetheless, fundamental treatment principles remain universal, and the goals of national guidelines are uniform: standardizing patient care, providing the highest quality treatments, incorporating cutting-edge clinical trial results, and consensus in guidelines to help formulate governmental policies. This review highlights how the guidelines are constructed, the unique elements of each guideline, how they differ, and why they differ.
Collapse
Affiliation(s)
- Emily E Stroobant
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, South Korea
| | - Maria Bencivenga
- General and Upper GI Surgery Unit, Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, Verona, Italy
| | - Takahiro Kinoshita
- Department of Gastric Surgery, National Cancer Center Hospital East, Chiba, Japan
| | - Tae-Han Kim
- Department of Surgery, Gyeongsang National University Changwon Hospital, Changwon, Gyeongsangnam-do, South Korea
| | - Takeshi Sano
- Department of Gastroenterological Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Giovanni de Manzoni
- General and Upper GI Surgery Unit, Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, Verona, Italy
| | - Han-Kwang Yang
- Department of Surgery, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, South Korea
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Vivian E Strong
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
2
|
Ma Y, Li Z, Wei C, Zhang J, Fu Q, Zhang Z, Shang C, Wang J, Wan X, Zhang B, Zhang Y, Li J, Zhang H, Bie L, Xia Q, Luo S, Li N. Neoadjuvant camrelizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastroesophageal junction adenocarcinoma: a single-arm, phase 2 trial. Gastric Cancer 2025; 28:652-661. [PMID: 40184004 DOI: 10.1007/s10120-025-01606-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND The impact of neoadjuvant combined chemotherapy, immunotherapy, and targeted therapy on pathologic responses and survival outcomes in HER2-positive locally advanced gastric cancer remains unclear. PATIENTS AND METHODS In this single-arm, phase 2 trial, patients with HER2-positive resectable cT4 and/or N + M0 gastric or gastroesophageal junction (G/GEJ) adenocarcinoma received four cycles of neoadjuvant camrelizumab plus trastuzumab and CapOx, followed by D2 gastrectomy and four cycles of CapOx. The primary endpoint was pathological complete response (pCR, ypT0N0) rate. RESULTS Twenty-five patients were enrolled and received neoadjuvant combination treatment. Of these patients, 11 (44%) were in cT3 and 14 (56%) in cT4a; all had positive nodal status. Of the 23 patients who underwent surgery, 5 (21.7%, 95% CI: 7.5-43.7) achieved pCR (ypT0N0), and 7 (30.4%, 95% CI: 13.2-52.9) achieved near pCR (ypT0). The R0 resection rate was 100%. During a median follow-up of 41.0 months, no patients with pCR had recurrence or death. In contrast, five of 18 patients with non-pCR had recurrence, and four of them died. The three-year disease-free survival rate was 78.3%. During neoadjuvant treatment, grade 3 adverse events were observed in 36% of patients, with no grade 4 or 5 adverse events reported. No treatment-related surgical delay or reoperation occurred. CONCLUSION Neoadjuvant camrelizumab plus trastuzumab and chemotherapy demonstrated favorable response and tolerable safety in HER2-positive G/GEJ adenocarcinoma.
Collapse
Affiliation(s)
- Yijie Ma
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China
| | - Zhi Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Chen Wei
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China
| | - Jian Zhang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Qiang Fu
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Zhandong Zhang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Chuang Shang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Jinbang Wang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Xiangbin Wan
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Bin Zhang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Yongchao Zhang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Jing Li
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - He Zhang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Liangyu Bie
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China
| | - Qingxin Xia
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China
| | - Suxia Luo
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China
| | - Ning Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China.
| |
Collapse
|
3
|
Chen QX, Zhang YB, Zeng WM, Cai YC, Lv CB, Lian MQ, Huang RJ, Lian MJ, Lian WL, Xu QH, Sun YQ, Cai LS. Efficacy and safety of sintilimab combined with nab-paclitaxel plus S-1 for neoadjuvant treatment of locally advanced gastric cancer. World J Gastrointest Surg 2025; 17:106361. [DOI: 10.4240/wjgs.v17.i6.106361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/28/2025] [Accepted: 05/13/2025] [Indexed: 05/30/2025] Open
Abstract
BACKGROUND Gastric cancer is a leading global cause of cancer mortality, with poor survival in locally advanced stages. While immune checkpoint inhibitors (ICIs) like sintilimab have improved outcomes in advanced disease, their role as neoadjuvant therapy remains understudied. This study investigates sintilimab combined with nab-paclitaxel/S-1 as preoperative treatment for locally advanced gastric cancer (LAGC), addressing an unmet need for effective neoadjuvant strategies.
AIM To explore the efficacy and safety of combination treatment with sintilimab and nab-paclitaxel plus S-1 as neoadjuvant therapy for LAGC.
METHODS Clinical data from 82 patients diagnosed with LAGC, who underwent preoperative treatment and surgery between April 2020 and December 2022, were included. Patients were divided into 2 groups according to treatment regimen: ICI (sintilimab + nab-paclitaxel + S-1; and non-ICI (nab-paclitaxel + S-1). Imaging and pathological efficacy, intra- and postoperative conditions, molecular subtypes, short-term survival outcomes, and safety were compared between the 2 groups.
RESULTS Imaging evaluation of therapeutic efficacy revealed that the inclusion of ICI yielded a significantly higher complete response rate (13.2% vs 0.0%; P = 0.048), and objective response rate (69.8% vs 31.0%, P = 0.001) compared with non-ICI treatment. Pathological evaluation revealed that the ICI group exhibited a significantly higher pathological complete response rate (13.2% vs 0.0%; P = 0.048) and major pathological response rate (35.8% vs 13.8%; P = 0.041) than those in the non-ICI group. The two-year disease-free survival rate in the ICI group was greater than that in the non-ICI group (83.0% vs 55.2%; P = 0.043). The use of ICI did not increase the incidence of adverse reactions (47.2% vs 41.4%; P = 0.614) or perioperative adverse events (18.9% vs 13.8%; P = 0.761).
CONCLUSION The combination of sintilimab with nab-paclitaxel + S-1 for neoadjuvant treatment of LAGC improved efficacy in patients without increasing adverse drug reactions and perioperative adverse events, suggesting that this treatment regimen is safe and feasible.
Collapse
Affiliation(s)
- Qiu-Xian Chen
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| | - Yong-Bin Zhang
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| | - Wei-Ming Zeng
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| | - Yi-Chen Cai
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, Jiangsu Province, China
| | - Chen-Bin Lv
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| | - Ming-Qiao Lian
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| | - Rong-Jie Huang
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| | - Ming-Jie Lian
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| | - Wei-Long Lian
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| | - Qian-Hui Xu
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| | - Yu-Qin Sun
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| | - Li-Sheng Cai
- Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian Province, China
| |
Collapse
|
4
|
He J, Liu H, Dong Y, Hu C, Liang H, Shou D, Cao D, Du Y, Shao X, Lu Y, Wang H, Tuo C, Deng J, Zhang R, Wang X, Li B, Zhong N, Liu Y, Zhong Y, Gong W. Evolution of HER2 expression after neoadjuvant therapy in locally advanced gastric cancer. iScience 2025; 28:112710. [PMID: 40520113 PMCID: PMC12164010 DOI: 10.1016/j.isci.2025.112710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/10/2025] [Accepted: 05/16/2025] [Indexed: 06/18/2025] Open
Abstract
HER2 is a crucial biomarker in gastric cancer, influencing both treatment strategies and patient prognosis. A multicenter retrospective study was conducted, including 398 patients with locally advanced gastric cancer who received neoadjuvant treatment (NAT) followed by gastrectomy between 2018 and 2023 at three medical centers in China. Alterations in HER2 expression after NAT were detected in more than 40% of patients, with a higher rate of decreased expression (26.0%) compared to increased expression (17.3%). Multivariate analysis indicated that HER2 status at diagnosis significantly influenced HER2 expression alteration. Patients with HER2 IHC 2+ tumors before NAT demonstrated an increased tendency for HER2 expression alterations after NAT. Decreased HER2 expression was associated with improved recurrence-free survival and overall survival. PD-1/PD-L1 inhibitors and trastuzumab both increased pCR rates, but neither significantly impacted the rate of HER2 expression alterations among non-pCR patients. Reassessing HER2 status after NAT is essential for guiding HER2-targeted therapies.
Collapse
Affiliation(s)
- Jun He
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hongming Liu
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yansong Dong
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Chengyu Hu
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Han Liang
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Diwen Shou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hosptital of Chinese Medicine), Hangzhou 310006, China
| | - Dong Cao
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yongxing Du
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xinxin Shao
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yiming Lu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Haikuo Wang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chuanlei Tuo
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jingyu Deng
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Rupeng Zhang
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xuejun Wang
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Bin Li
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ning Zhong
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yong Liu
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yuxin Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Weihua Gong
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
5
|
Wang G, Zheng HL, Zhang LK, Sun YQ, Li BL, Wang YH, Wu J, Wu SC, Zheng HH, Xue Z, Weng CM, Lin JX, Tang JA, Wang JB, Huang CM. Appraisal of Residual Tumor Burden on Survival Prognosis Following Neoadjuvant Chemotherapy in Locally Advanced Gastric Cancer: A Multicenter Cohort Study. Ann Surg Oncol 2025:10.1245/s10434-025-17601-5. [PMID: 40517199 DOI: 10.1245/s10434-025-17601-5] [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: 12/02/2024] [Accepted: 05/14/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND The eighth edition American Joint Committee on Cancer (AJCC) guidelines introduced the postneoadjuvant therapy pathologic tumor, node, metastasis (ypTNM) staging system to assess gastric cancer prognosis postneoadjuvant chemotherapy; however, it overlooks other residual tumor burden aspects beyond tumor infiltration depth and positive lymph nodes. METHODS Data from 537 locally advanced gastric cancer patients (cT2-4NanyM0) treated with neoadjuvant chemotherapy (2010-2021) across six tertiary centers were analyzed. Prognostic factors for overall survival (OS) and recurrence-free survival (RFS) were identified using Cox regression. Reduction of carcinoembryonic antigen (ΔCEA), Response Evaluation Criteria in Solid Tumors (RECIST) criteria, and pathologic nodal stage after neoadjuvant therapy (ypN) were incorporated into a nomogram, validated by receiver operating characteristic (ROC), C-index, calibration plots, and decision curve analyses. A novel prognostic staging system was developed on the basis of the interquartile range (IQR) of the nomogram scores. RESULTS The study included a training cohort of 320 patients and a validation cohort of 217. In the training cohort, multivariate Cox regression identified ypN, RECIST criteria, and ΔCEA as independent prognostic factors for OS and RFS. Nomograms for OS and RFS were developed, showing superior prognostic ability against the American Joint Committee on Cancer (AJCC) eighth edition ypTNM staging for OS (C-index: 0.792 versus 0.689; AIC: 731.268 versus 782.089; BIC: 733.737 versus 784.459) and RFS (C-index: 0.763 versus 0.679; AIC: 868.146 versus 906.772; BIC: 893.301 versus 915.299). The new system improved 3-year OS (89.7%, 64.2%, 60.2%, and 23.0%, p < 0.001) and RFS (84.3%, 79.0%, 65.1%, and 30.0%, p < 0.001) stratification over AJCC stages. Similar results were observed in the validation cohort. CONCLUSIONS The novel staging system, based on residual tumor burden, significantly outperforms ypTNM staging, enhancing prognostic prediction accuracy in postneoadjuvant chemotherapy patients with gastric cancer.
Collapse
Affiliation(s)
- Gang Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Hua-Long 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
| | - Ling-Kang 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-Qin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 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
- Department of General Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Yong-Hong Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Gastrointestinal Surgery, Leshan People's Hospital, Leshan, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Shi-Chao Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Gastrointestinal Surgery, The First Hospital of Putian, Putian, China
| | - Hong-Hong 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
| | - 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
| | - 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
| | - Jian-Xian 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
| | - Ji-Ao Tang
- Department of General Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Jia-Bin 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.
| | - 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.
| |
Collapse
|
6
|
Masuda Y, Fong KL, Yeo D, Yeo C, Chue KM, Araba SB, Lim CW, Yeung B, Lee J, Lin J, Chia C, Ng M, Ng K, Samol J, Chia D, Teh JL, Sundar R, Yong WP, Tan HL, Muro K, Lordick F, Wainburg Z, Tan BC, Kim G, Suda K, Law S, Sano T, Gurunathan R, Chiu P, Woo E, Duong C, Yang HK, Long VD, Kim HH, Mahendren HA, Lee HJ, Samarasam I, Gotoda T, Liew R, Shabbir A, Aung MO, Terashima M, Cheong E, So J, Tan J. Asia Pacific Gastroesophageal Cancer Congress (APGCC) 2024 consensus statement on stage 2 and 3 locally advanced gastric and Siewert 3 junctional adenocarcinoma. J Gastroenterol 2025:10.1007/s00535-025-02266-4. [PMID: 40514519 DOI: 10.1007/s00535-025-02266-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND While the development in multimodal therapies has helped improve treatment outcomes for patients with locally advanced gastric adenocarcinoma (LAGC), there still exist disparities in opinion with an optimal treatment plan. This consensus hopes to provide clinicians with structured guidelines to aid in the decision-making for treatment options for LAGC. METHODS The consensus statement was initiated by establishing a taskforce in collaboration with the Asia Pacific Gastroesophageal Cancer Congress (APGCC) and a multidisciplinary expert panel was selected. Clinical questions on LAGC where perceived variance in practice or opinion may exist were formulated. Studies involving patients with Stage 2 or 3 gastric or Siewert 3 junctional cancers with treatment arms of perioperative chemotherapy, neoadjuvant chemotherapy, adjuvant chemotherapy, immunotherapy and surgery were included. A total of two rounds of voting were performed. Consensus was determined to be reached when a single answer or a combination of either "strongly agree/agree" or "strongly disagree/disagree" responses exceeded 75%. RESULTS A total of thirteen clinical questions were developed. They were identified through five main categories: Distal LAGC, Proximal LAGC, Deficient mismatch repair tumors, Chemotherapy and Immunotherapy, and Elderly/Unfit patients. After two rounds of voting by our multidisciplinary expert panel, eleven out of a total thirteen clinical questions had reached consensus. No consensus was reached for two clinical questions. CONCLUSION The APGCC consensus statement aims to guide clinicians in the treatment options for LAGC and Siewert 3 junctional cancer and has clarified some of the roles of perioperative chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yoshio Masuda
- Department of Upper Gastrointestinal & Bariatric Surgery, Division of Surgery, Singapore General Hospital, Singapore, Singapore
- Ministry of Health Holdings, Singapore, Singapore
| | - Kang Ler Fong
- Department of Upper Gastrointestinal & Bariatric Surgery, Division of Surgery, Singapore General Hospital, Singapore, Singapore
| | - Danson Yeo
- Upper Gastrointestinal & Bariatric Surgery Service, Department of General Surgery, Tan Tock Seng Hospital, Singapore, Singapore
| | - Charleen Yeo
- Upper Gastrointestinal & Bariatric Surgery Service, Department of General Surgery, Tan Tock Seng Hospital, Singapore, Singapore
| | - Koy Min Chue
- Upper Gastrointestinal & Bariatric Surgery Service, Department of General Surgery, Sengkang General Hospital, Singapore, Singapore
| | - Said Bani Araba
- Division of General Surgery (Upper Gastrointestinal Surgery), Department of Surgery, National University Hospital, Singapore, Singapore
| | - Chiew Woon Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Baldwin Yeung
- Upper Gastrointestinal & Bariatric Surgery Service, Department of General Surgery, Sengkang General Hospital, Singapore, Singapore
| | - June Lee
- Upper Gastrointestinal & Bariatric Surgery Service, Department of General Surgery, Changi General Hospital, Singapore, Singapore
| | - Jinlin Lin
- Upper Gastrointestinal & Bariatric Surgery Service, Department of General Surgery, Changi General Hospital, Singapore, Singapore
| | - Claramae Chia
- Division of Surgery and Surgical Oncology, Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), National Cancer Centre Singapore and Singapore General Hospital, Singapore, Singapore
- SingHealth Duke-NUS Surgery & Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Matthew Ng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Kennedy Ng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Jens Samol
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- John Hopkins University School of Medicine, Baltimore, USA
| | - Daryl Chia
- Division of General Surgery (Upper Gastrointestinal Surgery), Department of Surgery, National University Hospital, Singapore, Singapore
| | - Jun Liang Teh
- Upper Gastrointestinal & Bariatric Surgery Service, Department of General Surgery, Ng Teng Fong Hospital, Singapore, Singapore
| | - Raghav Sundar
- Center for Gastrointestinal Cancers, Yale Cancer Center, New Haven, United States of America
| | - Wei-Peng Yong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Hon Lyn Tan
- OncoCare Cancer Centre, Singapore, Singapore
| | - Kei Muro
- Department of Clinical Oncology and Outpatient Treatment Centre, Alchi Cancer Centre Hospital, Nagoya, Japan
| | - Florian Lordick
- Department of Medical Oncology, University Cancer Centre, Leipzig, Germany
| | - Zev Wainburg
- Department of Medicine, University of California Los Angeles Medical Centre, Los Angeles, CA, USA
| | - Bo Chuan Tan
- Department of Upper Gastrointestinal Surgery, Clinic for Digestive Surgery, Singapore, Singapore
| | - Guowei Kim
- Department of Upper Gastrointestinal Surgery, Crest Surgical Practice, Singapore, Singapore
| | - Koichi Suda
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, Japan
| | - Simon Law
- Division of Esophageal and Upper Gastrointestinal Surgery, Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Takeshi Sano
- Gastroenterological Center, The Cancer Institute Hospital, Tokyo, Japan
| | - Ramesh Gurunathan
- Department of Surgery, Sunway Medical Centre, Subang, Selangor, Malaysia
| | - Philip Chiu
- Division of Upper GI and Metabolic Surgery, Department of Surgery, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong S.A.R, China
| | - Emile Woo
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Cuong Duong
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, East Melbourne, Vic., Australia
| | | | - Vo Duy Long
- Gastro-Intestinal Surgery Department, University Medical Center, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Hyung Ho Kim
- Comprehensive Cancer Center, Seoul National University Bundang Hospital, Seongnam, South Korea
| | | | - Hyuk Joon Lee
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Inian Samarasam
- Upper Gastrointestinal Surgery Unit, Department of General Surgery Unit 3, Christian Medical College, Vellore, India
| | - Takuji Gotoda
- Department of Gastroenterology, Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Reis Liew
- Division of General Surgery (Upper Gastrointestinal Surgery), Department of Surgery, National University Hospital, Singapore, Singapore
| | - Asim Shabbir
- Division of General Surgery (Upper Gastrointestinal Surgery), Department of Surgery, National University Hospital, Singapore, Singapore
| | - Myint Oo Aung
- Upper Gastrointestinal & Bariatric Surgery Service, Department of General Surgery, Tan Tock Seng Hospital, Singapore, Singapore
| | - Masanori Terashima
- Division of Gastric Surgery, Shizuoka Cancer Center, 1007 Monagakubo, Nagaizumi-Cho, Sunto-Gun, Nagaizumi, Shizuoka, Japan
| | - Edward Cheong
- Department of Upper Gastrointestinal Surgery, PanAsia Surgery, Singapore, Singapore
| | - Jimmy So
- Division of General Surgery (Upper Gastrointestinal Surgery), Department of Surgery, National University Hospital, Singapore, Singapore.
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 5 Lower Kent Ridge Road, Singapore, 119074, Singapore.
- Division of Surgical Oncology, National University Cancer Institute of Singapore (NCIS), 5 Lower Kent Ridge Road, Singapore, 119074, Singapore.
| | - Jeremy Tan
- Department of Upper Gastrointestinal & Bariatric Surgery, Division of Surgery, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
7
|
Yao K, Okuno K, Watanabe S, Shigeno T, Ogo T, Fujiwara H, Tanioka T, Kawada K, Tokunaga M, Ban D, Kinugasa Y. A Novel Transcriptomic Signature for Prediction of Response to Adjuvant Chemotherapy in Patients With Stages II and III Gastric Cancer. Ann Surg Oncol 2025:10.1245/s10434-025-17487-3. [PMID: 40415152 DOI: 10.1245/s10434-025-17487-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/28/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Predicting patients who will benefit from postoperative adjuvant chemotherapy is crucial for precision medicine. Therefore, this study comprehensively analyzed messenger RNA (mRNA) expression profiles to identify novel biomarkers and developed a prediction signature for postoperative adjuvant chemotherapy in patients with gastric cancer (GC). METHODS Biomarkers were discovered by analyzing two publicly available genome-wide datasets from 343 patients with pathologic stages (pStages) II and III GC. A novel prediction signature was developed based on a quantitative reverse transcription polymerase chain reaction (qRT-PCR) assay using 137 pStages II and III GC frozen tissue specimens. RESULTS Nine novel mRNAs were identified as candidate biomarkers in biomarker discovery, and a Gene Expression-based ADJuvant chemotherapy Response prediction for stages II and III GC (GEx-ADJ-Res) signature was developed using these candidate biomarkers and key clinicopathologic features by qRT-PCR assay. The GEx-ADJ-Res signature robustly predicted postoperative recurrence in clinical tissue samples (area under the curve [AUC], 0.84). The signature demonstrated sufficient potential for predicting response to postoperative adjuvant chemotherapy (AUC, 0.82) and was shown to be an independent predictor of postoperative recurrence and survival in multivariate analysis. Finally, the GEx-ADJ-Res signature was successfully validated using independent multi-institutional datasets (AUC, 0.91, 0.85, and 0.78, respectively). CONCLUSIONS We identified the novel mRNA biomarkers and developed a novel signature that allowed robust prediction of response to postoperative adjuvant chemotherapy in patients with GC. This signature could become a precision medicine tool in GC treatment.
Collapse
Affiliation(s)
- Kenta Yao
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Keisuke Okuno
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan.
| | - Shuichi Watanabe
- Department of Hepatobiliary and Pancreatic Surgery, Institute of Science Tokyo, Tokyo, Japan
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Shigeno
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Taichi Ogo
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Hisashi Fujiwara
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Toshiro Tanioka
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Kenro Kawada
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Masanori Tokunaga
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepatobiliary and Pancreatic Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Yusuke Kinugasa
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Shen ZZ, Li EZ, Zhang RL, Cao MX, Zhang YQ, Yang Q, Hu C, Pan SW, Xu ZY, Ye ZS, He JY. Adjuvant chemotherapy may have no significant survival benefit in older patients with stage II/III gastric cancer: a multicenter retrospective study. J Cancer Res Clin Oncol 2025; 151:175. [PMID: 40419812 PMCID: PMC12106140 DOI: 10.1007/s00432-025-06230-w] [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: 04/01/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025]
Abstract
AIM Postoperative adjuvant chemotherapy is known to enhance cure rates and is thus recommended for stages pII to pIII. However, specific guidelines for such treatment in elderly gastric cancer (GC) patients are currently lacking. This study examines the impact of adjuvant chemotherapy on the postoperative survival of these patients. METHODS We reviewed a total of 7749 patients with GC who underwent radical gastrectomy at Zhejiang Cancer Hospital and Fujian Cancer Hospital from January 2007 to December 2019. We conducted univariate and multivariate Cox regression analyses to investigate the impact of clinicopathological factors on overall survival (OS) and cancer-specific survival (CSS) in these patients. Additionally, we created a meta-analysis forest plot and employed propensity score matching (PSM) to mitigate confounding bias. RESULTS Age and adjuvant chemotherapy were independent risk factors for OS and CSS. Stratified analysis based on chemotherapy use revealed a statistically significant difference in OS and CSS between younger patients who did and did not receive adjuvant chemotherapy. In contrast, no significant differences in OS and CSS were observed between older patients with or without adjuvant chemotherapy. These findings remained consistent after propensity score matching (PSM). CONCLUSIONS Age and adjuvant chemotherapy are independent risk factors for OS and CSS in patients with stage II/III GC; for patients with stage II/III gastric cancer aged ≥ 75 years, shared decision-making should be made taking into account functional status and comorbidities, rather than conventional adjuvant chemotherapy.
Collapse
Affiliation(s)
- Zheng-Zheng Shen
- Department of Radiation Physics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - En-Ze Li
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang Province, China
| | - Ruo-Lan Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Meng-Xuan Cao
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yan-Qiang Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Qing Yang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang Province, China
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Si-Wei Pan
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Zhi-Yuan Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Zai-Sheng Ye
- Department of Gastric Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, 350014, Fujian Province, China.
| | - Jing-Yang He
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang Province, China.
| |
Collapse
|
9
|
Ling T, Zuo Z, Huang M, Ma J, Wu L. Stacking classifiers based on integrated machine learning model: fusion of CT radiomics and clinical biomarkers to predict lymph node metastasis in locally advanced gastric cancer patients after neoadjuvant chemotherapy. BMC Cancer 2025; 25:834. [PMID: 40329193 PMCID: PMC12057267 DOI: 10.1186/s12885-025-14259-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND The early prediction of lymph node positivity (LN+) after neoadjuvant chemotherapy (NAC) is crucial for optimizing individualized treatment strategies. This study aimed to integrate radiomic features and clinical biomarkers through machine learning (ML) approaches to enhance prediction accuracy by focusing on patients with locally advanced gastric cancer (LAGC). METHODS We retrospectively enrolled 277 patients with LAGC and randomly divided them into training (n = 193) and validation (n = 84) sets at a 7:3 ratio. In total, 1,130 radiomics features were extracted from pre-treatment portal venous phase computed tomography scans. These features were linearly combined to develop a radiomics score (rad score) through feature engineering. Then, using the rad score and clinical biomarkers as input features, we applied simple statistical strategies (relying on a single ML model) and integrated statistical strategies (including classification model integration techniques, such as hard voting, soft voting, and stacking) to predict LN+ post-NAC. The diagnostic performance of the model was assessed using receiver operating characteristic curves with corresponding areas under the curve (AUC). RESULTS Of all ML models, the stacking classifier, an integrated statistical strategy, exhibited the best performance, achieving an AUC of 0.859 for predicting LN+ in patients with LAGC. This predictive model was transformed into a publicly available online risk calculator. CONCLUSIONS We developed a stacking classifier that integrates radiomics and clinical biomarkers to predict LN+ in patients with LAGC undergoing surgical resection, providing personalized treatment insights.
Collapse
Affiliation(s)
- Tong Ling
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Zhichao Zuo
- School of Mathematics and Computational Science, Xiangtan University, Xiangtan, Hunan province, China
| | - Mingwei Huang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Jie Ma
- Department of Medical Imaging, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Liucheng Wu
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.
| |
Collapse
|
10
|
Cheng H, Chen J, Li Y, Li Y, Tse C, Shen B, Li S, Chen Q, Tang L, Mai H, Liu L. Determining the optimal duration of oral adjuvant chemotherapy in locoregionally advanced nasopharyngeal carcinoma. Br J Cancer 2025:10.1038/s41416-025-03033-1. [PMID: 40328916 DOI: 10.1038/s41416-025-03033-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Concurrent chemoradiotherapy (CCRT) followed by adjuvant chemotherapy (AC) is the standard treatment for locoregionally advanced nasopharyngeal carcinoma (LA-NPC). However, the optimal duration of oral AC remains poorly defined. METHODS This study examined newly diagnosed patients between April 2017 and December 2020. The primary endpoint was overall survival (OS). Restricted cubic splines (RCS) and Kaplan-Meier method were used to evaluate the relationship between AC maintenance and survival. Inverse probability of treatment weighting (IPTW) was used to control for confounding factors. RESULTS The RCS demonstrated an L-shaped association between oral AC maintenance and OS. The risk of mortality was relatively flat after 12 months. Patients with oral AC duration >186 days (defined by RCS) had a significantly better OS (HR 0.23 [95% CI 0.10-0.55], log-rank p < 0.001), with a higher 3-year OS rate (98.7% [95% CI 96.8-100.0] vs 88.3% [95% CI 82.5-94.5]). For patients with pretreatment Epstein-Barr virus (EBV) DNA level >4000 copies/mL, mortality risk decreased to 1 at 194 days of AC duration. CONCLUSIONS The optimal duration of oral AC after CCRT was >186 days (6 months) for LA-NPC. And the maintenance beyond 12 months may not bring additional benefits.
Collapse
Affiliation(s)
- Hui Cheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Jie Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yifu Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yuchen Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Chunfung Tse
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Bowen Shen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Shibing Li
- Department of Clinical Laboratory, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Qiuyan Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Linquan Tang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Haiqiang Mai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Liting Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| |
Collapse
|
11
|
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
|
12
|
Zeng S, Yin S, Lian S, Luo M, Feng L, Liao Y, Huang Z, Zheng Y, Xie C, Zhuo S. A Clinical-Radiomic Combined Model based on Dual-Layer Spectral CT for Predicting Pathological T4 in Gastric Cancer. Acad Radiol 2025:S1076-6332(25)00383-6. [PMID: 40328540 DOI: 10.1016/j.acra.2025.04.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025]
Abstract
RATIONALE AND OBJECTIVES This study aimed to develop and validate a dual-layer spectral CT based clinical-radiomic model for pre-treatment prediction of pathological T4 (pT4) in gastric cancer (GC) patients. MATERIALS AND METHODS This retrospective study included 148 surgically confirmed GC patients who underwent dual-layer spectral CT scanning before surgery and were divided into a training (n=104) and test (n=44) cohorts. Subjective assessments were performed based on conventional 120-kV CT images by two readers. Clinical models were developed using patient demographics, serum tumor markers, and image features from CT scans. Radiomics model included features extracted from conventional 120-kV CT and dual-layer CT-derived spectral base image (SBI), such as virtual monoenergetic images (40 keV, 70 keV, 100 keV), iodine density (ID), effective atomic number (Zeff), and electron density (ED) images for both the arterial phase (AP) and portal venous phase (PVP). A clinical-radiomic combined model was developed and visualized using a nomogram. RESULTS Tumor thickness on CT and serum level of CA19-9 levels were identified as independent predictors. The clinical-radiomic combined model demonstrated superior performance compared to subjective image interpretation and other models, with an AUC of 0.906 (95% CI, 0.848-0.963) in the training cohort and 0.873 in the test cohort. The nomogram was significantly associated with pT4 status, supporting its potential utility in clinical prediction. CONCLUSION The integration of clinical characteristics with radiomic features from conventional CT and dual-layer CT-derived SBI achieved a high diagnostic accuracy for predicting pT4 in GC patients. This combined approach could assist in treatment planning and patient management in GC.
Collapse
Affiliation(s)
- Sihui Zeng
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Shaohan Yin
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Shanshan Lian
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Ma Luo
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Lili Feng
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Yuting Liao
- Philips Healthcare, Guangzhou 510000, PR China (Y.L., Z.H.)
| | - Zhijie Huang
- Philips Healthcare, Guangzhou 510000, PR China (Y.L., Z.H.)
| | - Yuquan Zheng
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Chuanmiao Xie
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.).
| | - Shuiqing Zhuo
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.).
| |
Collapse
|
13
|
Zhu W, Zhang J, Pan S, Zhang R, Zhang Y, Yang Q, Hu C, Xu Z. Identifying beneficial gastric cancer patient on prognosis after treating with perioperative or postoperative-only chemotherapy: a single-center real-world study. World J Surg Oncol 2025; 23:177. [PMID: 40320534 PMCID: PMC12051334 DOI: 10.1186/s12957-025-03827-w] [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: 02/06/2025] [Accepted: 04/25/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND This study evaluates the impact of perioperative S-1 and oxaliplatin (SOX) versus postoperative SOX or capecitabine and oxaliplatin (XELOX) on patient prognosis to identify suitable candidates for each therapy. METHOD A retrospective real-world cohort study was conducted using data from Zhejiang Cancer Hospital on gastric cancer patients treated between 2010 and 2019. Patients were divided into perioperative SOX and postoperative SOX or XELOX groups. Propensity score matching (PSM) was used to control for selection bias. Overall survival (OS) was the primary outcome, analyzed using the Kaplan-Meier method and Cox regression. RESULT A total of 816 patients were included: 293 in the perioperative SOX group and 523 in the postoperative chemotherapy group (408 SOX and 115 XELOX). In the perioperative SOX group, the tumor regression grade (TRG) 2-3 subgroup demonstrated a significantly worse overall survival (OS) compared to the postoperative XELOX group (95% CI = 1.064-3.444, P = 0.027). Subgroup analysis revealed that older patients (95% CI = 0.210-0.950, P = 0.036), and those at the cT3 (95% CI = 0.05-1.19, P = 0.008) stage experienced greater benefits from postoperative chemotherapy. When comparing the benefited populations, it was found that patients with CA125 positivity had an advantage trend with adjuvant chemotherapy compared to perioperative SOX chemotherapy. CONCLUSION Real-world data suggest that perioperative SOX chemotherapy does not benefit all patients with advanced gastric cancer. Patients with TRG 2-3, older age, or cT3 stage may achieve better outcomes with postoperative chemotherapy. Additionally, an exploratory analysis indicated that CA125 positivity may be associated with improved survival following adjuvant treatment.
Collapse
Affiliation(s)
- Weiwei Zhu
- Second clinical medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Jiaqing Zhang
- Second clinical medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Siwei Pan
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Ruolan Zhang
- Second clinical medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yanqiang Zhang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Qing Yang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Can Hu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China.
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China.
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| | - Zhiyuan Xu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China.
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China.
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| |
Collapse
|
14
|
Anyaduba UL, Orababa OQ, Faye Z, Rashid N, Shafrin J, Reardon G. Meta-Analysis of Recurrence-Free Survival or Disease-Free Survival as a Potential Surrogate Endpoint for Overall Survival in Esophageal Cancer Trials. Cancer Rep (Hoboken) 2025; 8:e70195. [PMID: 40387359 PMCID: PMC12087002 DOI: 10.1002/cnr2.70195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/20/2025] [Accepted: 03/21/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Cancer trials increasingly use surrogate endpoints, but it is unclear how well recurrence-free survival (RFS) or disease-free survival (DFS) specifically predict overall survival (OS) in resectable esophageal cancer (EC). METHODS A systematic literature review identified trials with RFS/DFS and OS endpoints. A meta-analysis assessed RFS/DFS as surrogates for OS, estimating pooled hazard ratios (HRs) from trial HRs. Forest plots and heterogeneity tests showed effect sizes and pooled estimates. Unweighted linear regression and weighted sensitivity analysis estimated the correlation between OS and RFS/DFS, producing a regression plot. RESULTS Of 975 articles identified, 11 met the criteria. The pooled HR for OS and RFS/DFS was 0.90 and 0.87, respectively. The primary analysis showed a strong Pearson correlation between RFS/DFS and OS (ρ = 0.89, p < 0.001). CONCLUSION Subject to known methodological limits, RFS/DFS was demonstrated to be a potentially suitable surrogate endpoint for OS in resectable EC.
Collapse
Affiliation(s)
| | | | - Zion Faye
- Henry E. Riggs School of Applied Life SciencesKeck Graduate InstituteClaremontCaliforniaUSA
| | - Nazia Rashid
- School of PharmacyKeck Graduate InstituteClaremontCaliforniaUSA
| | - Jason Shafrin
- Center for Healthcare Economics and PolicyFTI ConsultingLos AngelesCaliforniaUSA
| | - Gregory Reardon
- School of PharmacyKeck Graduate InstituteClaremontCaliforniaUSA
| |
Collapse
|
15
|
Liu Q, Meng C, Cao S, Liu X, Tian Y, Li Z, Zhong H, Sun Y, Yu J, Zhou Y. Comparison of short- and long-term outcomes of robotic versus laparoscopic gastrectomy for locally advanced gastric cancer after neoadjuvant therapy: a high-volume center retrospective study with propensity score matching. Surg Endosc 2025; 39:2814-2827. [PMID: 40064692 DOI: 10.1007/s00464-025-11626-3] [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: 10/28/2024] [Accepted: 02/18/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Although neoadjuvant therapy (NAT) for advanced gastric cancer (AGC) can benefit patient survival, few studies have compared the short- and long-term outcomes of robotic and laparoscopic gastrectomy for AGC after NAT. METHODS The clinical data of 321 AGC patients who received NATs and who underwent robotic gastrectomy (RG, n = 109) or laparoscopic gastrectomy (LG, n = 212) between May 2017 and September 2022 were collected and analyzed retrospectively at our center. After propensity score matching (PSM) for 1:1 matching to eliminate bias, both groups had 106 cases. Short-term clinical outcomes and long-term survival-related indicators were compared between the two groups of patients. RESULTS A total of 212 patients were included in the groups after matching. There were fewer overall complications (13.2% vs. 28.3%, P = 0.007) and surgical complications (8.5% vs. 17.9%, P = 0.043) in the RG group than in the LG group. Compared with the LG group, the RG group had more harvested overall lymph nodes (35.25 ± 4.99 vs. 31.45 ± 6.31, P < 0.001) and more suprapancreatic lymph nodes (13.12 ± 4.38 vs. 10.05 ± 4.13, P < 0.001). Patients in the RG group had significantly shorter surgery times (217.62 ± 47.49 vs. 267.25 ± 70.68, P < 0.001) and less blood loss (46.51 ± 27.02 vs. 70.75 ± 37.25, P < 0.001) than patients in the LG group. The RG group had significantly faster bowel function recovery, earlier liquid diet, and shorter hospital stay. Compared with LG, RG had a better 3-year RFS (75.5% vs. 62.3%, P = 0.017). CONCLUSION Compared with laparoscopic surgery, robotic surgery significantly increased the number of lymph node dissected, reduced intraoperative blood loss, and postoperative surgical complications rate. Although RG did not statistically improve 3-year overall survival, there was a significant improvement in RFS and could be an alternative surgical method for GC patients after NAC.
Collapse
Affiliation(s)
- Qi Liu
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Cheng Meng
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Shougen Cao
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaodong Liu
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Yulong Tian
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Zequn Li
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Hao Zhong
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Yuqi Sun
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Junjian Yu
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Yanbing Zhou
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China.
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China.
| |
Collapse
|
16
|
Sundar R, Nakayama I, Markar SR, Shitara K, van Laarhoven HWM, Janjigian YY, Smyth EC. Gastric cancer. Lancet 2025:S0140-6736(25)00052-2. [PMID: 40319897 DOI: 10.1016/s0140-6736(25)00052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/13/2024] [Accepted: 01/09/2025] [Indexed: 05/07/2025]
Abstract
Gastric cancer remains a major health challenge worldwide, with nearly 1 million new cases annually contributing to more than 650 000 deaths. Epidemiologically, gastric cancer shows substantial geographical variation in incidence, with higher rates in Asia, South America, and eastern Europe, and a rapid increase in early-onset cases among people younger than 50 years. Key risk factors for gastric cancer include Helicobacter pylori infection, diet, obesity, smoking, and genetic predisposition. Early detection through comprehensive diagnostic procedures is crucial for optimising treatment outcomes. Standard treatment approaches for locally advanced gastric cancer include surgical resection, particularly D2 lymphadenectomy, complemented by chemotherapy and radiotherapy. There is increasing implementation of minimally invasive surgical techniques for operable disease and integration of immune checkpoint inhibitors and targeted therapies for advanced stages. Emerging therapies, such as novel targeted treatments and next-generation immunotherapies, show promise in improving survival and quality of life. Future directions in the management of gastric cancer focus on precision medicine, continued advancement in immunotherapy, novel early detection methods, and a multidisciplinary approach to care. These strategies aim to enhance the overall effectiveness of treatment and prognosis worldwide.
Collapse
Affiliation(s)
- Raghav Sundar
- Department of Medicine, Section of Medical Oncology, Yale School of Medicine, New Haven, CT, USA; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Izuma Nakayama
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Sheraz R Markar
- Surgical Intervention Trials Unit, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands; Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Yelena Y Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Elizabeth C Smyth
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
17
|
Shang-Guan ZX, Zhong Q, Zhang ZQ, Liu ZY, Sun YQ, Ma YB, Ding FH, Wu SC, Zhu JY, Wang YH, Zhao W, Yu JH, Li BL, Wu J, Ye W, Li P, Xie JW, Zheng CH, Chen QY, Huang CM. Surgical and oncological outcomes of laparoscopic versus open gastrectomy after neoadjuvant chemotherapy in patients with locally advanced gastric cancer: A multicenter analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110060. [PMID: 40398315 DOI: 10.1016/j.ejso.2025.110060] [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: 01/31/2025] [Revised: 03/31/2025] [Accepted: 04/13/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Evidence based on large-scale samples comparing the efficacy of laparoscopic gastrectomy (LG) and open gastrectomy (OG) in patients with locally advanced gastric cancer (LAGC) after neoadjuvant chemotherapy (NACT) remains limited. This multicenter study aimed to evaluate the short -term and oncological outcomes of LG and OG after NACT. METHODS Data from a multicenter database of LAGC patients undergoing radical gastrectomy after NACT across 12 centers in China. Propensity score matching (PSM 3:1) was used to balance baseline characteristics. Short-term outcomes, 5-year overall survival (OS), disease-free survival (DFS), and recurrence patterns were compared. RESULTS In total, 962 patients fulfilled the inclusion criteria, of whom 753 underwent LG and 209 underwent OG. After PSM, 627 LG and 209 OG patients were analyzed. Both groups had comparable clinical and pathological characteristics (SMD≤0.100). Intraoperative blood loss was lower in the LG group, with earlier ambulation and diet initiation than that in the OG group (P < 0.05). The number of lymph nodes retrieved was higher in LG group (P < 0.001). Although no significant differences were shown in 5-year OS (LG vs. OG: 51.7vs.43.4 %) and 5-year DFS (LG vs. OG, 48.8 vs. 41.4 %; P > 0.05), landmark analysis revealed improved OS (77.6vs. 65.9 %; P = 0.024) and DFS (84.1vs.71.4 %; P = 0.031) after a landmark time of 28 months in the LG group. Most recurrences occurred within 3 years postoperatively, with similar recurrence patterns in both groups (P > 0.05). CONCLUSIONS In LAGC patients after NACT, LG yielded faster recovery while demonstrating comparable surgical efficacy and oncological outcomes to OG, with potential long-term survival benefits.
Collapse
Affiliation(s)
- Zhi-Xin Shang-Guan
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yu-Qin Sun
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Fang-Hui Ding
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Shi-Chao Wu
- Gastrointestinal Surgery Unit 2, Teaching Hospital of Putian First Hospital of Fujian Medical University, Putian, China
| | - Ji-Yun Zhu
- Department of Hepato-pancreato-biliary Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yong-Hong Wang
- Department of Gastrointestinal Surgery, The People's Hospital of Leshan, Leshan, China
| | - Wei Zhao
- Department of General Surgery, The General Hospital of Ningxia Medical University, Ningxia, China
| | - Jun-Hua Yu
- Department of General Surgery, The Quzhou Affiliated Hospital of Wenzhou University, Quzhou, China
| | - Bao-Long Li
- Department of General Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Ju Wu
- Department of General Surgery, The Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Wen Ye
- Department of Gastrointestinal Surgery, The Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
18
|
Liu G, Cao S, Liu X, Tian Y, Yu W, Chai J, Li L, Wang X, Chu X, Duan Q, Qu J, Wang H, Zhang H, Wang X, Hui X, Yang D, Zhou S, Ding Y, Wang H, Zhou F, Hu B, Guo P, Jiang L, Zhang G, Pan Q, Zhou X, Zhou Y. Effect of perioperative probiotic supplements on the short-term clinical outcomes of patients undergoing laparoscopic or robotic radical gastrectomy after neoadjuvant chemotherapy: Study protocol for a multicenter randomized controlled trial (GISSG2023 - 01 Study). BMC Cancer 2025; 25:776. [PMID: 40281451 PMCID: PMC12023430 DOI: 10.1186/s12885-025-14115-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Gastric cancer is a common malignant tumor, and radical gastrectomy can markedly improve the prognosis of gastric cancer patients. However, some patients are diagnosed with advanced gastric cancer before receiving any antitumor therapy and need to receive neoadjuvant chemotherapy (NACT). Previous studies have shown that NACT may cause gut barrier dysfunction and intestinal dysbacteriosis which may further lead to infections. Probiotics have the potential to reduce postoperative infections and improve short-term outcomes after abdominal surgery; however, no large-sample, multicenter, randomized clinical trials have been conducted to explore the effectiveness of probiotics in gastric cancer patients receiving NACT. So we proposed a hypothesis that probiotics can improve short-term outcomes after minimally invasive radical gastrectomy in gastric cancer patients receiving NACT and designed this multicenter randomized controlled trial with the objective to verify this hypothesis. METHODS/DESIGN The GISSG 2023-01 study will be a prospective, open-label, multicenter RCT to verify whether perioperatively probiotic supplementation (begin from the end of the last cycle of NACT to postoperative day 7 or the discharge day) can reduce postoperative infections and improve recovery of gastrointestinal function and other short-term outcomes after minimally invasive radical gastrectomy in gastric cancer patients receiving NACT. A total of 318 patients who meet the inclusion criteria will be enrolled in this study and randomly divided into two groups in a 1:1 ratio: the probiotic group (n = 159) and the control group (n = 159). The participants in the probiotic group will receive perioperative probiotic supplementation, and those in the control group will receive blank control management. The other perioperative management protocols will be the same between the two groups. The primary outcome is postoperative infection compared between the two groups, and the secondary outcomes are postoperative recovery of gastrointestinal function, quality of life, laboratory parameters of systemic inflammation and other short-term outcomes. DISCUSSION The results of this RCT should clarify whether perioperative probiotic supplementation would reduce postoperative infection, promote recovery of gastrointestinal function, reduce laboratory parameters of systemic inflammation and improve symptoms and quality of life after minimally invasive radical gastrectomy in gastric cancer patients receiving NACT. It is hoped that our data will provide evidence that probiotic supplementation improves short-term outcomes in gastric cancer patients receiving NACT. TRIAL REGISTRATION This trial has been registered on https://clinicaltrials.gov/(NCT05901779 ).
Collapse
Affiliation(s)
- Gan Liu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, China
| | - Shougen Cao
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, China
| | - Xiaodong Liu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, China
| | - Yulong Tian
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, China
| | - Wenbin Yu
- Qilu Hospital of Shandong University, Jinan, China
| | - Jie Chai
- Shandong Cancer Hospital, Jinan, China
| | - Leping Li
- Shandong Provincial Hospital, Jinan, China
| | - Xixun Wang
- Yantai Yuhuangding Hospital, Yantai, China
| | - Xianqun Chu
- Shandong Jining No.1 People's Hospital, Jining, China
| | - Quanhong Duan
- Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jianjun Qu
- Weifang People's Hospital, Weifang, China
| | - Hao Wang
- Dongying People's Hospital, Dongying, China
| | | | | | | | - Daogui Yang
- Liaocheng People's Hospital, Liaocheng, China
| | | | - Yinlu Ding
- The Second Hospital of Shandong University, Jinan, China
| | - Hongbo Wang
- The People's Hospital of Jimo, Qingdao, China
| | | | - Baoguang Hu
- Binzhou Medical University Hospital, Yantai, China
| | | | | | | | - Qiang Pan
- Rushan People's Hospital, Weihai, China
| | - Xiaobin Zhou
- Department of Epidemiology and Health Statistics, School of Public Health of Qingdao University, Qingdao, China
| | - Yanbing Zhou
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, China.
| |
Collapse
|
19
|
Chen J, Hu K, Lu J, Pan H, Huang Y, Yu Z, Gao T, Fan Y, Li C, Liu F. Optimizing surgical timing following neoadjuvant therapy for gastric Cancer: Insights from a multicenter retrospective analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110063. [PMID: 40288218 DOI: 10.1016/j.ejso.2025.110063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/16/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Neoadjuvant therapy (NAT) is a cornerstone in the treatment of locally advanced gastric cancer, improving surgical outcomes and survival. However, the optimal timing for surgery following NAT remains controversial. This study evaluates the impact of the interval between NAT and surgery on overall survival (OS) and explores associated clinicopathological factors. METHODS A retrospective analysis of 893 patients undergoing NAT and curative surgery for gastric adenocarcinoma across three centers in China was conducted. Surgical intervals were categorized (∼28 days, 29-42 days, >42 days). Survival analyses employed restricted cubic spline (RCS) models, Kaplan-Meier methods, and Cox proportional hazards regression. RESULTS Patients operated on within 28 days post-NAT had the most favorable OS, while intervals longer than 28 days were independently associated with worse outcomes. RCS analysis revealed increased risks for intervals longer than 28 days. Prolonged intervals showed declining effectiveness in tumor regression. Stratified analyses indicated that patients with poor NAT response (TRG 3) particularly benefited from surgery within 4 weeks, while delays were detrimental. CONCLUSIONS Timely surgery, especially within 4 weeks post-NAT, optimizes survival outcomes, particularly in patients with limited NAT response. This study underscores the need for individualized surgical timing and calls for prospective multicenter validation.
Collapse
Affiliation(s)
- Jie Chen
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Keshu Hu
- Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jun Lu
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hongda Pan
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yakai Huang
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhenjia Yu
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tianyu Gao
- Department of General Surgery, The Second Hospital of Lanzhou University, 730030, China
| | - Yong Fan
- Department of General Surgery, The Second Hospital of Lanzhou University, 730030, China.
| | - Chen Li
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Fenglin Liu
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
20
|
Chen Y, Jia K, Xie Y, Yuan J, Liu D, Jiang L, Peng H, Zhong J, Li J, Zhang X, Shen L. The current landscape of gastric cancer and gastroesophageal junction cancer diagnosis and treatment in China: a comprehensive nationwide cohort analysis. J Hematol Oncol 2025; 18:42. [PMID: 40234884 PMCID: PMC12001465 DOI: 10.1186/s13045-025-01698-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: 12/20/2024] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Gastric cancer is the fifth most common cancer globally and is associated with significant morbidity and mortality. Despite its alarming prevalence, limited comparative evidence exists on its treatment efficacy and prognosis across diverse China populations. METHODS To address this, our study used a large-scale dataset from the National Cancer Information Database, including data from 220,304 patients from 53 leading hospitals across 27 provinces in China. RESULTS From 2017 to 2023, early-stage (Stages I-II) gastric cancer diagnoses increased to 35.63% of all cancer cases. Our study evaluated the neoadjuvant treatment strategies, adjuvant post-operative therapy, first- and second-line management for progressive stages, alongside current gastric cancer treatment guidelines in China. Notably, immunotherapy accounted for 16.17% and 23.28% of first- and second-line treatments for late-stage gastric cancers, and 14.56% and 5.00% for neoadjuvant and adjuvant therapies, respectively. Analysis of survival rates revealed that the 1-, 2-, 3-, 4-, and 5-year survival rates were 74.07%, 54.89%, 44.21%, 37.97%, and 33.53%, respectively. The 5-year survival rates across stages I-IV were 85.07%, 49.34%, 35.56%, and 13.15%, respectively. CONCLUSIONS These findings offer critical insights into the current state of gastric cancer treatment in China and can inform future initiatives to improve therapeutic outcomes for patients with gastric cancer.
Collapse
Affiliation(s)
- Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Keren Jia
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Dan Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lei Jiang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | | | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
21
|
Tan Y, Xing Y, Yuan S, Sun F, Lin X, Bao S, Jiang D, Zhang J, Sun SL. Potential Value of AURKA and CDK6 Amplification for the Response of Patients With Gastric Cancer to Neoadjuvant Chemotherapy. Mol Carcinog 2025. [PMID: 40222043 DOI: 10.1002/mc.23921] [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: 12/16/2024] [Revised: 03/19/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025]
Abstract
Many patients respond poorly to neoadjuvant chemotherapy (NACT), negatively affecting the surgical success rate. Identifying effective biomarkers and understanding the potential resistance mechanisms are urgently needed. Data of 18 patients with advanced stomach cancer who were treated with NACT categorized according to tumor regression grade into major histological response (MJHR) and nonhistological response (NHR) groups were retrospectively analyzed. Genomic signatures associated with the response to NACT were identified using whole-exome and RNA sequencing. Extraction of molecular signatures revealed increased deficient mismatch repair signature and tumor mutation levels in the NHR group. Compared to the MJHR group, the NHR group was also characterized by a greater number of copy number alterations (p = 0.08), which was further confirmed by RNA sequencing, and upregulation of aurora kinase A (AURKA) (p = 0.05) and cyclin-dependent kinase 6 (CDK6) (p = 0.049). Western blot analysis and immunohistochemical analyses further confirmed high CDK6 (p < 0.01/p < 0.0001) and AURKA (p < 0.01/p < 0.001) expression levels in the NHR group. Finally, palbociclib, an inhibitor of CDK4/6, effectively inhibited the proliferation (p < 0.05) and induced apoptosis of oxaliplatin-resistant gastric cancer cells (p < 0.01) in vitro. These findings support the potential value of AURKA and CDK6 amplification, as well as their effects on the tumor microenvironment, in predicting poor outcomes of NACT in patients with locally advanced gastric cancer. Thus, CDK4/6 inhibitors could be used to treat NACT-resistant patients with gastric cancer.
Collapse
Affiliation(s)
- Yuen Tan
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Yao Xing
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Shuai Yuan
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Fan Sun
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Xiaohui Lin
- School of Computer Science and Technology, Dalian University of Technology, Dalian, China
| | - Simeng Bao
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Dongyue Jiang
- Key Laboratory of Ocean Energy Utilization and Energy Conservation of Ministry of Education, Dalian University of Technology, Dalian, China
| | - Jianjun Zhang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Shu-Lan Sun
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| |
Collapse
|
22
|
Zhu K, Pan Z, Qin M, Huang J. The cost effectiveness of penpulimab with paclitaxel and carboplatin in first-line treatment of metastatic squamous non-small cell lung cancer. Sci Rep 2025; 15:12679. [PMID: 40221588 PMCID: PMC11993585 DOI: 10.1038/s41598-025-97591-2] [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: 11/30/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
This study aimed to evaluate the cost-effectiveness of Penpulimab versus placebo in treating metastatic squamous non-small cell lung cancer (NSCLC) from the perspective of Chinese payers. A three-state Markov model was developed to simulate clinical efficacy and cost consumption using Kaplan-Meier curves from clinical trials. The model considered only direct medical costs, with utility values derived from the published literature. The primary outcome measure was the incremental cost-effectiveness ratio (ICER), and sensitivity analysis was performed to assess the impact of parameter uncertainty on the model's robustness. The base case analysis indicated that the Penpulimab group incurred higher costs ($33,592 vs. $9,351) than the placebo group, while also providing more quality-adjusted life years (QALYs) (3.30 vs. 2.11), resulting in an incremental cost-effectiveness ratio (ICER) of $20,389.38 per QALY. Sensitivity analyses revealed that the cost of Penpulimab, along with the utilities of progression-free survival (PFS) and progression of disease (PD), were the parameters that most significantly influenced the model's outcomes. From the perspective of Chinese payers, Penpulimab offers a cost-effectiveness advantage over placebo in treating metastatic squamous NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/economics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/mortality
- Cost-Benefit Analysis
- Paclitaxel/administration & dosage
- Paclitaxel/therapeutic use
- Paclitaxel/economics
- Lung Neoplasms/drug therapy
- Lung Neoplasms/economics
- Lung Neoplasms/pathology
- Lung Neoplasms/mortality
- Carboplatin/administration & dosage
- Carboplatin/therapeutic use
- Carboplatin/economics
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/economics
- Quality-Adjusted Life Years
- Markov Chains
- Antibodies, Monoclonal, Humanized/economics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Male
- Female
- Neoplasm Metastasis
- Progression-Free Survival
- Cost-Effectiveness Analysis
Collapse
Affiliation(s)
- Kaiqi Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhaoyi Pan
- Medical Record Management and Statistics Information Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Mengyao Qin
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
23
|
Sugimura K, Motoori M, Kentaro K, Yamamoto K, Takeno A, Hara H, Hamakawa T, Murakami K, Nakahara Y, Masuzawa T, Omori T, Kurokawa Y, Fujitani K, Doki Y. Comparison of laparoscopic and open gastrectomy after neoadjuvant chemotherapy for locally advanced gastric cancer: a propensity score matching analysis. Surg Endosc 2025; 39:2304-2315. [PMID: 39948263 DOI: 10.1007/s00464-025-11595-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/29/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND We investigated the safety and efficacy of laparoscopic gastrectomy (LG) in patients with locally advanced gastric cancer who underwent neoadjuvant chemotherapy (NAC). METHODS This study included 247 consecutive patients with advanced gastric cancer who underwent NAC followed by gastrectomy between 2007 and 2017 at one of six institutions. The patients were divided into the open gastrectomy (OG) and LG groups. The short- and long-term outcomes in both groups were investigated after propensity score matching. RESULTS After propensity score matching, 72 pairs of patients were selected. The baseline characteristics were not significantly different after matching. Compared with the OG group, the LG group had a significantly longer operative time (360 vs. 305 min, P = 0.002) and less intraoperative blood loss (271 vs. 652 mL, P < 0.001). The LG group had more harvested lymph nodes than the OG group (57.4 vs. 45.1, P < 0.001). The frequency of Clavien-Dindo grade ≥ 2 postoperative complications was not significantly different (26% vs. 22%, P = 0.698). The interval between surgery and postoperative chemotherapy was significantly shorter in the LG group (48.7 vs. 68.6 days, P = 0.048). The 5-year overall survival rates in the OG and LG groups were 54.4% and 53.5%, respectively. The overall survival was similar between the two groups (P = 0.773). No significant differences were observed between the two groups in terms of the type of recurrence, including lymph node, hematogenous, and peritoneal recurrences (P = 1.000, P = 1.000, and P = 0.686, respectively). CONCLUSIONS Based on both short- and long-term results, LG is a potential therapeutic option for patients with gastric cancer who undergo NAC.
Collapse
Affiliation(s)
- Keijiro Sugimura
- Department of Surgery, Kansai Rosai Hospital, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan.
| | - Masaaki Motoori
- Department of Surgery, Osaka General Medical Center, Osaka, Japan
| | - Kishi Kentaro
- Department of Surgery, Osaka Police Hospital, Osaka, Japan
| | - Kazuyoshi Yamamoto
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Atsushi Takeno
- Department of Surgery, National Hospital Organization, Osaka National Hospital, Osaka, Japan
| | - Hisashi Hara
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Takuya Hamakawa
- Department of Surgery, National Hospital Organization, Osaka National Hospital, Osaka, Japan
| | - Kohei Murakami
- Department of Surgery, Kansai Rosai Hospital, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | | | - Toru Masuzawa
- Department of Surgery, Kansai Rosai Hospital, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Takeshi Omori
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Yukinori Kurokawa
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | - Yuichiro Doki
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
24
|
Kano M, Tokumoto N, Tanabe K, Hihara J, Toyota K, Hotta R, Saeki Y, Tazawa H, Fujikuni N, Hiroshima Surgical Study Group of Clinical Oncology (HiSCO). Gastrectomy after neoadjuvant chemotherapy in five cases of locally advanced gastric cancer with pancreatic head invasion. Int Cancer Conf J 2025; 14:147-154. [PMID: 40160878 PMCID: PMC11950593 DOI: 10.1007/s13691-025-00750-5] [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: 07/25/2024] [Accepted: 01/13/2025] [Indexed: 04/02/2025] Open
Abstract
Locally advanced gastric cancer (LAGC) with pancreatic head invasion (T4b) carries a poor prognosis despite radical surgery. Herein, we report the effectiveness of neoadjuvant chemotherapy (NAC) in improving the resectability of advanced gastric cancer with pancreatic invasion. A total of 2191 cases of gastric cancer were retrospectively analyzed from 13 institutions within the Hiroshima Surgical Study Group of Clinical Oncology (Hisco) database from 2018 to 2020. Among them, 5 of the 24 patients with Stage cT4b gastric cancer underwent NAC for three-to-eight cycles. Following chemotherapy, three patients underwent total gastrectomy, two patients underwent distal gastrectomy, and no patient underwent pancreaticoduodenectomy (PD). All five patients achieved a chemotherapeutic response of Grade 1b or higher, and only one case showed residual pancreatic invasion on pathology. This study suggests that NAC for Stage T4b LAGC with pancreatic head invasion may have the potential to obviate the need for PD.
Collapse
Affiliation(s)
- Mikihiro Kano
- Department of Digestive Surgery, Hiroshima City North Medical Center Asa Citizens Hospital, 1-2-1 Kameyama-minami, Asakita-ku, Hiroshima, 7310293 Japan
| | - Noriaki Tokumoto
- Department of Digestive Surgery, Hiroshima City North Medical Center Asa Citizens Hospital, 1-2-1 Kameyama-minami, Asakita-ku, Hiroshima, 7310293 Japan
| | - Kazuaki Tanabe
- Department of Perioperative and Critical Care Management, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Jun Hihara
- Department of Digestive Surgery, Hiroshima City North Medical Center Asa Citizens Hospital, 1-2-1 Kameyama-minami, Asakita-ku, Hiroshima, 7310293 Japan
| | - Kazuhiro Toyota
- Department of Surgery, Hiroshima Memorial Hospital, Hiroshima, Japan
| | - Ryuichi Hotta
- Department of Surgery, National Hospital Organization Higashihiroshima Medical Center, Higashihiroshima, Japan
| | - Yoshihiro Saeki
- Department of Gastroenterological and Transplant Surgery, Hiroshima University, Hiroshima, Japan
| | - Hirofumi Tazawa
- Department of Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Nobuaki Fujikuni
- Department of Digestive Surgery, Hiroshima Prefectural Hospital, Hiroshima, Japan
| | - Hiroshima Surgical Study Group of Clinical Oncology (HiSCO)
- Department of Digestive Surgery, Hiroshima City North Medical Center Asa Citizens Hospital, 1-2-1 Kameyama-minami, Asakita-ku, Hiroshima, 7310293 Japan
- Department of Perioperative and Critical Care Management, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Surgery, Hiroshima Memorial Hospital, Hiroshima, Japan
- Department of Surgery, National Hospital Organization Higashihiroshima Medical Center, Higashihiroshima, Japan
- Department of Gastroenterological and Transplant Surgery, Hiroshima University, Hiroshima, Japan
- Department of Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Japan
- Department of Digestive Surgery, Hiroshima Prefectural Hospital, Hiroshima, Japan
| |
Collapse
|
25
|
Li R, Li J, Wang Y, Liu X, Xu W, Sun R, Xue B, Zhang X, Ai Y, Du Y, Jiang J. The artificial intelligence revolution in gastric cancer management: clinical applications. Cancer Cell Int 2025; 25:111. [PMID: 40119433 PMCID: PMC11929235 DOI: 10.1186/s12935-025-03756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/18/2025] [Indexed: 03/24/2025] Open
Abstract
Nowadays, gastric cancer has become a significant issue in the global cancer burden, and its impact cannot be ignored. The rapid development of artificial intelligence technology is attempting to address this situation, aiming to change the clinical management landscape of gastric cancer fundamentally. In this transformative change, machine learning and deep learning, as two core technologies, play a pivotal role, bringing unprecedented innovations and breakthroughs in the diagnosis, treatment, and prognosis evaluation of gastric cancer. This article comprehensively reviews the latest research status and application of artificial intelligence algorithms in gastric cancer, covering multiple dimensions such as image recognition, pathological analysis, personalized treatment, and prognosis risk assessment. These applications not only significantly improve the sensitivity of gastric cancer risk monitoring, the accuracy of diagnosis, and the precision of survival prognosis but also provide robust data support and a scientific basis for clinical decision-making. The integration of artificial intelligence, from optimizing the diagnosis process and enhancing diagnostic efficiency to promoting the practice of precision medicine, demonstrates its promising prospects for reshaping the treatment model of gastric cancer. Although most of the current AI-based models have not been widely used in clinical practice, with the continuous deepening and expansion of precision medicine, we have reason to believe that a new era of AI-driven gastric cancer care is approaching.
Collapse
Affiliation(s)
- Runze Li
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Jingfan Li
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Yuman Wang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Xiaoyu Liu
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Weichao Xu
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China
| | - Runxue Sun
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China
| | - Binqing Xue
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Xinqian Zhang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Yikun Ai
- North China University of Science and Technology, Tanshan 063000, China
| | - Yanru Du
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China.
- Hebei Provincial Key Laboratory of Integrated Traditional and Western Medicine Research on Gastroenterology, Hebei, 050011, China.
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, 050011, China.
| | - Jianming Jiang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China.
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China.
| |
Collapse
|
26
|
Li TH, Sun X, Li CG, Yin YP, Tao KX. Hypercoagulation after neoadjuvant immunochemotherapy as a new prognostic indicator in patients with locally advanced gastric cancer undergoing surgery. World J Gastrointest Oncol 2025; 17:100927. [PMID: 40092957 PMCID: PMC11866221 DOI: 10.4251/wjgo.v17.i3.100927] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/06/2024] [Accepted: 12/25/2024] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Coagulation status is closely related to the progression of malignant tumors. In the era of neoadjuvant immunochemotherapy (NICT), the prognostic utility of coagulation indicators in patients with locally advanced gastric cancer (LAGC) undergoing new treatments remains to be determined. AIM To determine whether hypercoagulation is an effective prognostic indicator in patients with LAGC who underwent radical resection after NICT. METHODS A retrospective analysis of clinical data from 104 patients with LAGC, who underwent radical resection after NICT between 2020 and 2023, was performed. D-dimer and fibrinogen concentrations were measured one week before NICT, and again one week before surgery, to analyze the association between these two indicators and their combined indices [non-hypercoagulation (D-dimer and fibrinogen concentrations within the upper limit of normal) vs hypercoagulation (D-dimer or fibrinogen concentrations above the upper limit of normal)] with prognosis. After radical resection, patients were followed-up periodically. The median follow-up duration was 21 months. RESULTS Data collected after NICT revealed that the three-year overall survival (OS) and disease-free survival (DFS) rates the non-hypercoagulation group were significantly better than those in the hypercoagulation group [94.4% vs 78.0% (P = 0.019) and 87.0% vs 68.0% (P = 0.027), respectively]. Multivariate analysis indicated that hypercoagulation after NICT was an independent factor for poor postoperative OS [hazard ratio (HR) 4.436, P = 0.023] and DFS (HR 2.551, P = 0.039). Pre-NICT data demonstrated no statistically significant difference in three-year OS between the non-hypercoagulation and hypercoagulation groups (88.3% vs 84.1%, respectively; P = 0.443). CONCLUSION Hypercoagulation after NICT is an effective prognostic indicator in patients with LAGC undergoing radical gastrectomy.
Collapse
Affiliation(s)
- Tian-Hao Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Xiong Sun
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Cheng-Guo Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Yu-Ping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Kai-Xiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
27
|
Yu X, Cui R, Guo P. Evaluation of the efficacy and safety of toripalimab combination therapy for treatment of advanced gastric cancer: a meta-analysis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2025; 18:96-109. [PMID: 40226111 PMCID: PMC11982770 DOI: 10.62347/gzow5960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 02/12/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND To systematically evaluate the efficacy and safety of combination therapy with toripalimab in the treatment of advanced gastric cancer (GC). METHODS We conducted a thorough search for relevant studies in PubMed, Embase, Cochrane Library, and Web of Science. Effect estimates were computed utilizing Stata software (version 14.0) and either random or fixed effects models, as applicable. A subgroup analysis was undertaken to assess the effect of various combination therapies on overall response rate (ORR). Begg and Egger's tests were employed to assess publication bias. RESULTS The study consisted of 8 trials, which included 277 participants with advanced gastric cancer. The overall ORR was 41.4% (95% CI, 32.4%-50.3%), with a disease control rate (DCR) of 83.6% (95% CI, 74.6%-92.7%), a median overall survival (mOS) of 11.0 months (95% CI, 9.6-12.4), and a median progression-free survival (mPFS) of 4.2 months (95% CI, 2.5-6.0) for the combination therapy with toripalimab. Subgroup analysis revealed that the combination of toripalimab and chemotherapy achieved a greater ORR compared to the non-chemotherapy group, with ORR rates of 49.8% (95% CI, 42.2%-57.4%) and 31.9% (95% CI, 26.7%-37.1%), respectively. The combination therapy with toripalimab led to adverse events (AEs) of any grade at 94.0% of cases (95% CI, 89.5%-98.5%) and grade 3 AEs at 32.4% (95% CI, 17.8%-47.1%). The sensitivity analysis indicated that no single study affected the overall results. CONCLUSIONS Combination therapy of toripalimab can improve clinical efficacy, although with increased but manageable toxicity. Additional clinical trials are required to assess comprehensively the efficacy and safety of alternative toripalimab regimens. The review agreement has been recorded with PROSPERO (CRD42024585696).
Collapse
Affiliation(s)
- Xinlin Yu
- Department of Oncology, Affiliated Hospital Chengdu UniversityChengdu, Sichuan, China
| | - Ran Cui
- Department of Emergency, The First People’s Hospital of NeijiangNeijiang, Sichuan, China
| | - Ping Guo
- Department of Cardiology, Affiliated Hospital Chengdu UniversityChengdu, Sichuan, China
| |
Collapse
|
28
|
Kang YK, Kim HD, Cho H, Park YS, Lee JS, Ryu MH. Phase 2 study of neoadjuvant durvalumab plus docetaxel, oxaliplatin, and S-1 with surgery and adjuvant durvalumab plus S-1 for resectable locally advanced gastric cancer. J Immunother Cancer 2025; 13:e010635. [PMID: 40081945 PMCID: PMC11907044 DOI: 10.1136/jitc-2024-010635] [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: 09/21/2024] [Accepted: 02/01/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Based on the phase 3 PRODIGY study, neoadjuvant docetaxel, oxaliplatin, and S-1 (DOS) have emerged as a viable treatment option for Asian patients with resectable locally advanced gastric cancer (LAGC). This phase 2 study evaluated the efficacy and safety of combining neoadjuvant durvalumab with DOS, followed by surgery and adjuvant durvalumab plus S-1 chemotherapy, for resectable LAGC. METHODS Patients with LAGC with cT2/3N+or cT4Nany tumors were enrolled in this study. Patients with proficient mismatch repair protein (pMMR) tumors received three cycles of neoadjuvant durvalumab plus DOS, administered every 3 weeks, followed by surgery and adjuvant S-1 plus durvalumab (main study arm). The primary endpoints were the rate of pathologic complete regression (pCR) and safety. An exploratory arm evaluated patients with deficient mismatch repair protein (dMMR) tumors, who received three cycles of neoadjuvant durvalumab and tremelimumab, followed by surgery and adjuvant durvalumab. RESULTS In the main study arm, 50 pMMR patients were enrolled, and received at least one dose of neoadjuvant treatment. The median age was 63 years, with 72.0% being men. 18 and 32 patients presented with clinical stage II and III tumors, respectively. 49 (98.0%) underwent surgery, with 45 achieving R0 resection. A pCR rate of 30.0% was observed, meeting the prespecified primary efficacy endpoint. With a median follow-up of 21.8 months, the 3-year progression-free survival and overall survival rates were 69.9% and 88.1%, respectively. 10% of patients experienced predefined unacceptable severe toxicities, including febrile neutropenia (n=3) and persistent G4 neutropenia (n=2) lasting more than 7 days, thereby meeting the primary safety endpoint. Nine patients with dMMR tumors were enrolled in the exploratory arm. All nine underwent surgery, with a pCR rate of 22.2%. CONCLUSIONS This study met its primary efficacy and safety endpoints. The combination of neoadjuvant durvalumab plus DOS, followed by surgery and adjuvant durvalumab plus S-1 chemotherapy, warrants further investigation in a phase 3 trial for Asian patients with LAGC. CLINICAL TRIAL INFORMATION 04221555.
Collapse
Affiliation(s)
- Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea (the Republic of)
| | - Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea (the Republic of)
| | - Hyungwoo Cho
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea (the Republic of)
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Korea (the Republic of)
| | - Jong Seok Lee
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea (the Republic of)
| |
Collapse
|
29
|
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
|
30
|
Ou H, Zhuang J, Jian M, Zheng X, Wu T, Cheng H, Qian R. Perioperative versus adjuvant chemotherapy for resectable gastric cancer: a meta-analysis of randomized controlled trials. Front Oncol 2025; 15:1432596. [PMID: 40115020 PMCID: PMC11922704 DOI: 10.3389/fonc.2025.1432596] [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: 05/14/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Objectives To report the latest systematic review and meta-analysis of randomized controlled trials (RCT) to compare perioperative versus adjuvant chemotherapy for resectable gastric cancer. Methods We conducted a systematic literature retrieval via PubMed, Embase, Web of Science, and Cochrane until April, 2024 for RCT which compared perioperative versus adjuvant chemotherapy for resectable gastric cancer. Outcomes measured were overall survival (OS) and progression-free survival (PFS). Results 5 RCTs including 2,735 patients were included for meta-analysis. Meta-analysis revealed a significant longer PFS in the neoadjuvant chemotherapy (NAC) group (HR: 0.77; 95% CI: 0.69, 0.85; P<0.00001) compared with adjuvant chemotherapy (AC) group. Subgroup analysis found that there was still a significant superiority of NAC in female (HR: 0.53; 95% CI: 0.40, 0.70; P<0.0001) and cN+ (HR: 0.77; 95% CI: 0.67, 0.89; P=0.0005) patients, while the superiority disappeared in male (HR: 0.87; 95% CI: 0.74, 1.01; P=0.07) and cN- patients (HR: 0.91; 95% CI: 0.46, 1.78; P=0.77). In addition, meta-analysis observed a trend towards improved OS with NAC (HR: 0.86; 95% CI: 0.70, 1.07; P = 0.17), and sensitivity analysis demonstrated instability in OS. Conclusions NAC can significantly prolong PFS in patients with resectable gastric cancer compared to AC, and the benefit is more significant in women and cN+ patients. Besides, our analysis indicated that NAC has a potential to improve OS compared with AC. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42024546165.
Collapse
Affiliation(s)
- Haiya Ou
- Department of Gastroenterology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jiamei Zhuang
- Department of Nephrology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Mingwei Jian
- Department of Gastroenterology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xinyi Zheng
- Department of Gastroenterology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Tingping Wu
- Department of Gastroenterology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Honghui Cheng
- Department of Gastroenterology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Rui Qian
- Department of Gastroenterology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
31
|
Gan X, Jia Y, Shan F, Ying X, Li S, Zhang Y, Pang F, Li Z. Comprehensive evaluation of tumor response better evaluates the efficacy of neoadjuvant chemotherapy and predicts the prognosis in gastric cancer - a post hoc analysis of a single-center randomized controlled trial. BMC Cancer 2025; 25:401. [PMID: 40045265 PMCID: PMC11884205 DOI: 10.1186/s12885-024-13372-6] [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: 04/06/2024] [Accepted: 12/19/2024] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Perioperative chemotherapy combined with D2 radical gastrectomy has been proven to be the standard treatment for local advanced gastric cancer. However, tumor regression grading (TRG) is the only neoadjuvant chemotherapy (NACT) response evaluation criterion recommended by the NCCN guideline for gastric cancer (GC). Given TRG's limitations, we aim to explore a better comprehensive response evaluation method in this study. METHODS Clinical information of 96 GC patients who received NACT was collected prospectively. Clinicopathological variables predictive of the response to NACT were identified by comparing the pre- and post-NACT examination results. The correlations between the response mode and long-term survival rate were assessed. RESULTS Univariate Cox regression analysis showed that CT-based evaluation of the primary lesion thickness (CT-thickness) and tumor markers (TMs) were significantly associated with prognosis. The comprehensive evaluation method, including CT-thickness, TRG, and TMs, was constructed and proved to have a higher Harrell's C index. Significant differences in overall survival (OS) and recurrence-free survival (RFS) were observed between responders and non-responders distinguished by the comprehensive evaluation method. CONCLUSIONS The combination of CT-thickness, TRG, and TMs could be used to construct a pragmatic NACT efficacy evaluation method with both high sensitivity and specificity, which could facilitate clinical decision-making, NACT-related clinical research conduction, and efficacy predictive biomarker exploration.
Collapse
Affiliation(s)
- Xuejun Gan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), gastrointestinal surgery of department, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yongning Jia
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, gastrointestinal surgery of department, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Fei Shan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, gastrointestinal surgery of department, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiangji Ying
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), gastrointestinal surgery of department, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shuangxi Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), gastrointestinal surgery of department, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), gastrointestinal surgery of department, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Fei Pang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), gastrointestinal surgery of department, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Ziyu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), gastrointestinal surgery of department, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
32
|
Zhang X, Liang H, Li Z, Xue Y, Wang Y, Zhou Z, Yu J, Bu Z, Chen L, Du Y, Wang X, Wu A, Li G, Su X, Xiao G, Cui M, Wu D, Chen L, Wu X, Zhou Y, Zhang L, Dang C, He Y, Zhang Z, Sun Y, Li Y, Chen H, Bai Y, Wang Y, Yu P, Zhu G, Suo J, Jia B, Li L, Huang C, Li F, Ye Y, Xu H, Wang X, Yuan Y, E J, Ying X, Yao C, Shen L, Ji J. Perioperative or postoperative adjuvant oxaliplatin with S-1 versus adjuvant oxaliplatin with capecitabine in patients with locally advanced gastric or gastro-oesophageal junction adenocarcinoma undergoing D2 gastrectomy (RESOLVE): final report of a randomised, open-label, phase 3 trial. Lancet Oncol 2025; 26:312-319. [PMID: 39952264 DOI: 10.1016/s1470-2045(24)00676-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 02/17/2025]
Abstract
BACKGROUND The multicentre RESOLVE trial examined the efficacy of perioperative and postoperative S-1 and oxaliplatin (SOX) compared with postoperative capecitabine and oxaliplatin (CapOx) in gastric or gastro-oesophageal junction cancer. Initial analyses did not encompass overall survival owing to the immature data. This paper provides an updated analysis of the survival data from the RESOLVE trial. METHODS In this randomised, open-label, phase 3 study, participants aged 18 years or older with cT4a N+ M0 or cT4b Nany M0 gastric or gastro-oesophageal junction adenocarcinoma who were feasible for D2 lymphadenectomy and had a Karnofsky performance score of 70 or higher were enrolled. Participants were randomly assigned in a 1:1:1 ratio via an interactive web response system, stratified by participating centres and Lauren classification, to receive adjuvant CapOx (eight postoperative cycles of intravenous oxaliplatin 130 mg/m2 on day 1 of each 21-day cycle plus oral capecitabine 1000 mg/m2 twice a day on days 1-14, adjuvant SOX (eight postoperative cycles of intravenous oxaliplatin 130 mg/m2 on day 1 of each 21-day cycle plus oral S-1 40-60 mg twice a day on days 1-14), or perioperative SOX (intravenous oxaliplatin 130 mg/m2 on day 1 of each 21-day cycle plus oral S-1 40-60 mg twice a day for three cycles preoperatively and five cycles postoperatively followed by three cycles of S-1 monotherapy. The primary endpoint, assessed in the modified intention-to-treat population, was 3-year disease-free survival to assess the superiority of perioperative-SOX compared with adjuvant-CapOx and the non-inferiority (hazard ratio [HR] non-inferiority margin of 1·33) of adjuvant-SOX compared with adjuvant-CapOx, and has been reported previously. This final report focuses on the secondary endpoint of 5-year overall survival, also assessed in the modified intention-to-treat population. Other secondary endpoints-R0 resection rate and safety-were not updated in this analysis. The study is registered at ClinicalTrials.gov, NCT01534546, and is complete. FINDINGS Between Aug 15, 2012, and Feb 28, 2017, 1094 patients were enrolled and randomly assigned, of whom 1022 participants were included in the modified intention-to-treat population: 345 (259 male, 86 female) in the adjuvant-CapOx group, 340 (238 male, 102 female) in the adjuvant-SOX group, and 337 (271 male, 66 female) in the perioperative-SOX group. As of April 7, 2022, the median duration of follow-up was 62·8 months (IQR 52·0-75·1). The 5-year overall survival rates were 52·1% (95% CI 46·3-57·5) for the adjuvant-CapOx group, 61·0% (55·3-66·2) for the adjuvant-SOX group, and 60·0% (54·2-65·3), for the perioperative-SOX group. Overall survival was significantly prolonged with perioperative-SOX (HR 0·79; 95% CI 0·62-1·00, p=0·049) and adjuvant-SOX (HR 0·77, 0·61-0·98, p=0·033), compared with adjuvant-CapOx. INTERPRETATION Consistent with the initial analysis of 3-year disease-free survival, the extended 5-year overall survival analysis from the RESOLVE trial confirmed the survival advantage of perioperative-SOX and adjuvant-SOX compared with the standard adjuvant-CapOx regimen. The SOX regimen, given perioperatively or as an adjuvant treatment, emerges as a potential standard treatment modality for locally advanced gastric or gastro-oesophageal junction cancer management in Asian patients. FUNDING The National Key Research and Development Program of China, the National Natural Science Foundation of China, the Capital's Funds for Health Improvement and Research, the Beijing Natural Science Foundation, National Natural Science Foundation of China, the Beijing Natural Science Foundation, Taiho, Hengrui Pharmaceutical and Sanofi-Aventis. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Xiaotian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Han Liang
- Department of Abdominal Oncology Surgery, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Ziyu Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yingwei Xue
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yanong Wang
- Department of Stomach Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhiwei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jiren Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhaode Bu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Chen
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China; Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yian Du
- Department of General Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xinbao Wang
- Department of Hepatopancreatobiliary Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Aiwen Wu
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Guoli Li
- Department of General Surgery, Jinling Hospital, Nanjing, China
| | - Xiangqian Su
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Gang Xiao
- Department of General Surgery, Beijing Hospital, Beijing, China
| | - Ming Cui
- Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Dan Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Li Chen
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China; Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojiang Wu
- Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanbing Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lianhai Zhang
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chengxue Dang
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yulong He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Li
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Huanqiu Chen
- Department of General Surgery, Jiangsu Cancer Hospital, Nanjing, China
| | - Yuxian Bai
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yakun Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Peiwu Yu
- Department of General Surgery, The First Hospital Affiliated to Army Medical University, Chongqing, China
| | - Guanbao Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Suo
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Baoqing Jia
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Jinan, China
| | - Changming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Yingjiang Ye
- Department of General Surgery, Peking University People's Hospital, Beijing, China
| | - Huimian Xu
- Department of Gastrointestinal Oncology Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Xin Wang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yannan Yuan
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jianyu E
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Xiangji Ying
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chen Yao
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Jiafu Ji
- Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
33
|
van Laarhoven H. Management of locally advanced gastric cancer. Lancet Oncol 2025; 26:267-269. [PMID: 39952263 DOI: 10.1016/s1470-2045(24)00727-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 02/17/2025]
Affiliation(s)
- Hanneke van Laarhoven
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam 1105 AZ, Netherlands; Cancer Treatment & Quality of Life, Cancer Center Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
34
|
He J, Zhang B, Zhou S, Yang Y, Han Z, Wu T, Qiao Q, Yang H, He X, Wang N. Phase II study of perioperative camrelizumab and XELOX for locally advanced gastric or gastroesophageal junction adenocarcinoma. Cancer Sci 2025; 116:736-743. [PMID: 39656600 PMCID: PMC11875781 DOI: 10.1111/cas.16425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/15/2024] [Accepted: 11/24/2024] [Indexed: 12/17/2024] Open
Abstract
Immune checkpoint inhibitors combined with chemotherapy have shown promising efficacy in treating gastric or gastroesophageal junction (G/GEJ) adenocarcinoma in the neoadjuvant setting. This phase II trial (NCT05715632) aimed to investigate the efficacy and safety of perioperative camrelizumab plus XELOX in patients with locally advanced G/GEJ adenocarcinoma. Treatment-naive patients with cT3-4aN1-3 M0 resectable locally advanced G/GEJ adenocarcinoma were recruited to receive camrelizumab (200 mg, intravenously) on Day 1 combined with XELOX (oxaliplatin at 130 mg/m2 on Day 1 and capecitabine at 1000 mg/m2 on Days 1-14) every 3 weeks for four cycles, followed by surgery and adjuvant camrelizumab combined with XELOX every 3 weeks for four cycles. The primary endpoint was the pathological complete response (pCR; ypT0N0) rate. From September 2020 to January 2023, 46 patients were enrolled, and all patients completed neoadjuvant therapy. Among them, 43 underwent D2 resection. In the intention-to-treat population, pCR was achieved in nine patients (19.6%, 95% confidence interval [CI]: 9.9%-34.4%), and the major pathological response was achieved in 25 patients (54.3%, 95% CI: 39.2%-68.8%). The objective response rate was 69.6%, of which 12 patients achieved a complete response and 20 patients achieved a partial response. The 1-year event-free survival and disease-free survival rates were both 93.1%. Treatment-related adverse events (TRAEs) occurred in 42 (91.3%) patients, and grade 3 TRAEs occurred in nine (19.6%) patients. No grades 4-5 TRAEs were observed. Perioperative camrelizumab combined with XELOX showed promising pathological response with an acceptable safety profile in patients with resectable locally advanced G/GEJ adenocarcinoma.
Collapse
Affiliation(s)
- Jiaxing He
- Department of General SurgeryAir Force Medical University Tangdu HospitalXi'anChina
| | - Bo Zhang
- Department of General SurgeryAir Force Medical University Tangdu HospitalXi'anChina
| | - Shuai Zhou
- Department of General SurgeryAir Force Medical University Tangdu HospitalXi'anChina
| | - Ying Yang
- Department of General SurgeryAir Force Medical University Tangdu HospitalXi'anChina
| | - Zhuo Han
- Department of General SurgeryAir Force Medical University Tangdu HospitalXi'anChina
| | - Tao Wu
- Department of General SurgeryAir Force Medical University Tangdu HospitalXi'anChina
| | - Qing Qiao
- Department of General SurgeryAir Force Medical University Tangdu HospitalXi'anChina
| | - Haicheng Yang
- Department of General SurgeryAir Force Medical University Tangdu HospitalXi'anChina
| | - Xianli He
- Department of General SurgeryAir Force Medical University Tangdu HospitalXi'anChina
| | - Nan Wang
- Department of General SurgeryAir Force Medical University Tangdu HospitalXi'anChina
| |
Collapse
|
35
|
Leng A, Madan V, Shah M, Gurau A, Johnston FM, Greer JB. Characterizing Patients with ypT0N1 Gastric Adenocarcinoma Within the AJCC Staging System. Ann Surg Oncol 2025; 32:2122-2128. [PMID: 39739259 DOI: 10.1245/s10434-024-16408-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/09/2024] [Indexed: 01/02/2025]
Abstract
INTRODUCTION The standard of care for gastric cancer in the United States involves perioperative chemotherapy. While most post-therapy pathologic staging results are concordant (i.e. ypT+N+ or ypT0N0), patients occasionally display discordant results, such as ypT0N1. Herein, we characterized the survival of patients with ypT0N1 staging to better determine their categorization within the American Joint committee on Cancer (AJCC) staging system. METHODS Using the National Cancer Database (NCDB), we queried all patients diagnosed with gastric adenocarcinoma from 2004 to 2021 who received neoadjuvant chemotherapy. Patients were stratified by their ypTNM stage: (1) ypT0N0; (2) ypT+N0; (3) ypT+N1; and (4) ypT0N1. Multivariable Cox proportional hazard regression was used to assess 5- and 10-year survival between ypTNM stages. RESULTS A total of 28,985 patients received neoadjuvant chemotherapy, of whom 2378 (8.2%) had ypT0N0, 9402 (32.4%) had ypT+N0, 5339 (18.4%) had ypT+N1, and 318 (1.1%) had ypT0N1 staging. Overall, patients had a median age of 64 years, with the majority being male (74.9%) or White (82.2%). Additionally, 50.2% received care from an academic center, and 53.3% received neoadjuvant chemotherapy and radiotherapy. On multivariable analysis, patients with ypT0N1 had a 105% higher risk for mortality within 5 years (adjusted hazards ratio [aHR] 2.05, 95% confidence interval [CI] 1.69-2.50) and 86% increased risk within 10 years (aHR 1.86, 95% CI 1.54-2.23) when compared with ypT0N0. CONCLUSION Patients with stage ypT0N1 disease have worse 5- and 10-year outcomes than those with node-negative disease. Thus, their survival pattern most closely matches patients with ypstage IIB and III disease.
Collapse
Affiliation(s)
- Albert Leng
- Division of Gastrointestinal Surgical Oncology, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vrinda Madan
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Manuj Shah
- Division of Gastrointestinal Surgical Oncology, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrei Gurau
- Division of Gastrointestinal Surgical Oncology, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fabian M Johnston
- Division of Gastrointestinal Surgical Oncology, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan B Greer
- Division of Gastrointestinal Surgical Oncology, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
36
|
Yamada Y, Seto Y, Yoshikawa T, Takeuchi H, Kitagawa Y, Kodera Y, Doki Y, Yoshida K, Muro K, Kabeya Y, Kamada A, Nagashima K, Kumamaru H, Tachimori H, Sasako M, Katai H, Konno H, Kakeji Y. Postoperative adjuvant chemotherapy in patients with gastric cancer based on the Nationwide Gastric Cancer Registry in Japan. Glob Health Med 2025; 7:13-27. [PMID: 40026857 PMCID: PMC11866910 DOI: 10.35772/ghm.2024.01080] [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: 10/23/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 03/05/2025]
Abstract
The nationwide registry of the Japanese Gastric Cancer Association contains data related to the efficacy of adjuvant chemotherapy and prognostic factors across this patient population; elderly patients with advanced resectable gastric cancer are especially prevalent. Here, we analyzed data from 34,931 patients, who were treated between 2011 and 2013 at 421 hospitals in Japan. Although adjuvant chemotherapy was effective overall, 75 years or older elderly patients had a worse prognosis compared to younger patients. The most administered adjuvant chemotherapy was S-1 monotherapy. Adjuvant S-1 monotherapy was also effective for patients with pT1N2, pT1N3, and pT3N0 stage II tumors, as well as patients with other stage II and III malignancies. Independent prognostic factors for poor overall and relapse-free survival in patients at both stage II and stage III were age 75 or older, male, preoperative Eastern Cooperative Oncology Group performance status (ECOG-PS) 1 or more, preoperative renal dysfunction, undifferentiated adenocarcinoma, undergoing total gastrectomy, open laparotomy, no adjuvant chemotherapy, D1 lymphadenectomy, residual tumor R1 or R2, and Clavien-Dindo classification grade II or higher. Age 75 or older, renal dysfunction, ECOG-PS 1 and total gastrectomy were also significant risk factors for postoperative complications and lower compliance with adjuvant chemotherapy. Our analysis also revealed that adjuvant chemotherapy after resection of cancer of gastric remnant and postoperative chemotherapy against CY1 gastric cancer were also effective. We conclude that adjuvant chemotherapy is effective for all stage II and III patients including age 75 or older gastric cancer patients, in addition to distal gastrectomy, proximal gastrectomy, and pylorus-preserving surgery to avoid total gastrectomy may improve surgical outcomes and quality of life for elderly patients.
Collapse
Affiliation(s)
- Yasuhide Yamada
- Department of Medical Research, National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Takaki Yoshikawa
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroya Takeuchi
- Department of Surgery, Hamamatsu University, School of Medicine, Hamamatsu, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University, School of Medicine, Tokyo, Japan
| | | | | | | | - Kei Muro
- Department of Pharmacotherapy, Aichi Cancer Center, Nagoya, Japan
| | | | - Ami Kamada
- Healthcare & Life Sciences, IBM Japan, Ltd, Tokyo, Japan
| | - Kengo Nagashima
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan
| | - Hiraku Kumamaru
- Department of Healthcare Quality Assessment, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hisateru Tachimori
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | | | | | - Hiroyuki Konno
- Hamamatsu University, School of Medicine, Hamamatsu, Japan
| | - Yoshihiro Kakeji
- Database Committee, The Japanese Society of Gastroenterological Surgery, Tokyo, Japan
- Division of Gastrointestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
37
|
Xu G, Liu T, Shen J, Guan Q. Neoadjuvant therapy with immune checkpoint inhibitors in combination with chemotherapy vs . chemotherapy alone in HER2(-) locally advanced gastric cancer: A propensity score-matched cohort study. Chin Med J (Engl) 2025; 138:459-471. [PMID: 38420853 PMCID: PMC11845188 DOI: 10.1097/cm9.0000000000003028] [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: 10/18/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND This study aims to compare the efficacy between neoadjuvant immune checkpoint inhibitors (ICIs) plus chemotherapy vs . chemotherapy, and neoadjuvant triplet vs . doublet chemotherapeutic regimens in locally advanced gastric/esophagogastric junction cancer (LAGC). METHODS We included LAGC patients from 47 hospitals in China's National Cancer Information Database (NCID) from January 2019 to December 2022. Using propensity score matching (PSM), we retrospectively analyzed the efficacy between neoadjuvant ICIs plus chemotherapy vs . chemotherapy alone, and neoadjuvant triplet vs . doublet chemotherapeutic regimens. The primary study result was the pathologic complete response (pCR) rate. The secondary study results were disease-free survival (DFS) and overall survival (OS). RESULTS A total of 1205 LAGC patients were included. After PSM, the ICIs plus chemotherapy and the chemotherapy cohorts had 184 patients each, while the doublet and triplet chemotherapy cohorts had 246 patients each. The pCR rate (14.13% vs . 7.61%, χ2 = 4.039, P = 0.044), and the 2-year (77.60% vs . 61.02%, HR = 0.67, 95% con-fidence interval [CI] 0.43-0.98, P = 0.048) and 3-year (70.55% vs . 61.02%, HR = 0.58, 95% CI 0.32-0.93, P = 0.048) DFS rates in the ICIs plus chemotherapy cohort were improved compared to those in the chemotherapy cohort. No significant increase was observed in the OS rates at both 1 year and 2 years. The pCR rates, DFS rates at 1-3 years, and OS rates at 1-2 years did not differ significantly between the doublet and triplet cohorts, respectively. No differences were observed in postoperative complications between any of the group comparisons. CONCLUSIONS Neoadjuvant ICIs plus chemotherapy improved the pCR rate and 2-3 years DFS rates of LAGC compared to chemotherapy alone, but whether short-term benefit could translate into long-term efficacy is unclear. The triplet regimen was not superior to the doublet regimen in terms of efficacy. The safety after surgery was similar between either ICIs plus chemotherapy and chemotherapy or the triplet and the doublet regimen.
Collapse
Affiliation(s)
- Gehan Xu
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Tianjiao Liu
- Department of Medical Data, Beijing Yiyong Technology Co., Ltd., Beijing 100102, China
| | - Jingyi Shen
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Quanlin Guan
- Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China
| |
Collapse
|
38
|
Vásquez-Jaico ER, Yan-Quiroz EF, Bonilla-Feria N, Gonzalez MNV, Guzmán LS, Vásquez-Tirado GA, Serna-Alarcon V. Upfront surgery versus chemotherapy neoadjuvant in the survival of patients with locally advanced gastric signet-ring-cell adenocarcinoma. A scoping review. Ecancermedicalscience 2025; 19:1843. [PMID: 40248270 PMCID: PMC12003976 DOI: 10.3332/ecancer.2025.1843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Indexed: 04/19/2025] Open
Abstract
Background Recent research suggests that neoadjuvant chemotherapy is not effective for gastric cancer with signet ring cells. Objective The present study performs a scoping review of research that seeks to determine whether neoadjuvant chemotherapy is more effective than upfront surgery in the survival of locally advanced signet ring gastric adenocarcinoma. Design Online databases such as Pubmed, scopus and embase were used to identify articles from the last 20 years that used survival, as an initial or secondary outcome variable, after upfront surgery or neoadjuvant chemotherapy as initial treatment in locally advanced gastric signet ring cells adenocarcinoma. Results After a systematic selection process, five primary studies were selected that evaluated neoadjuvant chemotherapy compared to primary surgery. Conclusion Neoadjuvant chemotherapy does not appear to have greater benefit than initial surgery in gastric adenocarcinoma with locally advanced sign ring cells, it is necessary to define which is the most appropriate qt scheme for adenocarcinoma with sign ring cells, clinical trials type studies are required to improve the evidence. Finally, a national clinical practice guide is required as an interpretative map for the management of gastric cancer which may be appropriate as a first step to know the reality.
Collapse
Affiliation(s)
- Erick R Vásquez-Jaico
- School of Medicine, Faculty of Medicine, Antenor Orrego Private University, Trujillo 13008, Perú
- Virgen De La Puerta High Complexity Hospital Essalud, Trujillo 13013, Perú
- https://orcid.org/0000-0003-4350-7461
| | - Edgar Fermín Yan-Quiroz
- School of Medicine, Faculty of Medicine, Antenor Orrego Private University, Trujillo 13008, Perú
- Virgen De La Puerta High Complexity Hospital Essalud, Trujillo 13013, Perú
- https://orcid.org/0000-0002-9128-4760
| | - Nicol Bonilla-Feria
- School of Medicine, Faculty of Medicine, Antenor Orrego Private University, Trujillo 13008, Perú
- https://orcid.org/0009-0006-3659-7277
| | | | - Luis Salas Guzmán
- National University of San Marcos, Lima 15001, Perú
- https://orcid.org/0009-0008-2893-272X
| | - Gustavo Adolfo Vásquez-Tirado
- School of Medicine, Faculty of Medicine, Antenor Orrego Private University, Trujillo 13008, Perú
- https://orcid.org/0000-0002-2109-6430
| | - Victor Serna-Alarcon
- School of Medicine, Faculty of Medicine, Antenor Orrego Private University, Trujillo 13008, Perú
- Jose Cayetano Heredia EsSalud Regional Hospital, Piura 20002, Perú
- https://orcid.org/0000-0002-9803-6217
| |
Collapse
|
39
|
Chen Y, Liu D, Wei K, Lin Y, Wang Z, Sun Q, Wang H, Peng J, Lian L. Carcinoembryonic antigen trajectory predicts pathological complete response in advanced gastric cancer after neoadjuvant chemotherapy. Front Oncol 2025; 15:1525324. [PMID: 39995833 PMCID: PMC11847669 DOI: 10.3389/fonc.2025.1525324] [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: 11/09/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Aims This study aims to develop a simple, clinically applicable classification system to predict pCR based on carcinoembryonic antigen (CEA) trajectory during NAC. Methods This study included 366 AGC patients who received NAC followed by radical gastrectomy. CEA levels were measured before, during, and after NAC, with changes classified into three trajectory types: Type I (>=80% decline), Type II (>=40% but <80% decline), and Type III (<40% decline or increase). We analyzed associations between these CEA trajectories, pCR, lymph node remission, and survival. Results pCR was achieved in 10.4% (38/366) of patients. pCR rates were significantly higher in Type I (41%) and Type II (15.8%) trajectories compared to Type III (6.7%). Lymph node remission also correlated with CEA trajectories, with Type I having the highest proportion of ypN0 (79.2%). Multivariate analysis identified CEA trajectory subtypes and tumor differentiation as independent predictors of pCR. This classification system proved robust across subgroups. Although no significant differences in overall survival were observed between subtypes, higher initial CEA levels were associated with worse survival. Conclusion The trajectory of CEA change during NAC is a promising predictor of pCR in AGC. This simple and accessible classification system may facilitate personalized surgical strategies for patients with AGC.
Collapse
Affiliation(s)
- Yonghe Chen
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dan Liu
- Department of Laboratory Science, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaikai Wei
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Lin
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Wang
- School of Nursing, Sun Yat-sen University, Guangzhou, China
| | - Qian Sun
- School of Nursing, Sun Yat-sen University, Guangzhou, China
| | - Huashe Wang
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junsheng Peng
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei Lian
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
40
|
Nie RC, Yuan SQ, Ding Y, Chen YM, Li YF, Liang CC, Cai MY, Chen GM, Wang W, Sun XW, Weng DS, Li DD, Zhao JJ, Chen XJ, Guan YX, Liu ZM, Liang Y, Luo M, Chi J, Qiu HB, Zhou ZW, Zhang XS, Chen YB. Perioperative tislelizumab plus chemotherapy for locally advanced gastroesophageal junction adenocarcinoma (NEOSUMMIT-03): a prospective, nonrandomized, open-label, phase 2 trial. Signal Transduct Target Ther 2025; 10:60. [PMID: 39910052 PMCID: PMC11799164 DOI: 10.1038/s41392-025-02160-8] [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: 07/30/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/07/2025] Open
Abstract
This prospective, nonrandomized, open-label phase 2 trial (Chinese Clinical Trial Registry, ChiCTR2200061906) aimed to evaluate the effectiveness of adding the PD-1 antibody tislelizumab to perioperative chemotherapy in patients with locally advanced gastroesophageal junction adenocarcinoma (GEJA). This study enrolled patients with GEJA clinically staged as cT3-4aNanyM0 or cT1-2N+M0 from October 2022 to June 2023. Eligible patients were administered three preoperative and five postoperative 3-week cycles of treatment with PD-1 antibody tislelizumab plus SOX (S-1 and oxaliplatin) regimen. The primary endpoint was major pathological response (MPR) rate. Thirty-two patients were enrolled. The median age was 60 years (range: 28-74 years), and 53.1% (17/32) patients were Siewert III type. All patients received at least one cycle of assigned preoperative treatment, and 93.8% (30/32) patients completed three cycles of assigned preoperative tislelizumab and SOX. The R0 resection rate was 96.9% (31/32). MPR, pathological complete response (pCR) of primary tumors and ypT0N0 rates were 50.0% (16/32, 95% CI: 31.9-68.1%), 28.1% (9/32, 95% CI: 13.7-46.7%) and 25.0% (8/32, 95% CI: 11.5-43.4%), respectively. The surgical morbidity rate was 15.6% (5/32), and no 30-day mortality was observed. In the preoperative and postoperative treatment periods, the rate of treatment-related grade 3-4 adverse events was 31.2% (10/32). At the date of 7th Jan 2025, 8 (25.0%) patients occurred recurrence. Therefore, perioperative tislelizumab plus chemotherapy demonstrated significantly improved pathological regression and might be a promising option for patients with locally advanced resectable GEJA.
Collapse
Affiliation(s)
- Run-Cong Nie
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.
| | - Shu-Qiang Yuan
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Ya Ding
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Yong-Ming Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Yuan-Fang Li
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Cheng-Cai Liang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Mu-Yan Cai
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Guo-Ming Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Xiao-Wei Sun
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - De-Sheng Weng
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Dan-Dan Li
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Jing-Jing Zhao
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Xiao-Jiang Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Yuan-Xiang Guan
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Zhi-Min Liu
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Yao Liang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Ma Luo
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Jun Chi
- Department of Endoscopy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Hai-Bo Qiu
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Zhi-Wei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Xiao-Shi Zhang
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.
| | - Ying-Bo Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.
| |
Collapse
|
41
|
Nakayama I, Nakamura Y, Shitara K. The immunotherapy challenge in locally advanced gastroesophageal cancer: VESTIGE trial's insights and future pathways. Ann Oncol 2025; 36:130-133. [PMID: 39627086 DOI: 10.1016/j.annonc.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 01/31/2025] Open
Affiliation(s)
- I Nakayama
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa City, Chiba, Japan
| | - Y Nakamura
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa City, Chiba, Japan
| | - K Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa City, Chiba, Japan.
| |
Collapse
|
42
|
Tanaka T, Suda K, Nakauchi M, Fujita M, Suzuki K, Umeki Y, Serizawa A, Akimoto S, Watanabe Y, Shibasaki S, Matsuoka H, Inaba K, Uyama I. Safety and feasibility of laparoscopic stomach-partitioning gastrojejunostomy combined with neoadjuvant chemotherapy followed by minimally invasive gastrectomy for resectable gastric cancer with gastric outlet obstruction. Surg Endosc 2025; 39:837-849. [PMID: 39623174 DOI: 10.1007/s00464-024-11427-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/12/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Advanced gastric cancer with gastric outlet obstruction (GOO) causes malnutrition and medication adherence issues, leading to a poor prognosis. We developed a novel multimodal, less invasive treatment approach for gastric cancer patients with symptomatic GOO: laparoscopic stomach-partitioning gastrojejunostomy (LSPGJ) combined with neoadjuvant chemotherapy (NAC), followed by minimally invasive gastrectomy with reuse of gastrojejunostomy. This study is a retrospective analysis of the safety and feasibility of our treatment strategy. METHODS In this single-institution retrospective study, we enrolled 54 patients (NAC group, n = 26; upfront gastrectomy group, n = 28) who achieved R0 resection through a minimally invasive approach between 2007 and 2020 and evaluated their short- and long-term outcomes. RESULTS After LSPGJ, the Gastric Outlet Obstruction Scoring System score significantly improved (p < 0.001). The median relative dose intensity of NAC was 88.2%. Regarding short-term outcomes, there were no differences in postoperative complications, length of postsurgical hospital stay, and adjuvant chemotherapy administration. Although overall survival and relapse-free survival showed trends toward improvement in the NAC group, these differences were not statistically significant. The cumulative incidence curve for recurrence in the NAC group was significantly lower than that of the upfront gastrectomy group (p = 0.041). Recurrence and hematogenous metastasis were significantly lower in the NAC group (p = 0.031 and 0.041, respectively) than in the upfront gastrectomy group. A forest plot revealed that NAC yielded favorable outcomes, particularly for patients with a body mass index (BMI) < 18.5 kg/m2, cT4, or cN1. CONCLUSIONS LSPGJ combined with NAC followed by minimally invasive gastrectomy was a safe and feasible treatment strategy for patients with advanced gastric cancer with symptomatic GOO. This procedure may contribute to the early recovery of oral intake and help maintain NAC dose intensity, potentially improving prognosis, particularly for patients with low BMI and advanced-stage disease.
Collapse
Affiliation(s)
- Tsuyoshi Tanaka
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Koichi Suda
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan.
- Collaborative Laboratory for Research and Development in Advanced Surgical Intelligence, Fujita Health University, Toyoake, Japan.
| | - Masaya Nakauchi
- Department of Advanced Robotic and Endoscopic Surgery, Fujita Health University, Toyoake, Japan
| | - Masahiro Fujita
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Kazumitsu Suzuki
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Yusuke Umeki
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Akiko Serizawa
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Shingo Akimoto
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Yusuke Watanabe
- Department of Advanced Robotic and Endoscopic Surgery, Fujita Health University, Toyoake, Japan
| | - Susumu Shibasaki
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Hiroshi Matsuoka
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Kazuki Inaba
- Department of Advanced Robotic and Endoscopic Surgery, Fujita Health University, Toyoake, Japan
| | - Ichiro Uyama
- Department of Advanced Robotic and Endoscopic Surgery, Fujita Health University, Toyoake, Japan
- Collaborative Laboratory for Research and Development in Advanced Surgical Technology, Fujita Health University, Toyoake, Japan
| |
Collapse
|
43
|
Li C, Tian Y, Zheng Y, Yuan F, Shi Z, Yang L, Chen H, Jiang L, Wang X, Zhao P, Zhang B, Wang Z, Zhao Q, Dong J, Lian C, Xu S, Zhang A, Zheng Z, Wang K, Dang C, Wu D, Chen J, Xue Y, Liang B, Cheng X, Wang Q, Chen L, Xia T, Liu H, Xu D, Zhuang J, Wu T, Zhao X, Wu W, Wang H, Peng J, Hou Z, Zheng R, Chen Y, Yin K, Zhu Z. Pathologic Response of Phase III Study: Perioperative Camrelizumab Plus Rivoceranib and Chemotherapy Versus Chemotherapy for Locally Advanced Gastric Cancer (DRAGON IV/CAP 05). J Clin Oncol 2025; 43:464-474. [PMID: 39383487 PMCID: PMC11776878 DOI: 10.1200/jco.24.00795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/01/2024] [Accepted: 09/05/2024] [Indexed: 10/11/2024] Open
Abstract
PURPOSE This multicenter, randomized phase III trial evaluated the efficacy and safety of perioperative camrelizumab (an anti-PD-1 antibody) plus low-dose rivoceranib (a VEGFR-2 inhibitor) and S-1 and oxaliplatin (SOX) (SOXRC), high-dose rivoceranib plus SOX (SOXR), and SOX alone (SOX) for locally advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma. METHODS Patients with T3-4aN + M0 G/GEJ adenocarcinoma were randomly assigned (1:1:1) to receive perioperative treatment with SOXRC, SOXR, or SOX. The primary end points were pathologic complete response (pCR) and event-free survival. The Independent Data Monitoring Committee recommended stopping enrollment in the SOXR group on the basis of the safety data of the first 103 randomly assigned patients in the three groups. The patients were then randomly assigned 1:1 to the SOXRC or SOX groups. This report presents the pCR results obtained per protocol for the first 360 randomly assigned patients who had the opportunity for surgery in the SOXRC and SOX groups. RESULTS In the SOXRC and SOX groups, of the 180 patients in each group, 99% and 98% of patients received neoadjuvant therapy, 91% and 94% completed planned neoadjuvant therapy, and 86% and 87% underwent surgery, respectively. The pCR was significantly higher in the SOXRC group at 18.3% (95% CI, 13.0 to 24.8) compared with 5.0% (95% CI, 2.3 to 9.3) in the SOX group (difference of 13.7%; 95% CI, 7.2 to 20.1; odds ratio of 4.5 [95% CI, 2.1 to 9.9]). The one-sided P value was <.0001, crossing the prespecified statistical significance threshold of P = .005. Surgical complications and grade ≥3 neoadjuvant treatment-related adverse events were 27% versus 33% and 34% versus 17% for SOXRC and SOX, respectively. CONCLUSION The SOXRC regimen significantly improved pCR compared with SOX alone in patients with G/GEJ adenocarcinoma with a tolerable safety profile.
Collapse
Affiliation(s)
- Chen Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yantao Tian
- Pancreatogastric Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Yanan Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zheng Shi
- Gastrointestinal Surgery, Shanghai Changhai Hospital, Shanghai, China
| | - Lin Yang
- Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Hao Chen
- Oncological Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Lixin Jiang
- Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Yantai, China
| | - Xixun Wang
- Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Yantai, China
| | - Ping Zhao
- Gastrointestinal Surgery, Sichuan Provincial Cancer Hospital, Chengdu, China
| | - Benyan Zhang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zhenqiang Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Qun Zhao
- Gastrointestinal Oncology Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianhong Dong
- Minimally Invasive Surgery of Digestive Endoscopy, Shanxi Provincial Cancer Hospital, Taiyuan, China
| | - Changhong Lian
- Gastrointestinal Surgery, HePing Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Sanrong Xu
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Aimin Zhang
- Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Zhichao Zheng
- Gastric Cancer Surgery, Liaoning Cancer Hospital & Institution, Shenyang, China
| | - Kang Wang
- Gastrointestinal Surgery, Sichuan Provincial People's Hospital, Chengdu, China
| | - Chengxue Dang
- Surgical Oncology, The First Affiliated Hospital of XI'AN Jiaotong University, Xi'an, China
| | - Dan Wu
- Gastrointestinal Surgery, The Second Affiliated Hospital Zhejiang University, School of Medicine, Hangzhou, China
| | - Jian Chen
- Gastrointestinal Surgery, The Second Affiliated Hospital Zhejiang University, School of Medicine, Hangzhou, China
| | - Yingwei Xue
- Gastrointestinal Surgery, Harbin Medical University Affiliated Cancer Hospital, Harbin, China
| | - Bo Liang
- General Surgery, Jilin Guowen Hospital, Changchun, China
| | | | - Qian Wang
- Gastrointestinal Surgery, The Affiliated Hospital of GuiZhou Medical University, Guiyang, China
| | - Luchuan Chen
- Gastrointestinal Cancer Surgery Department, Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Tao Xia
- Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Heli Liu
- Gastrointestinal Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Dazhi Xu
- Gastrosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jing Zhuang
- General Surgery, Henan Cancer Hospital, Zhengzhou, China
| | - Tao Wu
- General Surgery, Tangdu Hospital of the Fourth Military Medical, Xi'an, China
| | - Xi Zhao
- Colorectal Surgery, Hainan Cancer Hospital, Haikou, China
| | - Wei Wu
- Geriatric Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Hongzhi Wang
- Center of Gastrointestinal Tumor, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Junsheng Peng
- Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiguo Hou
- Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | | | - Yuting Chen
- Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - Kai Yin
- Gastrointestinal Surgery, Shanghai Changhai Hospital, Shanghai, China
| | - Zhenggang Zhu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
44
|
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
|
45
|
Srikumar T, Sundar R. Multimodality Treatment for Locally Advanced Gastric Adenocarcinoma. Surg Clin North Am 2025; 105:75-94. [PMID: 39523078 DOI: 10.1016/j.suc.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Gastric cancer is prevalent worldwide and is a leading cause of cancer-related death. Patients with GC often present at advanced stages at diagnosis. Patients with locally advanced diseases experience poor survival rates with surgery alone. Multimodality therapy, including peri-operative therapy and adjuvant therapy, has improved outcomes. However, there is no consensus on the optimal treatment approach. Molecular characteristics of GC may help guide treatment choices and studies are currently underway to evaluate other treatment modalities including immunotherapy and targeted therapy.
Collapse
Affiliation(s)
- Thejal Srikumar
- Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
| | - Raghav Sundar
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore.
| |
Collapse
|
46
|
Zhong B, Xiong Z, Zheng J, Mohamed SA, Sun J, Huang D, Deng Z, Guo J, Peng J, Wang H, Lian L. Impact of Helicobacter pylori infection on neoadjuvant chemotherapy in locally advanced gastric cancer: a retrospective analysis. BMC Cancer 2025; 25:157. [PMID: 39871229 PMCID: PMC11773845 DOI: 10.1186/s12885-025-13494-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 01/12/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection may affect the efficacy of immunotherapy and adjuvant chemotherapy in gastric cancer patients. However, the role of H. pylori infection in neoadjuvant chemotherapy in patients with locally advanced gastric cancer (LAGC) remains unclear. This study investigated the effect of H. pylori infection on neoadjuvant chemotherapy and prognosis of patients with LAGC. METHODS This retrospective study utilized data from patients with LAGC who underwent neoadjuvant chemotherapy and surgical treatment at the Sixth Affiliated Hospital of Sun Yat-sen University from January 1, 2010, to January 31, 2021. Patients were grouped according to their H. pylori infection status. The responses of the two groups to neoadjuvant chemotherapy and oncological outcomes were then compared. RESULTS A total of 239 patients were included in the analysis, and the baseline characteristics of the H. pylori-positive (n = 51) and H. pylori-negative (n = 188) groups were comparable. Further analysis revealed that H. pylori infection was significantly associated with the major pathological response (P = 0.009). Multivariate analysis showed that factors related to major pathological response included; age ≤ 50 (OR: 0.423, 95% CI: 0.194-0.925), H. pylori infection (OR: 0.396, 95% CI: 0.183-0.854), pathological stage T 3/4 (OR: 0.524, 95% CI: 0.288-0.954), and CA12-5 > 35 U/mL (OR: 0.345, 95% CI: 0.132-0.904). Both overall survival (OS) and disease-free survival (DFS) rates were poorer in the H. pylori-positive group than in the H. pylori-negative group (OS: Log-Rank P = 0.035; DFS: Log-Rank P = 0.029). CONCLUSION This cohort study indicated that H. pylori infection may be associated with tumor response to neoadjuvant chemotherapy and survival outcomes in patients with LAGC.
Collapse
Affiliation(s)
- Bin Zhong
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhizhong Xiong
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiabo Zheng
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Saddam Ahmed Mohamed
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiachen Sun
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dayin Huang
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zijian Deng
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianping Guo
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junsheng Peng
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Huashe Wang
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Lei Lian
- Department of General Surgery (Department of Gastrointestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
47
|
Cao B, Zhang P, Shi Z. Association of Body Composition Parameters with the Short- and Long-Term Efficacy of Neoadjuvant Immunotherapy Combined with Chemotherapy in Advanced Gastric Cancer. Nutr Cancer 2025; 77:455-464. [PMID: 39865651 DOI: 10.1080/01635581.2025.2455762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Immunotherapy has become a prevalent strategy in the neoadjuvant treatment of advanced gastric cancer (AGC). This study investigates the predictive value of computed tomography (CT)-derived body composition parameters on the efficacy of neoadjuvant immunotherapy for AGC. METHODS Data on 103 patients with resectable AGC who received neoadjuvant immunotherapy combined with chemotherapy at a teaching hospital between March 2020 and August 2022 were collected. Body composition parameters, including the subcutaneous adipose index (SAI), visceral adipose index (VAI), and skeletal muscle index (SMI), were calculated from pretreatment CT images. Logistic regression and Cox proportional hazards models assessed the impact of these parameters on pathological responses and survival outcomes following treatment. RESULTS Of the patients, 34 (33.0%) achieved a major pathological response (MPR). Higher SAI, VAI, and SMI values were significantly linked to an increased likelihood of achieving MPR (p < 0.05). Multivariate regression analysis revealed that only SAI independently predicted MPR (OR 1.042, 95% CI 1.009-1.077, p = 0.013). Furthermore, patients with a high SAI had significantly improved 2-year overall survival (76.9% vs. 54.9%, log-rank p = 0.012) and 2-year event-free survival (71.2% vs. 51.0%, log-rank p = 0.022) compared to those with low SAI. The survival benefit associated with high SAI was partly due to its higher MPR rate (mediating proportion: 37.5%, 95% CI: 12%-110%). CONCLUSION Pretreatment SAI independently correlates with MPR and better oncological outcomes in patients with AGC receiving neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Bingyan Cao
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Xingtai Medical College, Xingtai, China
| | - Peifang Zhang
- Outpatient Department, The Second Affiliated Hospital of Xingtai Medical College, Xingtai, China
| | - Zhanying Shi
- Department of Internal Medicine, Xingtai Medical College, Xingtai, China
| |
Collapse
|
48
|
Sabbagh S, Jabbal IS, Herrán M, Mohanna M, Iska S, Itani M, Dominguez B, Sarna K, Nahleh Z, Nagarajan A. Evaluating survival outcomes and treatment recommendations in resectable gastric cancer. Sci Rep 2025; 15:2816. [PMID: 39843914 PMCID: PMC11754744 DOI: 10.1038/s41598-024-82807-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
No consensus exists on the optimal therapy for resectable gastric cancer (GC) and gastroesophageal junction (GEJ) tumors, including the effectiveness of chemoradiotherapy versus perioperative chemotherapy (PC). Our study aimed to compare overall survival (OS) outcomes associated with the recommended treatment modalities for GC and GEJ tumors and evaluate treatment trends from 2010 to 2020. A national registry cohort identified patients with ≥ cT2 nonmetastatic GC and GEJ cancer. Treatment modalities were classified as neoadjuvant chemotherapy (NC), neoadjuvant chemoradiotherapy (NCR), PC, adjuvant chemotherapy (AC), and adjuvant chemoradiation (ACR). Kaplan-Meier curve and multivariable Cox regression models evaluated factors associated with OS. A cohort of 7665 patients were included. Patients who received PC had the highest OS (median 86.80 months, 95% CI 73.40-NE), while chemoradiotherapy in the neoadjuvant and adjuvant settings had worse OS than PC and NC (NCR median 47.15 months, 95% CI 44.58-52.27, and ACR median 52.67 months, 95%CI 42.78-63.93). The Cox proportional hazards model showed that NCR and NC had worse survival than PC (HR 1.74, 95% CI 1.50-2.02, p < 0.001 and HR 1.26, 95% CI 1.10-1.44, p = 0.0008, respectively). Additionally, the most utilized modality during 2020 was NC (35.8%), followed by PC (28.0%) and NCR (24.9%). The utilization of PC and NC had the most substantial rise between 2010 and 2020, increasing by 11.0%. The study demonstrates the association of PC with improved OS outcomes for nonmetastatic GC and GEJ tumors. Therapies combining radiation with chemotherapy and extended lymph node dissection correlated with a worse prognosis compared to PC and NC. Despite the association with improved outcomes, national data reveals low utilization rates for PC.
Collapse
Affiliation(s)
- Saad Sabbagh
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, 33331, USA
| | - Iktej Singh Jabbal
- Department of Internal Medicine, Advent Health Sebring, Sebring, FL, USA
| | - María Herrán
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, 33331, USA
| | - Mohamed Mohanna
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, 33331, USA
| | - Sindu Iska
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, 33331, USA
| | - Mira Itani
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, 33331, USA
| | - Barbara Dominguez
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, 33331, USA
| | - Kaylee Sarna
- Department of Clinical Research, Cleveland Clinic Florida, Weston, FL, USA
| | - Zeina Nahleh
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, 33331, USA
| | - Arun Nagarajan
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, 33331, USA.
| |
Collapse
|
49
|
de Moraes FCA, Sano VKT, Silva BL, Silva ALS, Castro SCR, Kreuz M, Fernandes LR, Kelly FA, Burbano RMR. PD-1/PD-L1 Inhibitors Increase Pathological Complete Response in Locally Advanced Gastric Cancer: A Meta-analysis and Trial Sequential Analysis. J Gastrointest Cancer 2025; 56:49. [PMID: 39833372 DOI: 10.1007/s12029-024-01141-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND AND OBJECTIVE Gastric cancer (GC) remains a leading cause of morbidity and mortality worldwide. The current standard of care involves neoadjuvant chemotherapy (NACT) followed by radical gastrectomy. This study aims to evaluate the efficacy of neoadjuvant therapy with PD-1/PD-L1 inhibitors in comparison to chemotherapy alone for patients with locally advanced gastric cancer (LAGC). METHODS We conducted a systematic search of PubMed, Web of Science, and Embase to identify studies examining the addition of PD-1/PD-L1 inhibitors to neoadjuvant therapy for LAGC. Odds ratios (OR) were calculated for binary outcomes, such as pathological complete response (pCR), with corresponding 95% confidence intervals (CI). RESULTS Seven studies were included, encompassing a total of 1772 patients. Baseline median age ranged from 31 to 75 years. Most patients had an ECOG performance status score of 0 (942 patients), while 294 had an ECOG score of 1. The estimated pCR (OR 5.94, 95% CI 3.98-8.87; p < 0.000001) significantly favored the PD-1/PD-L1 inhibitors combined with chemotherapy over chemotherapy alone. Additionally, the incidence of certain adverse events increased significantly in the intervention group, including any-grade hypothyroidism (OR 4.55, 95% CI 2.27-9.10; p = 0.000019) and rash (OR 1.74, 95% CI 1.10-2.76; p = 0.017). Conversely, the control group showed a statistically significant lower incidence of grade ≥ 3 fatigue (OR 2.80, 95% CI 1.15-6.85; p = 0.024) compared to the intervention group. CONCLUSION This systematic review and meta-analysis indicate that the addition of PD-1/PD-L1 inhibitors to neoadjuvant chemotherapy is associated with a higher pathological complete response rate compared to chemotherapy alone in patients with locally advanced gastric cancer.
Collapse
Affiliation(s)
| | | | - Barbara Lins Silva
- Vancouver Island Health Authority, 1947 Cook St, Victoria, BC, V8T 3P7, Canada
| | | | | | - Michele Kreuz
- Lutheran University of Brazil, Rio Grande Do Sul, Canoas, 92425-020, Brazil
| | | | | | | |
Collapse
|
50
|
Shimonosono M, Arigami T, Matsushita D, Tsuruda Y, Sasaki K, Baba K, Ohtsuka T. The impact of delayed adjuvant chemotherapy on survival in gastric cancer patients with and without preoperative chemotherapy. Ann Gastroenterol Surg 2025. [DOI: 10.1002/ags3.12911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/03/2025] [Indexed: 02/11/2025] Open
Abstract
AbstractAimAdjuvant chemotherapy (AC) is the standard treatment for patients with advanced gastric cancer (GC), yet the optimal timing for its initiation remains unclear. Besides, no studies have definitively established when AC should begin in patients receiving preoperative chemotherapy (PC). This study aimed to determine the optimal timing for initiating AC in patients with GC who underwent curative gastrectomy, either with or without PC.MethodsA total of 446 patients who underwent curative gastrectomy were evaluated, including 140 who received AC: 72 without PC and 68 with PC. Patients were categorized into two groups based on when they began AC: the early initiation group (within 8 weeks post‐surgery), and the late initiation group (8 weeks or later post‐surgery).ResultsIn the non‐PC cohort, the 3‐year relapse‐free survival (RFS) rates were 71% in the early group versus 56% in the late group (p = 0.49), while the 3‐year overall survival (OS) rates were 94% versus 73% (p = 0.003). Similar trends were observed in the PC cohort; the 3‐year RFS rates were 59% versus 19% (p = 0.002), and the 3‐year OS rates were 69% versus 48% (p = 0.02). Multivariate analysis identified pretherapeutic distant metastasis (p < 0.001) and delayed AC initiation (≥8 weeks) (p = 0.001) as independent predictors of worse prognosis.ConclusionDelayed initiation of AC is associated with significantly poorer postoperative survival in patients with GC, irrespective of whether PC was administered. These findings emphasize the importance of timely AC initiation to improve long‐term outcomes in this patient population.
Collapse
Affiliation(s)
- Masataka Shimonosono
- Department of Digestive Surgery Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Takaaki Arigami
- Department of Digestive Surgery Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Daisuke Matsushita
- Department of Digestive Surgery Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Yusuke Tsuruda
- Department of Digestive Surgery Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Ken Sasaki
- Department of Digestive Surgery Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Kenji Baba
- Department of Digestive Surgery Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Takao Ohtsuka
- Department of Digestive Surgery Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| |
Collapse
|