1
|
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
|
2
|
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
|
3
|
Piao H, Wenping L, Chengde L, Haoming L, Xiaohan Z, Yingdi L, Xuezheng Z, Shumei M. The efficacy and safety of PD-1/PD-L1 inhibitors plus chemotherapy versus chemotherapy alone in advanced or metastatic gastric or gastroesophageal junction cancer: a meta-analysis of randomized controlled trials. Eur J Clin Pharmacol 2025:10.1007/s00228-025-03861-3. [PMID: 40493058 DOI: 10.1007/s00228-025-03861-3] [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: 02/15/2025] [Accepted: 05/31/2025] [Indexed: 06/12/2025]
Abstract
BACKGROUND The combination chemotherapy of alpha-PD-1/PD-L1 has become the standard treatment option for some cancer patients. However, studies have shown that not all patients benefit from improved survival rates, especially the use of PD-1/PD-L1 inhibitors in combination with chemotherapy for progression-free survival (PFS) in patients with gastric or gastroesophageal cancer (GC/GEJC) remains highly controversial. To address this issue, we conducted a meta-analysis of randomized controlled trials (RCTs) aimed at comparing the efficacy of PD-1/PD-L1 inhibitors combined with chemotherapy versus chemotherapy in GC/GEJC patients. METHOD By searching relevant databases, RCTs published up to November 2024 were collected, and the hazard ratios (HR) and 95% confidence intervals (CI) of overall survival (OS) and PFS were calculated. Meanwhile, the odds ratios (OR) and 95% CI of treatment-related adverse events (TRAEs) were evaluated. RESULT A total of 6842 patients were included in seven trials. In the summary analysis of OS, compared with the chemotherapy group, the PD-1/PD-L1 inhibitor combined with the chemotherapy group showed significant improvement in OS (HR = 0.80; 95% CI = 0.76-0.85; p < 0.0001) and PFS (HR = 0.86; 95% CI = 0.71-0.81; p < 0.0001). Additionally, there were significant differences in the incidence of TRAEs (OR = 1.59; 95% CI = 1.21-2.02; p = 0.0001) and grade 3-4 TRAEs (OR = 1.43; 95% CI = 1.30-1.58; p < 0.0001). CONCLUSION When compared to chemotherapy, the combination of PD-1/PD-L1 inhibitors with chemotherapy improves survival but with higher toxicity risks, requiring careful benefit-risk evaluation in clinical practice.
Collapse
Affiliation(s)
- Hu Piao
- Pharmacy College, Shandong Second Medical University, Weifang, China
| | - Li Wenping
- Pharmacy College, Shandong Second Medical University, Weifang, China
| | - Li Chengde
- Department of Clinical Pharmacy, Shandong Second Medical University, Weifang, China
| | - Li Haoming
- Clinical College of Shandong Second Medical University, Weifang, China
| | - Zhang Xiaohan
- Pharmacy College, Shandong Second Medical University, Weifang, China
| | - Liu Yingdi
- Pharmacy College, Shandong Second Medical University, Weifang, China
| | - Zhang Xuezheng
- Department of Health Care Medicine, Weifang Hospital of Traditional Chinese Medicine, Weifang, China.
| | - Mao Shumei
- Department of Pharmacology, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
4
|
Qian J, Tu X, Chen J, Chen S, Zhou Y, Sun C, Wu Z. Two decades of progress in gastric cancer peritoneal metastasis: a bibliometric perspective on molecular mechanisms and therapeutic innovations. Front Oncol 2025; 15:1583364. [PMID: 40519309 PMCID: PMC12162253 DOI: 10.3389/fonc.2025.1583364] [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: 02/25/2025] [Accepted: 05/12/2025] [Indexed: 06/18/2025] Open
Abstract
Background Gastric cancer (GC) is the fifth most common malignant tumor worldwide. The peritoneum is a common site of metastasis in advanced GC, and patients with gastric cancer peritoneal metastasis (GCPM) have a very low 5-year survival rate. Systemic therapy has limited efficacy for peritoneal metastases, and early diagnosis is difficult. In this paper, we analyzed the GCPM-related literature by bibliometric methods, aiming to identify the research hotspots and trends and to provide a basis for clinical practice and research planning. Methods Based on the Web of Science Core Collection database (WoSCC), we screened the GCPM-related literature published from 2004 to 2024. Countries, institutions, authors, journals, and keywords were analyzed and visualized by tools such as CiteSpace, VOSviewer, Scimago Graphica, RStudio, and the Bibliometrix package. Result A total of 2416 publications were included in this study. The growth rate of GCPM publications is positive until 2021, with a slowdown in the near future. Japan dominated the research output (842 publications), followed by China (748 publications) and the United States (268 publications). Japanese-affiliated organizations and researchers are extremely productive in the field of GCPM. The most frequently cited document was Japanese gastric cancer treatment guidelines 2014 (ver. 4) (citations = 2076). Research focuses on four major clusters: (1) molecular mechanisms of GCPM; (2) prognosis of GCPM; (3) chemotherapy of GCPM; and (4) intraperitoneal treatment of GCPM. Emerging trends include key pathways of GCPM, artificial intelligence (AI) and multi-omics-driven early diagnosis, novel intraperitoneal therapeutic modalities, and immunologic/targeted drugs. Conclusion Japan is a leader in GCPM research. Recently, the focus of GCPM research has shifted from basic treatment to precision and personalized treatment through the integration of molecular mechanisms, novel intraperitoneal therapeutic modalities, and AI technologies. Current challenges include the lack of standardized validation systems for emerging technologies and regional differences in clinical practice. In the future, there is a need to promote global collaborative trials and optimization of multimodality therapy. The results of this study provide a key direction and systematic basis for future exploration of GCPM.
Collapse
Affiliation(s)
- Jin Qian
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoxuan Tu
- Department of Oncology, Zhejiang Hospital, Hangzhou, China
| | - Jianglin Chen
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Sisi Chen
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Zhou
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuan Sun
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, Hangzhou, China
| | - Zhibing Wu
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, Hangzhou, China
- Department of Radiation Oncology, Zhejiang Hospital, Hangzhou, China
- Department of Oncology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Lu YM, Jin P, Wang HK, Shao XX, Hu HT, Jiang YJ, Li WY, Tian YT. Proximal gastrectomy with tubular stomach reconstruction vs total gastrectomy for proximal gastric cancer following neoadjuvant chemotherapy: A multicenter retrospective study. World J Gastrointest Surg 2025; 17:107579. [DOI: 10.4240/wjgs.v17.i5.107579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/08/2025] [Accepted: 04/25/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a major global health challenge, and the treatment of proximal GC in particular presents unique clinical and surgical complexities. Currently, there is no consensus on whether proximal gastrectomy (PG) or total gastrectomy (TG) should be used for advanced proximal GC, and the choice of postoperative gastrointestinal reconstruction method remains controversial.
AIM To compare the short-term efficacy, long-term survival, and postoperative reflux outcomes of PG with tubular stomach reconstruction vs TG with Roux-en-Y reconstruction in patients with proximal GC following neoadjuvant chemotherapy (NACT) in an effort to provide valuable insights for clinical decision-making regarding the optimal surgical approach.
METHODS A multicenter retrospective cohort study was conducted at two Chinese medical centers between December, 2012 and December, 2022. Patients with histologically confirmed proximal GC who received NACT followed by either PG with tubular stomach reconstruction or TG with Roux-en-Y reconstruction were included. Propensity score matching (PSM) was performed to balance baseline characteristics, and the primary endpoint was 5-year overall survival (OS). Secondary endpoints included recurrence-free survival (RFS), postoperative complications, and reflux severity.
RESULTS After PSM, 244 patients (122 PG, 122 TG) were finally included and all baseline characteristics were comparable between groups. The PG group had a significantly shorter operation time compared to the TG group (189.50 vs 215.00 minutes, P < 0.001), with no differences in intraoperative blood loss or postoperative complications (19.68% vs 14.75%, P = 0.792). The 5-year OS rates were 52.7% vs 45.5% (P = 0.330), and 5-year RFS rates were 54.3% vs 47.6% (P = 0.356) for the PG and TG groups, respectively. Reflux symptoms (18.0% vs 31.1%, P = 0.017) and clinically significant reflux based on gastroesophageal reflux disease questionnaire scores ≥ 8 (7.4% vs 21.3%, P < 0.001) were significantly less frequent in the PG group. Multivariate analysis identified histological differentiation (HR = 2.98, 95%CI: 2.03-4.36, P < 0.001) and tumor size (HR = 0.26, 95%CI: 0.17-0.41 for tumors ≤ 4 cm, P < 0.001) as independent prognostic factors.
CONCLUSION PG with tubular stomach reconstruction is comparable to TG in terms of surgical safety and long-term oncological outcomes for proximal GC patients following NACT. Additionally, PG has the advantages of shorter operation time and lower rates of postoperative reflux, suggesting potential benefits for patient quality of life. Notably, the analysis of postoperative prognostic factors, including histological differentiation and tumor size, further informs clinical decision-making and highlights the importance of individualized treatment strategies.
Collapse
Affiliation(s)
- Yi-Ming 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
| | - Peng Jin
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Hai-Kuo 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
| | - Xin-Xin 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
| | - Hai-Tao Hu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu-Juan Jiang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wang-Yao Li
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan-Tao Tian
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
6
|
Emiloju O, Miao R, Alese O. ASO Author Reflections: The Evolving Role of Immunotherapy for Gastro-Esophageal Malignancies. Ann Surg Oncol 2025:10.1245/s10434-025-17493-5. [PMID: 40402421 DOI: 10.1245/s10434-025-17493-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/23/2025]
Affiliation(s)
| | - Ruoyu Miao
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Olatunji Alese
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
7
|
Nakayama I. Therapeutic strategy for scirrhous type gastric cancer. Jpn J Clin Oncol 2025:hyaf081. [PMID: 40403741 DOI: 10.1093/jjco/hyaf081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
Scirrhous-type gastric cancer (SGC) is a rare but well-recognized subset of resectable gastric cancer (GC), accounting for ⁓10% of cases. Despite its long history of clinical recognition dating back to the pre-1900s, SGC remains one of the most challenging GC subtypes to treat. Traditionally, SGC has been clinically defined as Borrmann type 4 GC, with histological classifications such as signet ring cell carcinoma or diffuse-type histology serving as alternative diagnostic criteria. Therapeutic advancements for SGC have largely focused on locally advanced or oligometastatic disease, yet no SGC-specific treatment has been established. The phase III JCOG0501 trial failed to demonstrate a survival benefit of neoadjuvant S-1 plus cisplatin for Borrmann type 4 and large type 3 GC. Recent developments in biomarker-driven therapies may redefine SGC by molecular subtypes, with CLDN18.2-targeted therapy emerging as a potential option for some SGC cases. However, as the landscape of medical oncology evolves, SGC may not remain a distinct therapeutic entity. The focus should shift toward understanding the intrinsic biology of SGC. Treatment development for SGC is expected to continue advancing, becoming increasingly stratified based on molecular abnormalities while maintaining a commitment to addressing unmet needs, such as early-onset GC and GC with symptomatic peritoneal dissemination.
Collapse
Affiliation(s)
- Izuma Nakayama
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwanoha 6-5-1, Kashiwa, 277-8577, Japan
| |
Collapse
|
8
|
Tumour plasticity and immune dynamics promote resistance to neo-adjuvant treatment. NATURE CANCER 2025:10.1038/s43018-025-00954-x. [PMID: 40405021 DOI: 10.1038/s43018-025-00954-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
|
9
|
Walch HS, Borpatragohain R, Jee J, Chatila W, Fong C, Maron SB, Ku GY, Ilson DH, Janjigian YY, Wu AJ, Shah P, Coit DG, Bains MS, Rusch VW, Park BJ, Bott MJ, Gray K, Jones DR, Berger M, Schultz N, Strong VE, Molena D, Sihag S. Clinical Implications of The Cancer Genome Atlas Molecular Classification System in Esophagogastric Cancer. Clin Cancer Res 2025; 31:1912-1921. [PMID: 40299774 DOI: 10.1158/1078-0432.ccr-24-3473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/03/2024] [Accepted: 03/12/2025] [Indexed: 05/01/2025]
Abstract
PURPOSE The Cancer Genome Atlas (TCGA) project defined four distinct molecular subtypes of esophagogastric adenocarcinoma: microsatellite instable (MSI), Epstein-Barr virus (EBV)-associated, genomically stable (GS), and chromosomally instable (CIN). However, an association between molecular subtypes and clinical outcomes has not been clearly demonstrated. Given few actionable biomarkers, we investigated the clinical relevance of TCGA classification system. EXPERIMENTAL DESIGN We identified all patients with esophagogastric adenocarcinoma whose tumors underwent prospective next-generation sequencing using the Memorial Sloan Kettering-IMPACT assay from 2014 to 2023. We classified all tumors in accordance with TCGA methodology and correlated molecular subtypes with high-quality clinicopathologic data. RESULTS Among 1,438 included patients, 941 had CIN, 344 had GS, 103 had MSI, and 50 had EBV tumors. Accounting for the clinical stage and tumor grade, molecular classification was independently associated with overall cancer-specific survival (P < 0.001) on Cox multivariable analysis. Furthermore, genomic signatures, patient demographics, pathologic responses to neoadjuvant therapy, patterns of recurrence, and metastatic organotropism differed significantly by molecular subtype. Although most distal esophageal and gastroesophageal junction tumors were CIN, up to 25% of these included GS, MSI, or EBV subtypes in contrast to TCGA. Random forest machine learning demonstrated that the molecular subtype is more influential in predicting response to treatment than tumor location. CONCLUSIONS Molecular classification is independently prognostic and may warrant inclusion in future staging and treatment guidelines. Routine molecular profiling is clinically feasible and may play a role in the management of patients to help guide appropriate treatment selection and clinical trial enrollment in the place of tumor location.
Collapse
Affiliation(s)
- Henry S Walch
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Raktim Borpatragohain
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin Jee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Waleed Chatila
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christopher Fong
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven B Maron
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Geoffrey Y Ku
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David H Ilson
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yelena Y Janjigian
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Abraham J Wu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pari Shah
- Gastroenterology, Hepatology, and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniel G Coit
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Manjit S Bains
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Valerie W Rusch
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bernard J Park
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew J Bott
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katherine Gray
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Berger
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vivian E Strong
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Molena
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Smita Sihag
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
10
|
Emiloju O, Miao R, Alese O. The Evolving Role of Immunotherapy for Gastroesophageal Malignancies. Ann Surg Oncol 2025:10.1245/s10434-025-17386-7. [PMID: 40332652 DOI: 10.1245/s10434-025-17386-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025]
Abstract
The incorporation of immunotherapy has transformed the treatment landscape for advanced, unresectable, or metastatic gastroesophageal cancers (GECs), with improved survival outcomes. These improvements in outcomes for advanced GECs have led to clinical trials evaluating the role of immunotherapy in patients with resectable early-stage GECs. However, there remains a high burden of morbidity and mortality, and ongoing trials utilizing novel immunotherapy agents and combinations are underway. This review summarizes the findings of previous and ongoing clinical trials related to immunotherapy for patients with early- and late-stage GECs.
Collapse
Affiliation(s)
| | - Ruoyu Miao
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Olatunji Alese
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
11
|
Barroux M, Househam J, Lakatos E, Ronel T, Baker AM, Salié H, Mossner M, Smith K, Kimberley C, Nowinski S, Berner A, Gunasri V, Borgmann M, Liffers S, Jansen M, Caravagna G, Steiger K, Slotta-Huspenina J, Weichert W, Zapata L, Giota E, Lorenzen S, Alberstmeier M, Chain B, Friess H, Bengsch B, Schmid RM, Siveke JT, Quante M, Graham TA. Evolutionary and immune microenvironment dynamics during neoadjuvant treatment of esophageal adenocarcinoma. NATURE CANCER 2025; 6:820-837. [PMID: 40369175 PMCID: PMC12122370 DOI: 10.1038/s43018-025-00955-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] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 03/21/2025] [Indexed: 05/16/2025]
Abstract
Locally advanced esophageal adenocarcinoma remains difficult to treat and the ecological and evolutionary dynamics responsible for resistance and recurrence are incompletely understood. Here, we performed longitudinal multiomic analysis of patients with esophageal adenocarcinoma in the MEMORI trial. Multi-region multi-timepoint whole-exome and paired transcriptome sequencing was performed on 27 patients before, during and after neoadjuvant treatment. We found major transcriptomic changes during treatment with upregulation of immune, stromal and oncogenic pathways. Genetic data revealed that clonal sweeps through treatment were rare. Imaging mass cytometry and T cell receptor sequencing revealed remodeling of the tumor microenvironment during treatment. The presence of genetic immune escape, a less-cytotoxic T cell phenotype and a lack of clonal T cell expansions were linked to poor treatment response. In summary, there were widespread transcriptional and environmental changes through treatment, with limited clonal replacement, suggestive of phenotypic plasticity.
Collapse
Affiliation(s)
- Melissa Barroux
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK.
- Medical Clinic and Polyclinic II, TUM University Hospital, Klinikum rechts der Isar, Munich, Germany.
- German Cancer Consortium (DKTK) Heidelberg, Germany, Partner Site Munich, Munich, Germany.
| | - Jacob Househam
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Data Science Team, The Institute of Cancer Research, London, UK
| | - Eszter Lakatos
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Tahel Ronel
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Division of Infection and Immunity, University College London, London, UK
| | - Ann-Marie Baker
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Henrike Salié
- Clinic for Internal Medicine II, University Medical Center Freiburg, Freiburg, Germany
| | - Maximilian Mossner
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Kane Smith
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Chris Kimberley
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Salpie Nowinski
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Alison Berner
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Vinaya Gunasri
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Department of Pathology, UCL Cancer Institute, University College London, London, UK
| | - Martin Borgmann
- Clinic for Internal Medicine II, University Medical Center Freiburg, Freiburg, Germany
| | - Sven Liffers
- Bridge Institute of Experimental Tumor Therapy (BIT), Division of Solid Tumor Translational Oncology (DKTK) and Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Essen, Germany
| | - Marnix Jansen
- Department of Pathology, UCL Cancer Institute, University College London, London, UK
| | - Giulio Caravagna
- Department of Mathematics, Informatics and Geosciences, University of Triest, Triest, Italy
| | - Katja Steiger
- iBioTUM - Tissue, Institute of Pathology, School of Medicine, TUM, Munich, Germany
| | - Julia Slotta-Huspenina
- Institute of Pathology, Technical University of Munich, Munich, Germany
- Department of Nephrology, School of Medicine, Technical University Munich, Munich, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Luis Zapata
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Eleftheria Giota
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Sylvie Lorenzen
- Department of Internal Medicine III (Haematology/Medical Oncology), Technical University of Munich Hospital Rechts der Isar, Munich, Germany
| | - Markus Alberstmeier
- Department of General, Visceral and Transplantation Surgery, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Benny Chain
- Division of Infection and Immunity, University College London, London, UK
| | - Helmut Friess
- Department of Surgery, TUM University Hospital, rechts der Isar, School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Bertram Bengsch
- Clinic for Internal Medicine II, University Medical Center Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) Heidelberg, Germany, Partner Site Freiburg, Freiburg, Germany
| | - Roland M Schmid
- Medical Clinic and Polyclinic II, TUM University Hospital, Klinikum rechts der Isar, Munich, Germany
- German Cancer Consortium (DKTK) Heidelberg, Germany, Partner Site Munich, Munich, Germany
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy (BIT), Division of Solid Tumor Translational Oncology (DKTK) and Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Essen, Germany
| | - Michael Quante
- Medical Clinic and Polyclinic II, TUM University Hospital, Klinikum rechts der Isar, Munich, Germany
- Clinic for Internal Medicine II, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) Heidelberg, Germany, Partner Site Freiburg, Freiburg, Germany
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK.
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| |
Collapse
|
12
|
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
|
13
|
Zhang G, Zhang L, Feng Q, Ma P, Zheng C, Wang L, Xue Q, Li Y. Outcomes of Intraoperative Radiotherapy for Locally Advanced Adenocarcinoma of the Esophagogastric Junction After Neoadjuvant Therapy: A Single-Arm, Phase 1 Trial From the Chinese National Cancer Center. Ann Surg Oncol 2025; 32:3138-3146. [PMID: 39648241 DOI: 10.1245/s10434-024-16620-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: 08/20/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND The role of intraoperative radiotherapy (IORT) for adenocarcinoma of the esophagogastric junction (AEG) remains uncertain. Therefore, a prospective phase 1 trial was conducted to assess the safety and feasibility of IORT for locally advanced AEG. METHODS The study enrolled patients with AEG at stages II-IVA from January 2019 to September 2019. Eligible patients received esophagectomy and a single fraction of electron beam radiotherapy. The primary endpoint of the study was a safety profile for IORT. Additionally, survival outcomes and the locoregional recurrence rate (LRR) were compared between the non-IORT and IORT cohorts using propensity score-matching. RESULTS For 15 (93.8 %) of the 16 patients in the study, R0 resection was successfully achieved, with only one patient undergoing R1 resection. A total postoperative complication morbidity rate of 43.8 % (7/16) was observed, with major complications (Clavien-Dindo classification ≥3) in 12.5 % of the cases (2/16). Total treatment-related adverse events were reported for seven patients (43.8 %, 7/16). After matching, a lower LRR was observed in the IORT group than in the non-IORT group (0 % [0/12] vs 33.3 % [4/12]; p = 0.028). However, the two groups did not differ significantly in 3-year progression-free survival (PFS: IORT [50.9 %] vs non-IORT [53.4 %]; p = 0.93) or 3-year overall survival (OS: IORT [58.3 %] vs IORT [72.9 %]; p = 0.23). CONCLUSIONS The current study demonstrated favorable feasibility and safety of IORT for locally advanced AEG. Although IORT is beneficial for improving local control, it may not prolong PFS or OS for patients with locally advanced AEG. A phase 2 trial is warranted for further validation of these outcomes.
Collapse
Affiliation(s)
- Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Long Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qinfu Feng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pan Ma
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lide Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
14
|
Xu Q, Chu J, Hu Q, Sun Y, Jiang F, Li S, Liu L. The role and clinical significance of tumor-draining lymph nodes in tumor progression and immunotherapy. Crit Rev Oncol Hematol 2025; 212:104745. [PMID: 40315968 DOI: 10.1016/j.critrevonc.2025.104745] [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: 03/18/2025] [Revised: 04/12/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025] Open
Abstract
Tumor-draining lymph nodes (TDLNs) play a pivotal role in tumor growth and the immune response, activating immune cells such as CD8 + T cells and natural killer cells to combat tumors. However, tumors can subvert TDLNs to avoid immune attack. Initially, TDLNs stimulate a robust antitumor response, but as tumor evolve, they infiltrate with immunosuppressive cells that alter the TDLN environment and potentially promote metastasis. Immunotherapy, including immune checkpoint inhibitor (ICI), have emerged as a potential solution to this challenge by reconfiguring the TDLN environment to enhance immune responses and influence the immune status of the primary tumor. The integrity of the TDLNs is crucial for the efficacy of immunotherapy. Conventional surgery often removes TDLNs, but this may impede immune system function and the effectiveness of immunotherapy. It is therefore recommended that removal of TDLNs be considered after neoadjuvant treatment rather than before adjuvant treatment. Accurate identification of patients who require post-neoadjuvant TDLN removal and the determination of metastatic nodes is of paramount importance in tailoring treatment plans, optimizing of patient outcomes, and improving quality of life.
Collapse
Affiliation(s)
- Qian Xu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jiahui Chu
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qinqin Hu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yanheng Sun
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fan Jiang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Song Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lian Liu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
15
|
Qi TT, Zhou SJ, Yu Z, Li Y, Chen JQ. Unveiling the heterogeneity and immunotherapy potency of tumor-associated neutrophils in the tumor microenvironment of gastric cancer. BMC Gastroenterol 2025; 25:303. [PMID: 40295944 PMCID: PMC12036284 DOI: 10.1186/s12876-025-03920-0] [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: 01/13/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The differentiation characteristics of neutrophils within the gastric cancer (GC) tumor microenvironment (TME) and their interactions with malignant gastric epithelial cells require further investigation. Furthermore, the therapeutic potential of tumor-associated neutrophils (TANs) in immunotherapy remains inadequately explored. METHODS We integrated two single-cell transcriptome datasets comprising 12 samples, including gastric primary tumors, non-tumor tissues, and metastatic tumors, to profile the epithelial cells and TANs atlas within the TME and examine their interaction modules. In addition, these data were integrated with the bulk transcriptomic including the Cancer Genome Atlas - Stomach Adenocarcinoma (TCGA-STAD) and Asian Cancer Research Group (ACRG) datasets to analyze the expression levels of neutrophil-associated genes across the tumor-associated neutrophil subsets. RESULTS We analyzed 3,118 gastric epithelial cells and 2,365 TANs from all samples. Epithelial cells were classified into ten subclusters, while TANs were grouped into five subclusters. In gastric primary tumors, epithelial cell subtypes included primarily MUC16 + and stem-like populations. In metastatic tumors, the epithelial cell subset with high CXCL5 expression was a characteristic subtype. TANs mainly interacted with epithelial cells via the LGALS9-CD45 and CD46-JAG1 pathways. And RGS2 was highly expressed in N4, a tumor-associated neutrophils subcluster characterized by high MMP9 expression, highlighting its potential as an immunotherapy target. CONCLUSION TANs exhibit robust interactions with gastric malignant epithelial cell subsets. Furthermore, RGS2, which is highly expressed in N4, could serve as a promising target for immunotherapy.
Collapse
Affiliation(s)
- Tong-Tong Qi
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery After Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
| | - Si-Jiang Zhou
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery After Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
| | - Zhu Yu
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery After Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
| | - Yong Li
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery After Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
- Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun-Qiang Chen
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China.
- Guangxi Clinical Research Center for Enhanced Recovery After Surgery, Nanning, China.
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China.
| |
Collapse
|
16
|
Peng Z, Zhang X, Liang H, Zheng Z, Wang Z, Liu H, Hu J, Sun Y, Zhang Y, Yan H, Tong L, Xu J, Ji J, Shen L. Atezolizumab and Trastuzumab Plus Chemotherapy for ERBB2-Positive Locally Advanced Resectable Gastric Cancer: A Randomized Clinical Trial. JAMA Oncol 2025:2832721. [PMID: 40244574 PMCID: PMC12006909 DOI: 10.1001/jamaoncol.2025.0522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/10/2025] [Indexed: 04/18/2025]
Abstract
Importance Effective treatment of locally advanced gastric cancer (GC) or gastroesophageal junction (GEJ) cancer remains a challenge. Objective To compare the efficacy and safety of atezolizumab plus trastuzumab plus capecitabine and oxaliplatin chemotherapy (XELOX) vs trastuzumab plus XELOX in Chinese patients with locally advanced human epidermal growth factor receptor 2 (ERBB2; formerly HER2)-positive GC or adenocarcinoma of the GEJ. Design, Setting, and Participants This was an open-label phase 2 randomized clinical trial conducted at 8 study sites in China. Patient recruitment started on February 25, 2021, and this study is ongoing as participants are still being actively followed up. Chinese patients eligible for surgery with locally advanced ERBB2-positive GC or adenocarcinoma of the GEJ were included. Data were analyzed from March 2021 to October 2023. Interventions Eligible patients were enrolled and randomly assigned 1:1 to perioperative treatment with either atezolizumab plus trastuzumab plus XELOX (arm A) or trastuzumab plus XELOX (arm B) for 3 neoadjuvant cycles (3 weeks per cycle) and 5 adjuvant cycles. Main Outcomes and Measures The primary efficacy end point was the pathological complete response (pCR) rate following completion of neoadjuvant therapy and surgery. Results In total, 42 patients were screened and randomly assigned to arm A (n = 21) or arm B (n = 21). The median (range) ages were 61 (33-72) years and 65 (49-72) years in arm A and arm B, respectively, and 39 patients (93%) were male. The pCR rate was significantly higher in arm A (8 [38%]) than arm B (3 [14%]; difference, 23.8%; 90% CI, 1.3-44.7). Age younger than 65 years, male sex, and intestinal Lauren classification were significantly associated with a better pCR rate in arm A. Median event-free survival, disease-free survival, and overall survival were not reached. Based on the same way of interpretation, major pathologic response should be statistically significantly different between the 2 arms, while other outcome measures remained not significantly different. The incidence of treatment-emergent adverse events was 100% (21 of 21) and 100% (21 of 21) in arms A and B, respectively; grade 3 or higher TEAEs, 57% (12 of 21) and 67% (14 of 21), respectively; and serious TEAEs, 29% (6 of 21) and 10% (2 of 21), respectively. Conclusions and Relevance In this randomized clinical trial, add-on atezolizumab to trastuzumab plus XELOX therapy demonstrated promising efficacy in this patient population, and no new safety concerns were raised. Trial Registration ClinicalTrials.gov Identifier: NCT04661150.
Collapse
Affiliation(s)
- Zhi Peng
- 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 and Institute, Beijing, China
| | - 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 and Institute, Beijing, China
| | - Han Liang
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | | | - Zhenning Wang
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China
| | - Hao Liu
- General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiankun Hu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yanqiao Zhang
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Han Yan
- Shanghai Roche Pharmaceuticals, Shanghai, China
| | - Lin Tong
- Shanghai Roche Pharmaceuticals, Shanghai, China
| | - Jiahui Xu
- Shanghai Roche Pharmaceuticals, Shanghai, China
| | - Jiafu Ji
- Peking University Cancer Hospital and Institute, 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 and Institute, Beijing, China
| |
Collapse
|
17
|
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
|
18
|
An S, Liu FT, Wang C. Comment on "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:e012221. [PMID: 40234088 PMCID: PMC12001348 DOI: 10.1136/jitc-2025-012221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025] Open
Affiliation(s)
- Shuai An
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Fang-Tong Liu
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Chenglong Wang
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| |
Collapse
|
19
|
Lv XL, Peng QL, Wang XP, Fu ZC, Cao JP, Wang J, Wang LL, Jiao Y. Snail family transcriptional repressor 1 radiosensitizes esophageal cancer via epithelial-mesenchymal transition signaling: From bioinformatics to integrated study. World J Gastrointest Oncol 2025; 17:97644. [PMID: 40235866 PMCID: PMC11995309 DOI: 10.4251/wjgo.v17.i4.97644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 12/09/2024] [Accepted: 01/15/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Esophageal cancer (ESCA) poses a significant challenge in oncology because of the limited treatment options and poor prognosis. Therefore, enhancing the therapeutic effects of radiotherapy for ESCA and identifying relevant therapeutic targets are crucial for improving both the survival rate and quality of life of patients. AIM To define the role of the transcription factor Snail family transcriptional repressor 1 (SNAI1) in ESCA, particularly its regulation of radiosensitivity. METHODS A comprehensive analysis of TCGA data assessed SNAI1 expression in ESCA. Survival curves correlated SNAI1 levels with radiotherapy outcomes. Colony formation assays, flow cytometry, and a xenograft model were used to evaluate tumor radiosensitivity and apoptosis. Western blot validated protein expression, while Chromatin immunoprecipitation assays examined SNAI1's role in regulating epithelial-mesenchymal transition (EMT). RESULTS SNAI1 expression in ESCA cell lines and clinical specimens emphasizes its central role in this disease. Elevated SNAI1 expression is correlated with unfavorable outcomes in radiotherapy. Downregulation of SNAI1 enhances the sensitivity of ESCA cells to ionizing radiation (IR), resulting in remarkable tumor regression upon IR treatment in vivo. This study underscores the direct involvement of SNAI1 in the regulation of EMT, particularly under IR-induced conditions. Furthermore, inhibiting deacetylation effectively suppresses EMT, suggesting a potential avenue to enhance the response to radiotherapy in ESCA. CONCLUSION This study highlights SNAI1's role in ESCA radiosensitivity, offering prognostic insights and therapeutic strategies to enhance radiotherapy by targeting SNAI1 and modulating EMT processes.
Collapse
Affiliation(s)
- Xiao-Li Lv
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu Province, China
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Qi-Liang Peng
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu Province, China
| | - Xin-Peng Wang
- Department of Radiotherapy, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Zhi-Chao Fu
- Department of Radiotherapy, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Jian-Ping Cao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu Province, China
| | - Jian Wang
- Department of Radiotherapy, The Affiliated Jiangyin People’s Hospital of Nantong University, Jiangyin 214400, Jiangsu Province, China
| | - Li-Li Wang
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yang Jiao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu Province, China
| |
Collapse
|
20
|
Alcindor T, Tankel J, Fiset PO, Pal S, Opu T, Strasser M, Dehghani M, Bertos N, Zuo D, Mueller C, Cools-Lartigue J, Hickeson M, Marcus V, Camilleri-Broet S, Spatz A, Evaristo G, Farag M, Artho G, Elkrief A, Saleh R, Bailey S, Park M, Huang S, Sangwan V, Ferri L. Phase 2 trial of perioperative chemo-immunotherapy for gastro-esophageal adenocarcinoma: The role of M2 macrophage landscape in predicting response. Cell Rep Med 2025; 6:102045. [PMID: 40239627 PMCID: PMC12047487 DOI: 10.1016/j.xcrm.2025.102045] [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: 07/18/2024] [Revised: 11/28/2024] [Accepted: 03/06/2025] [Indexed: 04/18/2025]
Abstract
We present the clinical results of a phase 2 trial combining neoadjuvant docetaxel, cisplatin, 5 Flourouracil, and the PD-L1 inhibitor avelumab in locally advanced gastro-esophageal adenocarcinoma (GEA). Fifty-one patients receive neoadjuvant therapy with 50 proceeding to surgery. Grade 3-4 adverse events occur in 40%; complete/major pathological response is found in 7/50 (14%) and 9/50 (18%), with 2-year disease-free survival of 67.5%. There is no correlation between tumor regression and PD-L1 or mismatch repair (MMR) status. Multiplex immunohistochemistry and longitudinal single-cell transcriptomic profiling reveal alterations in certain innate immune cell populations, particularly noting an M2-tumor-associated macrophage (M2-TAM) proliferation in non-responding tumors. These findings describe the effective nature of this treatment regimen for GEA and reveal associated features of the inflammatory milieux associated with response to chemo-immunotherapy. The specific character of the inflammatory environment in non-responders may, in the future, help personalize treatment. This study was registered at ClinicalTrials.gov (NCT03288350).
Collapse
Affiliation(s)
- Thierry Alcindor
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada; Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada.
| | - James Tankel
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Pierre-Olivier Fiset
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Sanjima Pal
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Touhid Opu
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | | | - Mehrnoush Dehghani
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Nicholas Bertos
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Dongmei Zuo
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Carmen Mueller
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | | | - Marc Hickeson
- Department of Nuclear Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Victoria Marcus
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Sophie Camilleri-Broet
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Alan Spatz
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Gertruda Evaristo
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Mina Farag
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Giovanni Artho
- Department of Diagnostic Radiology, McGill University Health Centre, Montreal, QC, Canada
| | - Arielle Elkrief
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Ramy Saleh
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada; Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Swneke Bailey
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Morag Park
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Sui Huang
- Institute for Systems Biology, Seattle, WA, USA
| | - Veena Sangwan
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Lorenzo Ferri
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
21
|
Lemay F, Sandhu AS, Stein BD, Goodwin R. A Canadian algorithm for upper gastrointestinal cancer management. Front Oncol 2025; 15:1548637. [PMID: 40297809 PMCID: PMC12034531 DOI: 10.3389/fonc.2025.1548637] [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: 12/19/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Recent advances in immunotherapy have changed the treatment landscape for cancers of the upper gastrointestinal (GI) system. Immune checkpoint inhibitors can lead to better survival and improved quality of life for affected individuals. Adopting new treatment strategies in real-world practice can be challenging, and algorithms that are easy to implement in Canadian oncology practices would benefit clinicians and patients. In this study, we present expert opinion on best practices for upper GI cancer management, including a new algorithm that integrates the latest evidence for screening, workup, diagnosis, treatment, and survivorship. The algorithm is based on a novel approach comprising a case-based, accredited educational program with asynchronous discussion among clinicians practicing across Canada, with the input of expert medical oncologists and gastroenterologists. A needs assessment was employed to determine current areas of educational need in the field of upper GI cancers, and a patient representative provided insights into patient concerns and priorities. The best practices described here include seeking patient input throughout treatment, integrating immune checkpoint inhibitors into systemic therapy for both localized and advanced disease, and providing comprehensive supportive care throughout the treatment and survivorship journey.
Collapse
Affiliation(s)
- Frédéric Lemay
- Division of Gastroenterology, Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Amindeep S. Sandhu
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | | | - Rachel Goodwin
- Division of Medical Oncology, Department of Medicine, University of Ottawa, The Ottawa Hospital Regional Cancer Centre, Ottawa, ON, Canada
| |
Collapse
|
22
|
Li JJ, Rogers JE, Waters RE, Gan Q, Blum Murphy M, Ajani JA. Evolution of Therapeutics for Locally Advanced Upper Gastrointestinal Adenocarcinoma. Cancers (Basel) 2025; 17:1307. [PMID: 40282483 PMCID: PMC12025458 DOI: 10.3390/cancers17081307] [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: 03/07/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Upper gastrointestinal (GI) malignancies, including esophageal, gastroesophageal junction (GEJ), and gastric adenocarcinomas, remain a major global health concern, with poor overall survival and high recurrence rate despite aggressive treatment. Patients with very early tumors (cT1a) can benefit from endoscopic therapy. However, patients with locally advanced disease require multimodal therapies that may combine surgery, radiation, and systemic therapies. This review provides a comprehensive overview of recent advancements in the treatment of locally advanced upper GI adenocarcinomas. Surgical resection remains the cornerstone of curative treatment, with perioperative chemotherapy emerging as the standard of care. While preoperative chemoradiation has demonstrated some benefits in esophageal and GEJ cancers, recent data suggest a more limited role for radiation going forward. Immunotherapy has shown some promise in both the adjuvant and perioperative settings but has yet to establish definitive survival benefit. The integration of HER2-targeted therapies into treatment regimens for HER2-positive locally advanced gastroesophageal cancers has not yielded significant improvements, underscoring the need for more effective strategies. Ongoing research focuses on better predictive biomarkers, personalized treatment approaches, and potential organ preservation strategies for patients achieving a clinical complete response. Continued advancements in treatment modalities and precision medicine are critical to improving survival for patients with locally advanced upper GI adenocarcinomas.
Collapse
Affiliation(s)
- Jenny J. Li
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (M.B.M.); (J.A.A.)
| | - Jane E. Rogers
- Department of Pharmacy Clinical Program, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA;
| | - Rebecca E. Waters
- Department of Anatomical Pathology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.E.W.); (Q.G.)
| | - Qiong Gan
- Department of Anatomical Pathology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.E.W.); (Q.G.)
| | - Mariela Blum Murphy
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (M.B.M.); (J.A.A.)
| | - Jaffer A. Ajani
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (M.B.M.); (J.A.A.)
| |
Collapse
|
23
|
Bao Z, Jia N, Zhang Z, Hou C, Yao B, Li Y. Prospects for the application of pathological response rate in neoadjuvant therapy for gastric cancer. Front Oncol 2025; 15:1528529. [PMID: 40291912 PMCID: PMC12021903 DOI: 10.3389/fonc.2025.1528529] [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: 11/15/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
With the annual increase in the incidence and mortality rates of gastric cancer, it has gradually become one of the significant threats to human health. Approximately 90% of gastric cancer patients are diagnosed with adenocarcinoma. Although the 5-year survival rate for early-stage gastric cancer can exceed 90%, due to its concealed symptoms, less than half of the patients are eligible for radical surgical treatment upon diagnosis. For gastric cancer patients receiving palliative treatment, the current expected survival time is only about one year. In China, the majority of gastric cancer patients, accounting for about 80% of the total, are in the locally advanced stage. For these patients, radical surgery remains the primary treatment option; however, surgery alone is often inadequate in controlling tumor progression. In the pivotal MAGIC study, the recurrence rate was as high as 75%, and similar results were obtained in the French ACCORD07-FFCD9703 study. Numerous clinical trials are currently exploring preoperative neoadjuvant therapy for patients with locally advanced gastric cancer. Data indicates that preoperative neoadjuvant therapy can not only reduce the size of the local tumor but also shrink surrounding lymph nodes, thereby downstaging the tumor and improving the R0 resection rate. Additionally, it can decrease tumor cell activity and eliminate potential micrometastases. The emergence of various immunotherapies has ushered in a new era for neoadjuvant treatment options for gastric cancer.
Collapse
Affiliation(s)
| | | | - Zhidong Zhang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | | | | | | |
Collapse
|
24
|
Zhang C, Wu P, Li D, Zhou J, Lin C, Gu X, Shang D, Ma R, Liu J, Zhang G, Wang P, Che Y, Zeng Q, Peng J, Zhao B, Sun N, He J. Efficacy and safety of anti-PD-1 versus anti-PD-L1 in perioperative immunotherapy: A comprehensive reanalysis of randomized controlled trials. MED 2025:100669. [PMID: 40233750 DOI: 10.1016/j.medj.2025.100669] [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: 11/29/2024] [Revised: 12/16/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Perioperative anti-programmed cell death 1 (PD-1)/PD ligand 1 (PD-L1) immune checkpoint inhibitors improve outcomes, but optimal selection between agents remains debated. We compared the efficacy and safety of anti-PD-1 versus anti-PD-L1 in neoadjuvant/adjuvant settings. METHODS PubMed, Embase, Cochrane CENTRAL, and major oncology conferences (up to May 20, 2024) were systematically searched for randomized trials comparing anti-PD-1/PD-L1 with standard perioperative therapy. Data extraction followed PRISMA guidelines, including trial characteristics, efficacy outcomes (pathological response and survival outcome), and safety profiles. Indirect comparisons between agents were conducted through network meta-analysis employing the mirror principle, utilizing both frequentist and Bayesian methodologies. FINDINGS Thirty-one trials (14,974 patients) were analyzed. Anti-PD-1 demonstrated superior pathological complete response (relative risk [RR]: 1.65, 95% confidence interval [CI]: 1.18-2.29, p = 0.003), major pathological response (RR: 1.43, 95% CI: 1.04-1.96, p = 0.026), and disease-free survival (hazard ratio [HR] = 0.82, 95% CI: 0.71-0.96, p = 0.0106) versus anti-PD-L1. Safety profiles were comparable overall, though anti-PD-1 correlated with higher grade 3-5 immune-related adverse events (irAEs). Frequentist and Bayesian analyses yielded consistent results. CONCLUSIONS Perioperative anti-PD-1 therapy shows enhanced efficacy but increased severe irAEs compared to anti-PD-L1, supporting agent-specific considerations in clinical practice. Further tumor-specific evaluations and mature data are warranted. FUNDING This work is supported in part by the CAMS Innovation Fund for Medical Sciences (2024-I2M-ZD-004) and so on.
Collapse
Affiliation(s)
- Chaoqi Zhang
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Peng Wu
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Dongyu Li
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Junhan Zhou
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Chuqi Lin
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Xuanyu Gu
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Dexin Shang
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Ruijie Ma
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Jingjing Liu
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Pan Wang
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Yun Che
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Qingpeng Zeng
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Jilin Peng
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Bohui Zhao
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
25
|
Li C, Duan Y, Zhou S, Tang T, Yang Y, Zhou L. Evaluating the efficacy and safety of neoadjuvant immunochemotherapy versus chemotherapy in locally advanced gastric cancer undergoing radical gastrectomy: a retrospective study. World J Surg Oncol 2025; 23:121. [PMID: 40189585 PMCID: PMC11974004 DOI: 10.1186/s12957-025-03710-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/11/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Locally advanced gastric cancer (LAGC) is challenging to treat, with neoadjuvant chemotherapy (NCT) improving survival. Recent advances suggest that neoadjuvant immunochemotherapy (NICT) may enhance treatment outcomes. This study compares the efficacy and safety of NICT with NCT in LAGC patients who received radical surgery. METHODS We retrospectively analyzed 67 LAGC patients treated at China-Japan Friendship Hospital from January 2023 to January 2024. Patients were divided into two groups: NICT (chemotherapy plus PD-1/PD-L1 inhibitors) and NCT (standard chemotherapy). We compared pathological complete response (pCR), postoperative recovery, complications, and laboratory markers. RESULTS The NICT group demonstrated a significantly higher pCR rate (25.7% vs. 6.2%, P = 0.032) compared to the NCT group. Furthermore, the NICT group showed reduced rates of nerve and vascular invasion (28.6% vs. 31.4%, P = 0.041). Tumor regression grades (P = 0.001) were more favorable in the NICT group, with earlier ypN and ypTNM stages (P = 0.001). Laboratory analysis revealed a greater reduction in tumor markers CEA and CA19-9 in the two groups, with decreased white blood cell counts and elevated liver enzymes. Surgical outcomes, including operative time, blood loss, and hospital stay, were similar between the two groups, with no significant increase in postoperative complications in the NICT group. CONCLUSION NICT is more effective than traditional NCT in improving pathological responses and reducing tumor burden in LAGC patients. It also reduced nerve and vascular invasion without increasing surgical risks.
Collapse
Affiliation(s)
- Chaofeng Li
- General Surgery, Peking University First Hospital, Bejing, China
- Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yu Duan
- Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing, China
- Capital Medical University, Beijing, China
| | - Shengnan Zhou
- Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Tao Tang
- Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yinmo Yang
- General Surgery, Peking University First Hospital, Bejing, China.
| | - Lei Zhou
- Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
26
|
Mukherjee S, Fujiwara Y, Fountzilas C, Pattnaik H, Chatley S, Vadehra D, Kukar M, Attwood K, George A, Advani S, Yu H, Catalfamo K, Brown A, Spickard E, Fungtammasan A, George S, Liao C, Iyer R, Hatoum H. Trifluridine/Tipiracil and Oxaliplatin as Induction Chemotherapy in Resectable Esophageal and Gastroesophageal Junction Adenocarcinoma: A Phase II Study. Cancer Med 2025; 14:e70835. [PMID: 40200573 PMCID: PMC11978735 DOI: 10.1002/cam4.70835] [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: 11/03/2024] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Preoperative chemoradiation (CRT) followed by surgery for localized esophageal and gastroesophageal junction adenocarcinoma (EGAC) is a standard of care with a pathologic complete response (pCR) rate of 20%. We evaluated a novel combination of trifluridine/tipiracil with oxaliplatin as induction chemotherapy (IC) followed by CRT. METHODS We enrolled patients with potentially resectable localized EGAC (T3, T4aN0, or node-positive disease) in this open-label, single-arm, multicenter, Phase II trial between January 2020 and October 2022. Patients received three cycles of IC with trifluridine/tipiracil and oxaliplatin and then underwent concurrent CRT with weekly carboplatin and paclitaxel followed by surgery. The primary objective was to evaluate the pCR rate. The secondary objectives were to evaluate 2-year progression-free survival (PFS), 2-year overall survival (OS), and toxicities. Circulating tumor DNA (ctDNA) was measured at prespecified intervals to assess its correlation with clinical outcomes. RESULTS Of the 22 enrolled patients, 19 (86.4%) were male and 20 (90.9%) were Caucasian. The median age was 61 years, and 12 (54.5%) had their primary disease at the gastroesophageal junction. Twenty (90.9%) patients had T3 disease, and 15 (68.2%) had node-positive disease. Only two patients had pCRs, and an additional five had near pCRs. Since we could not meet our predefined pCR rate at the interim analysis, the study was closed. After a median follow-up of 15.8 months, 2-year OS and PFS were 43% and 41%, respectively. ctDNA clearance was associated with a significantly higher OS rate (p = 0.012) and PFS rate (p = 0.008). Nausea (59.1%) and fatigue (59.1%) were common treatment-related adverse events (AEs); nine (40.9%) patients had Grade 3 or higher AEs. CONCLUSION IC with trifluridine/tipiracil and oxaliplatin followed by CRT did not improve pCR rate in resectable EGAC compared to pCR from previous reports with CRT alone. We found a correlation between ctDNA clearance and improved survival, which merits further investigation. CLINICAL TRIAL INFORMATION NCT04097028.
Collapse
Affiliation(s)
- Sarbajit Mukherjee
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
- Miami Cancer Institute, Baptist Health South FloridaMiamiFloridaUSA
| | - Yu Fujiwara
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | | | - Sarah Chatley
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Deepak Vadehra
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Moshim Kukar
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | - Anthony George
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | - Han Yu
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | - Alyson Brown
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | | | - Sagila George
- Stephenson Cancer Center – University of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Chih‐Yi Liao
- The University of Chicago Medical Center & Biological SciencesChicagoIllinoisUSA
| | - Renuka Iyer
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Hassan Hatoum
- Stephenson Cancer Center – University of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| |
Collapse
|
27
|
Nowotny R, Rhode P, Niebisch S, Chon SH, Alakus H, Thieme R, Gockel I, Plum PS. [Innovations in the Current Guideline on Gastric Carcinoma and Outlook for the Future]. Zentralbl Chir 2025; 150:163-166. [PMID: 40199374 DOI: 10.1055/a-2529-5300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The third version of the national S3 guideline for gastric cancer is currently being finalised and will be published in the near future. Therefore, this review article primarily discusses the aspects of the current 2019 version and provides an outlook on the innovations in the upcoming version. Emphasis will be placed on the transfer of care to centres with appropriate expertise. Overall, previous revisions of the guideline have placed an increased focus on the genetic aspects of the disease and have specified corresponding risk groups for the occurrence of this neoplasia (including the presence of a CDH1 mutation or HNPCC carriers). In addition, more "borderline" cases for surgery, such as patients with oligometastasis or peritoneal carcinomatosis, have been included in the guidelines. However, for the time being, these patient groups should only be treated in trials. Finally, the guideline discusses the increasing use of immunotherapy in current and future treatment regimens, in addition to the current chemotherapeutic standard of care analogous to the FLOT regimen. New targeted therapeutic approaches such as monoclonal antibodies against claudin 18.2 will complement the treatment of gastric cancer and will certainly be included in the next version of the guideline.
Collapse
Affiliation(s)
- Robert Nowotny
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Philipp Rhode
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Stefan Niebisch
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Seung-Hun Chon
- Klinik und Poliklinik für Allgemein-, Viszeral-, Thorax- und Transplantationschirurgie, Uniklinik Köln, Köln, Deutschland
| | - Hakan Alakus
- Klinik und Poliklinik für Allgemein-, Viszeral-, Thorax- und Transplantationschirurgie, Uniklinik Köln, Köln, Deutschland
| | - René Thieme
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | | | - Patrick Sven Plum
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| |
Collapse
|
28
|
Sokol S, Bilusic M. Overcoming common emerging barriers to effective neoadjuvant immunotherapies. Expert Rev Anticancer Ther 2025; 25:393-403. [PMID: 40030884 DOI: 10.1080/14737140.2025.2474733] [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: 01/15/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Neoadjuvant immunotherapy has rapidly evolved as a novel approach in oncology, reshaping the standard treatment for several malignancies, including melanoma, lung, bladder, colorectal, and breast cancer. While it has an acceptable safety profile, challenges persist due to the complexity of the tumor microenvironment (TME), immune evasion, T-cell exhaustion, and identification of biomarkers. Addressing these issues is critical for optimizing treatment regimens, minimizing immune-related adverse events, and ensuring successful clinical integration. AREAS COVERED This review explores current research on neoadjuvant immunotherapy, emphasizing its impact on standard of care treatment, efficacy, safety, and clinical challenges. A literature search was conducted using PubMed and ClinicalTrials.gov for studies published in the last 5 years. Ongoing research aims to enhance the efficacy of neoadjuvant immunotherapy, identify resistance mechanisms, and broaden indications. Current clinical trials focus on biomarker-driven patient selection, refining immune response modulation through combination strategies, and developing evidence-based protocols for implementation into routine oncology practice. EXPERT OPINION Neoadjuvant immunotherapeutic options have rapidly changed the oncological treatment landscape in only a few years, and this treatment paradigm has quickly become a new standard of care in multiple solid tumors. With continued clinical investigation, neoadjuvant immunotherapy has the dramatic potential to further advance cancer care.
Collapse
Affiliation(s)
- Sophia Sokol
- University of Miami Miller School of Medicine, Miami, FL, USA
| | - Marijo Bilusic
- University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, Miami, FL, USA
| |
Collapse
|
29
|
SPACE-FLOT Investigators, Liu DS, Lee MM, Hall K, Watson DI, Ferri L, So J, Donohoe CL, Michael M, Tebbutt NC, Wong DJ, Duong CP, Bright T, Aly A, Gill S, Cheng C, Goh SK, Read M, Tan J, Stevens S, Wong E, Ooi G, Lam YH, Lee E, Williams D, Jackett L, Chan K, Smith G, Chan DL, Merrett N, Gananadha S, Kanhere H, Kennedy L, Smithers M, Thomas J, Bozin M, Chong L, Mori K, Johnson MA, Martin SA, Usatoff V, Jacobs R, Al-Habbal Y, Liew CH, Huynh F, Bohmer R, Pande G, Daruwalla J, Ballal M, Lee D, Ranjan R, MacCormick AD, Wilkins J, Pattison S, Evennett N, Wilkins J, Robertson J, Pang M, Gordon A, Bann S, Lim YK, Samarasam I, Gurunathan R, Yeung J, Allison F, Siblini A, Griffiths EA, Phillips AW, Prasad P, Markar S, Chidambaram S, Chan D, Murphy T, Reynolds J, Nilsson M, Klevebro F, Piessen G, Lerooy J, Wijnhoven B, van der Zijden C, van Hillegersberg R, Triemstra L, Ruurda J, van Berge Henegouwen MI, Gisbertz SS, Lombardi PM, Edmondson A, Wei JQ, Lorenzo A, Alhayo S, Narendra A, Rahim A, Ho R, Granger J, Tran S, Koullouros M, Nguyen A, McVeay C, Gan SW, Hopping E, et alSPACE-FLOT Investigators, Liu DS, Lee MM, Hall K, Watson DI, Ferri L, So J, Donohoe CL, Michael M, Tebbutt NC, Wong DJ, Duong CP, Bright T, Aly A, Gill S, Cheng C, Goh SK, Read M, Tan J, Stevens S, Wong E, Ooi G, Lam YH, Lee E, Williams D, Jackett L, Chan K, Smith G, Chan DL, Merrett N, Gananadha S, Kanhere H, Kennedy L, Smithers M, Thomas J, Bozin M, Chong L, Mori K, Johnson MA, Martin SA, Usatoff V, Jacobs R, Al-Habbal Y, Liew CH, Huynh F, Bohmer R, Pande G, Daruwalla J, Ballal M, Lee D, Ranjan R, MacCormick AD, Wilkins J, Pattison S, Evennett N, Wilkins J, Robertson J, Pang M, Gordon A, Bann S, Lim YK, Samarasam I, Gurunathan R, Yeung J, Allison F, Siblini A, Griffiths EA, Phillips AW, Prasad P, Markar S, Chidambaram S, Chan D, Murphy T, Reynolds J, Nilsson M, Klevebro F, Piessen G, Lerooy J, Wijnhoven B, van der Zijden C, van Hillegersberg R, Triemstra L, Ruurda J, van Berge Henegouwen MI, Gisbertz SS, Lombardi PM, Edmondson A, Wei JQ, Lorenzo A, Alhayo S, Narendra A, Rahim A, Ho R, Granger J, Tran S, Koullouros M, Nguyen A, McVeay C, Gan SW, Hopping E, Thomson I, Barbour A, Gotley D, Frankel A, Patel R, Chew SJH, Lah K, Gill S, Barnett SA, Muralidharan V, Phillips S, Jamel W, Ko BK, Joglekar S, Rajagopalan A, Jaya J, Chung YC, Peeroo S, Bak M, Tiong J, Zhou Z, Crowe A, Newbold R, Trainor B, Pac Soo ML, Khandelwal V, Eikelboom N, Kim K, Moran E, Hammerschlag J, Desmond B, D'Souza J, Lu J, McLay-Barnes R, Gower A, Choi J, Lim YK, Wood D, Whytock K, Surendran S, Paul N, Khan H F, Chia DKA, Leong EKF, Ijner T, Lin YTL, Liew MS, Jaretzke H, Dempster N, Bhanot K, Mian A, Mastoridis S, Gibbons B, Owen-Smith S, Walmsley J, Al Azzawi M, Doyle E, Okamura Y, Keywani K, Ferrari G, Gualtierotti M, De Martini P, Bushati F. Pathological response guides adjuvant 5-fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) chemotherapy in surgically resected gastro-oesophageal cancer (SPACE-FLOT): international cohort study. Br J Surg 2025; 112:znaf056. [PMID: 40156891 DOI: 10.1093/bjs/znaf056] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Many patients with locally advanced gastro-oesophageal cancers are unable to complete adjuvant 5-fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) chemotherapy, raising questions about its therapeutic utility. The aim of this study was to examine whether pathological response to neoadjuvant FLOT can guide its adjuvant use. METHODS Patients with non-metastatic gastro-oesophageal adenocarcinoma who received neoadjuvant FLOT and underwent surgery from 1 January 2017 to 1 January 2022 from 43 hospitals across 12 countries were analysed. Pathological response was assessed using tumour regression grading systems, trichotomized into minimal responders (MR; worst category), complete responders (CR; pCR), and partial responders (PR; between MR and CR). Survival outcomes of patients who did and did not receive adjuvant FLOT were compared using Kaplan-Meier, Cox regression, propensity score matched, and sensitivity analysis. RESULTS A total of 1887 patients (459 MR, 221 CR, and 1207 PR) were evaluated. The median follow-up was 25.5 (interquartile range 15.0-39.1) months. In the MR group, there was no difference in disease-free survival (DFS; HR 1.03 (95% c.i. 0.78 to 1.36), P = 0.836) between those who did and did not receive adjuvant FLOT. Whilst there was a difference in non-adjusted OS, this became statistically non-significant after adjusting for baseline characteristics (HR 0.96 (95% c.i. 0.70 to 1.30), P = 0.801). In the CR group, there was no difference in DFS (HR 0.88 (95% c.i. 0.41 to 1.85), P = 0.724) or OS (HR 0.69 (95% c.i. 0.31 to 1.54), P = 0.343) between those who did and did not receive adjuvant FLOT. In the PR group, adjuvant FLOT conferred a significant DFS (HR 0.68 (95% c.i. 0.55 to 0.86), P < 0.001) and OS (HR 0.55 (95% c.i. 0.44 to 0.69), P < 0.001) benefit. CONCLUSION Pathological response to neoadjuvant FLOT may guide the use of adjuvant FLOT, enabling personalized approaches to treatment.
Collapse
|
30
|
Chan LL, Chan SL. Future perspectives on immunotherapy for hepatocellular carcinoma. Ther Adv Med Oncol 2025; 17:17588359251323199. [PMID: 40144682 PMCID: PMC11938898 DOI: 10.1177/17588359251323199] [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: 10/26/2024] [Accepted: 02/05/2025] [Indexed: 03/28/2025] Open
Abstract
In recent years, several global phase III trials have shown that combinations of immune checkpoint inhibitors (ICIs) offer superior efficacy and survival compared to multi-kinase inhibitors, establishing them as the gold standard for treating patients with advanced hepatocellular carcinoma (HCC). This success has led to investigations into expanding the use of immunotherapy into various other settings and populations, including neoadjuvant and adjuvant therapies, patients with decompensated liver function and those awaiting liver transplantation. Despite its proven efficacy, a significant number of patients still develop resistance to immunotherapy, highlighting the need for innovative strategies to address this challenge. Approaches aimed at enhancing tumour immunogenicity, such as combining immunotherapy with transarterial chemoembolization or radiation therapies, show significant promise. Additionally, novel immunotherapeutics - such as triplet therapy, bispecific antibodies, adoptive T-cell therapy and cancer vaccines - are in early development for HCC. These agents have demonstrated potential for synergistic effects with existing ICIs, with initial studies yielding positive outcomes. In this review, we offer our future perspective on immunotherapy, emphasizing emerging indications, novel combination strategies and the development of new immunotherapeutic agents. Overall, the future of immunotherapy in HCC is brimming with extraordinary potential, set to transform the treatment landscape and redefine the possibilities for managing this challenging disease.
Collapse
Affiliation(s)
- Landon L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, SIRT, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Stephen L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, SIRT, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong, China
| |
Collapse
|
31
|
Aguiar-Ibáñez R, Mbous YPV, Sharma S, Chawla E. Assessing the clinical, humanistic, and economic impact of early cancer diagnosis: a systematic literature review. Front Oncol 2025; 15:1546447. [PMID: 40177242 PMCID: PMC11962897 DOI: 10.3389/fonc.2025.1546447] [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: 12/16/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction There is a clear consensus among healthcare providers on the advantages of early cancer detection and treatment. However, no in-depth review has yet fully presented the clinical, humanistic, and economic benefits of early cancer diagnosis compared to late detection across a broad range of tumor types. Methods A systematic literature review was conducted to determine the clinical, humanistic, and economic benefits of early cancer diagnosis, as opposed to late diagnosis, as reported in non-interventional studies conducted worldwide. Searches were conducted using electronic databases (MEDLINE and Embase), conference repositories and grey literature. Observational studies in adults diagnosed with bladder cancer, gastric cancer, head and neck cancer (HNC), melanoma, non-small cell lung cancer (NSCLC), renal-cell carcinoma (RCC), and triple negative breast cancer (TNBC) were eligible for inclusion if they reported survival, health-related quality of life (HRQoL), healthcare resource utilization and/or costs, according to stage at diagnosis. Identified records were screened and extracted by two independent reviewers, and discrepancies were resolved by a third reviewer. The quality of studies was assessed using the Newcastle-Ottawa scale and the Larg and Moss adapted checklist. Results Of the 3,159 records identified, 103 studies were included in this review. The general trend showed worse clinical, humanistic, and economic outcomes when patients were diagnosed at a later stage compared to an earlier stage. Patients diagnosed at an earlier stage, had on average, substantially higher survival rates and lower mortality rates across all cancer types and incurred lower resource utilization and costs (with available evidence for patients with NSCLC, TNBC, and HNC), compared to those diagnosed at a more advanced/later stage. Limited evidence on the humanistic burden suggested that with a more advanced stage at diagnosis, patients with bladder cancer experienced reduced HRQoL. Conclusion Early cancer diagnosis (i.e., cancer diagnosed at earlier stages or with lower grades) was associated with longer survival, improved quality of life and lower healthcare costs and resource utilization compared to diagnosis of cancer at later stages or higher grades, as reported by overall survival (OS) and HRQoL outcomes. These findings emphasize the importance of screening and early detection of cancer to improve outcomes among patients diagnosed with cancer.
Collapse
|
32
|
Chen Z, Ma Y, Chen J. Applications and challenges of immunotherapy in the management of gastric adenocarcinoma: current status and future perspectives. World J Surg Oncol 2025; 23:92. [PMID: 40108691 PMCID: PMC11921727 DOI: 10.1186/s12957-025-03752-y] [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: 01/21/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Gastric adenocarcinoma (GAC) remains a significant global public health challenge, characterized by high incidence and mortality rates. Progress in tumor immunology has introduced immune checkpoint inhibitors (ICIs) targeting the programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) pathways, demonstrating substantial potential in GAC therapy. Clinical research indicates that ICIs, particularly when combined with chemotherapy or targeted therapies, significantly enhance treatment efficacy in advanced GAC and specific molecular subtypes, including microsatellite instability-high (MSI-H) and human epidermal growth factor receptor 2 (HER2)-positive patients. However, immunotherapy is also associated with a range of immune-related adverse events (irAEs), necessitating effective management strategies to ensure treatment safety and maintain patients' quality of life. Future studies should focus on identifying new therapeutic targets, optimizing patient selection, and developing personalized treatment approaches to further improve the efficacy and safety of immunotherapy in GAC.
Collapse
Affiliation(s)
- Zhiyao Chen
- Department of Gastrointestinal & Esophageal Surgery, The 2nd Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yunbin Ma
- Department of General surgery, Yiling Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianan Chen
- Department of Clinical Sciences, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, USA.
| |
Collapse
|
33
|
Dong H, Peng Y, Wang X, Cui H. An updated review on immune checkpoint inhibitor-induced colitis: epidemiology, pathogenesis, treatment strategies, and the role of traditional Chinese medicine. Front Immunol 2025; 16:1551445. [PMID: 40165945 PMCID: PMC11955479 DOI: 10.3389/fimmu.2025.1551445] [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: 12/25/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Immune checkpoint inhibitor-induced colitis (irColitis) is a common and severe adverse reaction to immune checkpoint inhibitors (ICIs), significantly impacting the treatment outcomes and quality of life of cancer patients. Epidemiological studies indicate that the incidence of irColitis is associated with factors such as the type of ICIs, the patient's gender, age, and medical history. Although the exact pathophysiology remains unclear, irColitis is thought to be related to immune system activation and dysregulation, gut microbiota imbalance, and impaired epithelial barrier function. This review summarized the epidemiology, clinical presentation, diagnostic criteria, and pathogenesis of irColitis. Additionally, the standard and novel therapeutic strategies of irColitis, including corticosteroids, biologics, and gut microbiota interventions, more importantly the potential and application of Traditional Chinese Medicine (TCM). Future researches call for deeper mechanistic investigations, the development of biomarkers, and reveal the integration of TCM therapies within individual immunotherapy frameworks.
Collapse
Affiliation(s)
- Huijing Dong
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yanmei Peng
- Department of Oncology, Fangshan Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xinmeng Wang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Huijuan Cui
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
34
|
Long B, Zhou H, Yu Z, Zhu J, Yang H, Huang Z, Wei D, Chen S, Yang X, Zhao X, Zhang W, Yan H, Guan X, Li L, Zhang G, Yu H, Che S, Gao Z, Jiang X, Luo C, Mao J, Zhao D, Li Y, Jiang Z, Jiao Z. Neoadjuvant cadonilimab plus FLOT chemotherapy in locally advanced gastric/gastroesophageal junction adenocarcinoma: A multicenter, phase 2 study. MED 2025; 6:100531. [PMID: 39536755 DOI: 10.1016/j.medj.2024.10.008] [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: 07/15/2024] [Revised: 08/06/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Treatment with cadonilimab and chemotherapy has shown promise as a first-line treatment for gastric or gastroesophageal junction (G/GEJ) adenocarcinoma. However, its application in neoadjuvant settings has not yet been documented. METHODS This multicenter, phase 2 trial (ChiCTR2200066893) was conducted at four hospitals across China. Treatment-naive patients with locally advanced G/GEJ adenocarcinoma (cT3/4, N+, M0) and who were human epidermal growth factor receptor 2 negative received 3-cycle or 4-cycle neoadjuvant treatment of cadonilimab plus FLOT (5-fluorouracil, leucovorin, oxaliplatin, and docetaxel) chemotherapy, followed by gastrectomy and 4-cycle adjuvant FLOT chemotherapy. The primary endpoint was the pathological complete response (pCR) rate. Secondary endpoints included major pathological response (MPR), overall response rate (ORR), disease control rate (DCR), R0 resection rate, downstaging rate, and safety. FINDINGS Between December 23, 2022, and December 15, 2023, 32 of 38 patients completed the scheduled treatment, achieving an R0 resection rate of 100% (32/32). The pCR rate was 21.1% (8/38, 90% confidence interval [CI]: 9.7-32.4), and the MPR rate was 44.7% (17/38, 90% CI: 30.9-58.5). Radiological evaluations were available for 28 of 38 patients by blinded independent central review. The ORR was 60.7% (17/28, 90% CI: 44.7-76.7), and the DCR was 100.0% (28/28, 90% CI: 100.0-100.0). Tumor downstaging occurred in 71.9% of patients (23/32), with consistent efficacy across all populations observed in the subgroup analysis. Grade 3 adverse events occurred in 31.6% of patients without severe safety issues. CONCLUSIONS Neoadjuvant cadonilimab plus FLOT chemotherapy treatment exhibits promising efficacy with manageable toxicities in locally advanced G/GEJ adenocarcinoma, providing preliminary evidence for further investigation. FUNDING This study was funded by Akeso Biopharma.
Collapse
Affiliation(s)
- Bo Long
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Huinian Zhou
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Zeyuan Yu
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Junmin Zhu
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Hanteng Yang
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Zeping Huang
- Lanzhou University Second Hospital, The Oncological Surgery Department, Lanzhou, China
| | - Dengwen Wei
- Sun Yat-sen University Cancer Center Gansu Provincial Cancer Hospital, The Gastrointestinal Surgery Department, Lanzhou, China
| | - Shigong Chen
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Xiaojun Yang
- Gansu Provincial Hospital, The General Surgery Department, Lanzhou, China
| | - Xiaoning Zhao
- Sun Yat-sen University Cancer Center Gansu Provincial Cancer Hospital, The Gastrointestinal Surgery Department, Lanzhou, China
| | - Wenjuan Zhang
- Lanzhou University Second Hospital, The Radiology Department, Lanzhou, China
| | - Hong Yan
- Lanzhou University Second Hospital, The Pathology Department, Lanzhou, China
| | - Xiaoying Guan
- Lanzhou University Second Hospital, The Pathology Department, Lanzhou, China
| | - Long Li
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Gengyuan Zhang
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Hongwei Yu
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Shengfu Che
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Zhongti Gao
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Xiangyan Jiang
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Changjiang Luo
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Jie Mao
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Da Zhao
- The First Hospital of Lanzhou University, The Oncology Department, Lanzhou, China
| | - Yumin Li
- Lanzhou University Second Hospital, The Oncological Surgery Department, Lanzhou, China
| | - Zebin Jiang
- Gansu Provincial Hospital, The General Surgery Department, Lanzhou, China
| | - Zuoyi Jiao
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China; Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
35
|
Pazo-Cid R, Pons PG. Integrating immunotherapy in the treatment of resectable gastric cancer: Are we on the right track? MED 2025; 6:100543. [PMID: 40088881 DOI: 10.1016/j.medj.2024.10.020] [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: 10/20/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 03/17/2025]
Abstract
Long et al.1 evaluated neoadjuvant (NAT) cadonilimab plus FLOT chemotherapy (ChT) in locally advanced gastric (G) and gastroesophageal junction (GEJ) adenocarcinoma, showing a pathological complete response (pCR) rate of 21.1% and an R0 resection rate of 100%. These findings align with other perioperative immunotherapy trials, although challenges remain in optimizing event-free survival (EFS).
Collapse
Affiliation(s)
- Roberto Pazo-Cid
- Miguel Servet University Hospital, Medical Oncology Department, 50009 Zaragoza, Spain; University of Zaragoza, School of Medicine, 50009 Zaragoza, Spain; Aragon Institute of Health Sciences, 50009 Zaragoza, Spain.
| | - Paula Gomila Pons
- Miguel Servet University Hospital, Medical Oncology Department, 50009 Zaragoza, Spain
| |
Collapse
|
36
|
Ji X, Wang G, Pan D, Xu S, Lei X. Efficacy and safety of pembrolizumab in advanced gastric and gastroesophageal junction cancer: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:173. [PMID: 40087572 PMCID: PMC11908035 DOI: 10.1186/s12876-025-03754-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Pembrolizumab, a PD-1 inhibitor, has shown potential for treating advanced gastric and gastroesophageal junction (GEJ) cancer. This meta-analysis evaluates its efficacy and safety, alone or combined with chemotherapy, in this population. METHODS A systematic review and meta-analysis were conducted in accordance with PRISMA guidelines. Databases including PubMed, Embase, the Cochrane Central Register of Controlled Trials, and Web of Science were searched up to October 31, 2024. Twelve studies comprising 4,069 patients were included. The primary outcomes were overall survival (OS) and progression-free survival (PFS); secondary outcomes included objective response rate (ORR), adverse events (AEs), and grade ≥ 3 AEs. Effect sizes were calculated using mean differences (MDs) and odds ratios (ORs) with 95% confidence intervals (CIs). RESULTS Pembrolizumab combined with chemotherapy significantly improved OS (MD = 1.92 months; 95% CI: 0.94 to 2.91) and ORR (MD = 11.05%; 95% CI: 6.29 to 15.82) compared to chemotherapy alone. Pembrolizumab monotherapy did not show a significant effect on OS (MD = 0.24 months; 95% CI: -1.15 to 1.63) and was associated with a significant reduction in PFS (MD = -2.28 months; 95% CI: -2.85 to -1.71) compared to chemotherapy alone. For safety, pembrolizumab monotherapy significantly reduced the risk of AEs (OR = 0.68; 95% CI: 0.57 to 0.81) and grade ≥ 3 AEs (OR = 0.39; 95% CI: 0.30 to 0.51) compared to chemotherapy. Pembrolizumab combined with chemotherapy did not significantly alter the risk of AEs (OR = 1.01; 95% CI: 0.90 to 1.13) or grade ≥ 3 AEs (OR = 1.12; 95% CI: 0.99 to 1.27) compared to chemotherapy alone. CONCLUSION Pembrolizumab combined with chemotherapy improves survival and response rates with a manageable safety profile in advanced gastric and GEJ cancers. Monotherapy shows limited efficacy, highlighting the need for combination strategies and patient selection.
Collapse
Affiliation(s)
- Xiaoying Ji
- Department of Pharmacy, Yiwu Central Hospital, Yiwu, Zhejiang, 322000, China
| | - Guoping Wang
- Department of Pharmacy, Yiwu Central Hospital, Yiwu, Zhejiang, 322000, China
| | - Dandan Pan
- Department of Pharmacy, Yiwu Central Hospital, Yiwu, Zhejiang, 322000, China
| | - Shanxia Xu
- Quzhou Zhong Da Lang Yuan Nursing Home, Quzhou, Zhejiang, 324000, China
| | - Xinming Lei
- The Quzhou Afliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, China.
| |
Collapse
|
37
|
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
|
38
|
Alsina Maqueda M, Teijo Quintáns A, Cuatrecasas M, Fernández Aceñero MJ, Fernández Montes A, Gómez Martín C, Jiménez Fonseca P, Martínez Ciarpaglini C, Rivera Herrero F, Iglesias Coma M. Biomarkers in gastroesophageal cancer 2025: an updated consensus statement by the Spanish Society of Medical Oncology (SEOM) and the Spanish Society of Pathology (SEAP). Clin Transl Oncol 2025:10.1007/s12094-025-03865-6. [PMID: 40072752 DOI: 10.1007/s12094-025-03865-6] [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: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 03/14/2025]
Abstract
Gastroesophageal carcinomas, including gastroesophageal adenocarcinoma (GEA) and esophageal squamous cell carcinoma (ESCC), pose a global health challenge due to their heterogeneity. The approach to diagnosis and treatment should first differentiate between GEA and ESCC. Over the past decade, therapies for metastatic or advanced GEA/ESCC have expanded, with several new therapeutic targets alongside trastuzumab for metastatic HER2-positive GEA. Four key biomarkers are essential for targeted therapy: HER2 overexpression/amplification, deficient mismatch repair/microsatellite instability (dMMR/MSI), PD-L1, and Claudin18.2 expression. Immunohistochemistry is the recommended method for these biomarkers evaluation. In addition, the assessment of biomarkers like FGFR2b is likely to become routine in the near future. Experts from the Spanish Society of Pathology (SEAP) and the Spanish Society of Medical Oncology (SEOM) have formed a consensus to optimize biomarker detection and usage in clinical practice. Their recommendations aim to improve personalized treatment strategies for GEA and ESCC patients, integrating new diagnostic insights into routine care.
Collapse
Affiliation(s)
- Maria Alsina Maqueda
- Medical Oncology Department, Unidad de Oncología Médica Traslacional, Hospital Universitario de Navarra, Navarrabiomed - Centro de Investigación Sanitaria de Navarra, Pamplona, Spain.
| | - Ana Teijo Quintáns
- Pathology Department, Gastrointestinal and Neuroendocrine Tumors Research Group, Hospital Universitario 12 de Octubre, Research Institute (Imas12), Madrid, Spain
| | - Miriam Cuatrecasas
- Pathology Department, Hospital Clinic de Barcelona, Biomedical Research Institute IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Maria Jesús Fernández Aceñero
- Department of Legal Medicine, Psychiatry and Pathology, Surgical Pathology Department, Hospital Clínico Universitario San Carlos, nstituto de Investigación Sanitaria Clínico San Carlos (IdISSC), Universidad Complutense, Madrid, Spain
- , Madrid, Spain
| | - Ana Fernández Montes
- Medical Oncology Department, Complexo Hospitalario Universitario de Ourense, Ourense, Spain
| | - Carlos Gómez Martín
- Gastrointestinal Cancer and Early Clinical-Translational Research Units, Medical Oncology Division, 12 de Octubre University Hospital, Madrid, Spain
| | - Paula Jiménez Fonseca
- Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Carolina Martínez Ciarpaglini
- Pathology Department, Hospital Clínico Universitario de Valencia, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - Fernando Rivera Herrero
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Mar Iglesias Coma
- Pathology Department, Hospital del Mar, Pompeu Fabra University, Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
39
|
Cabrit N, Cheugoua-Zanetsie M, Tierney J, Thirion P, Nankivell M, Winter K, Yang H, Wijnhoven B, Vernerey D, Smithers BM, Piessen G, Nilsson M, Boonstra J, Ychou M, Law S, Cunningham D, Vathaire FD, Stahl M, Urba S, Valmasoni M, Williaume D, Thomas J, Lordick F, Tepper J, Gebski V, Burmeister B, Paoletti X, Sandick JV, Fu J, Pignon JP, Ducreux M, Faron M, Michiels S. Disease-free survival as surrogate for overall survival in esophageal cancer: An individual patient data meta-analysis of neoadjuvant chemotherapy and chemoradiotherapy. Eur J Cancer 2025; 218:115292. [PMID: 39938127 DOI: 10.1016/j.ejca.2025.115292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND The use of surrogate endpoints may expedite the reporting of study outcomes of clinical trials. The validity of disease-free survival (DFS) as a surrogate for overall survival (OS) in the neoadjuvant treatment of esophageal (E) or gastroesophageal junctional (GEJ) carcinomas remains uncertain. OBJECTIVE To evaluate DFS as a surrogate end-point for OS in E/GEJ using the meta-analytical approach DESIGN, SETTING, AND PARTICIPANTS: individual patient data from an international meta-analysis on operable locally advanced E/GEJ, which including randomized trials comparing at least two of the neo-adjuvant treatment strategies: upfront surgery (S), chemotherapy followed by surgery (CS), and/or chemoradiotherapy followed by surgery (CRS). MAIN OUTCOMES AND MEASURES Individual (Kendall's tau) and trial-level (R2) correlations between DFS and OS were estimated using a Clayton copula. RESULTS DFS and OS data were available for a total of 4518 pts: 2222 pts included in CS vs S, 1908 pts in CRS vs S, and 388 in CS vs CRS comparisons. 3440 patients had a DFS event and 3303 patients died. Kendall's tau was 0.73 [95 % CI 0.71 - 0.75] and R2 trial-level correlation was 0.95 [0.84 - 0.99] for CS vs S, Kendall's tau was 0.76 [0.74 - 0.77] and R2 was 0.96 [0.87 - 0.99] for CRS vs S, Kendall's tau was 0.87 [0.78 - 0.92] and R2 was 0.93 [0.43 - 1] for CRS vs CS. In a multistate model, the median time in the recurrence state was shorter in older vs more recent trials: mean time of 10.8 [10.2 - 11.4] vs 16.5 months [15.4-17.6]. CONCLUSIONS AND RELEVANCE DFS is a validated surrogate endpoint for OS in trials evaluating neoadjuvant chemotherapy or chemoradiotherapy in E/GEJ. DFS may be more useful as an endpoint when delays between recurrences and death become larger.
Collapse
Affiliation(s)
- Nicolas Cabrit
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif, France; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Maurice Cheugoua-Zanetsie
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif, France; Biostatistics and Epidemiology Office, Gustave Roussy, Villejuif, France; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Jayne Tierney
- MRC Clinical trial Unit at UCL, London, United Kingdom; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Pierre Thirion
- St Luke's Radiation Oncology Network Dublin, Trinity College Dublin, Ireland; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Matthew Nankivell
- MRC Clinical trial Unit at UCL, London, United Kingdom; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Kathryn Winter
- NRG Oncology Statistics and Data Management Center, Philadelphia, PA, USA; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Hong Yang
- Sun Yat-Sen University Cancer Center, Guangzhou, China; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Bas Wijnhoven
- Erasmus University Medical Center, Rotterdam, the Netherlands; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Dewi Vernerey
- CHRU Jean Minjoz, Besançon, France; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - B Mark Smithers
- University of Queensland, Princess Alexandra Hospital, Australia; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Guillaume Piessen
- Univ. Lille, CNRS, Inserm, Chu Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille F-59000, France; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Magnus Nilsson
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Jurjen Boonstra
- Leiden University Medical Center, Leiden, the Netherlands; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Marc Ychou
- Val d'Aurelles, Montpellier, France; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Simon Law
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Hong Kong; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - David Cunningham
- National Institute for Health Research, Biomedical Research Centres, Royal Marsden, London, UK; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Florent de Vathaire
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif, France; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Michael Stahl
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; Evang, Kliniken Essen-Mitte, Essen, Germany
| | - Susan Urba
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; University of Michigan, Ann Arbor, USA
| | - Michele Valmasoni
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; Padova University Hospital, Center for Esophageal Diseases, Department of Surgery, Oncology and Gastroenterology, Padova, Italy
| | - Danièle Williaume
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; Centre Eugène Marquis, Rennes, France
| | - Janine Thomas
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; Princess Alexandra Hospital, Woolloongabba, Australia
| | - Florian Lordick
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; Department of Oncology, University of Leipzig Medical Center, Leipzig, Germany
| | - Joel Tepper
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; University of North Carolina School of Medicine, Chapel Hill, USA
| | - Val Gebski
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; NHMRC, Sydney, Australia
| | - Bryan Burmeister
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; Padova University Hospital, Center for Esophageal Diseases, Department of Surgery, Oncology and Gastroenterology, Padova, Italy
| | - Xavier Paoletti
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; Institut Curie, Paris, France
| | - Johanna van Sandick
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Jianhua Fu
- NRG Oncology Statistics and Data Management Center, Philadelphia, PA, USA; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Jean-Pierre Pignon
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif, France; Biostatistics and Epidemiology Office, Gustave Roussy, Villejuif, France; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Michel Ducreux
- CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France; University Paris-Saclay, Medical Oncology Department, Gustave Roussy, France
| | - Matthieu Faron
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif, France; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France
| | - Stefan Michiels
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif, France; Biostatistics and Epidemiology Office, Gustave Roussy, Villejuif, France; CESP U1018, Inserm, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
40
|
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
|
41
|
de la Fouchardière C, Cammarota A, Svrcek M, Alsina M, Fleitas-Kanonnikoff T, Lordick Obermannová R, Wagner AD, Yap Wei Ting D, Enea D, Petrillo A, Smyth EC. How do I treat dMMR/MSI gastro-oesophageal adenocarcinoma in 2025? A position paper from the EORTC-GITCG gastro-esophageal task force. Cancer Treat Rev 2025; 134:102890. [PMID: 39933210 DOI: 10.1016/j.ctrv.2025.102890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
In less than a decade, immune checkpoint inhibitors (ICIs) have transformed the management of mismatch repair-deficient (dMMR) and microsatellite instability-high (MSI) cancers. However, beyond colorectal cancer (CRC), much of the evidence is mostly derived from non-randomized phase II studies or post-hoc analyses of broader clinical trials. dMMR/MSI tumours represent a specific subgroup of gastro-esophageal adenocarcinomas (GEA), accounting for approximately 9 % of cases, with a higher prevalence in early-stage compared to advanced-stage disease and older female patients. These tumours are predominantly sporadic, often linked to MLH1 promoter methylation, and rarely exhibit HER2 overexpression/ERBB2 amplification or other oncogenic drivers. The treatment landscape for early stage dMMR/MSI GEA is likely to change substantially soon, as ICIs have shown high pathological complete response (pCR) rates in small phase II trials, raising questions on optimisation of neoadjuvant therapy, and paving the way for organ preservation. The standard of treatment for untreated patients with advanced dMMR/MSI GEA is chemotherapy + ICI irrespectively of PDL-1 status. However, the role of chemotherapy-free regimen consisting of CTLA-4 plus PD-1 inhibitors remains undetermined. This review addresses these and other emerging questions, offering expert opinions and insights into the future therapeutic landscape for dMMR/MSI GEA.
Collapse
Affiliation(s)
- Christelle de la Fouchardière
- Institut PAOLI-CALMETTES, 232 Boulevard Sainte-Marguerite 13009, Marseille, France; Unicancer GI (UCGI) Group, Paris, France; EORTC-GITC Group, Brussels, Belgium.
| | - Antonella Cammarota
- EORTC-GITC Group, Brussels, Belgium; Hepatobiliary Immunopathology Lab, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Magali Svrcek
- Sorbonne Université, AP-HP, Saint-Antoine Hospital, Department of Pathology, France; LIMICS, UMRS 1142, Campus des Cordeliers 75006, Paris, France
| | - Maria Alsina
- EORTC-GITC Group, Brussels, Belgium; Hospital Universitario de Navarra, Navarrabiomed - IdiSNA, c. de Irunlarrea 3 31008, Pamplona, Spain
| | - Tania Fleitas-Kanonnikoff
- EORTC-GITC Group, Brussels, Belgium; Hospital Clínico Universitario de Valencia, INCLIVA, Valencia, Spain
| | - Radka Lordick Obermannová
- EORTC-GITC Group, Brussels, Belgium; Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute and Faculty of Medicine, Masaryk University, Czech Republic
| | - Anna Dorothea Wagner
- EORTC-GITC Group, Brussels, Belgium; Anna Dorothea Wagner, Department of Oncology, Division of Medical Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011, Lausanne, Switzerland
| | | | - Diana Enea
- Sorbonne Université, AP-HP, Saint-Antoine Hospital, Department of Pathology, France
| | - Angelica Petrillo
- EORTC-GITC Group, Brussels, Belgium; Medical Oncology Unit, Ospedale del Mare, Naples, Italy
| | - Elizabeth C Smyth
- EORTC-GITC Group, Brussels, Belgium; Oxford NIHRBiomedical Research Centre, Churchill Hospital, Oxford OX3 7LE, UK
| |
Collapse
|
42
|
Jiang W, Zhang B, Xu J, Xue L, Wang L. Current status and perspectives of esophageal cancer: a comprehensive review. Cancer Commun (Lond) 2025; 45:281-331. [PMID: 39723635 PMCID: PMC11947622 DOI: 10.1002/cac2.12645] [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: 07/01/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Esophageal cancer (EC) continues to be a significant global health concern, with two main subtypes: esophageal squamous cell carcinoma and esophageal adenocarcinoma. Prevention and changes in etiology, improvements in early detection, and refinements in the treatment have led to remarkable progress in the outcomes of EC patients in the past two decades. This seminar provides an in-depth analysis of advances in the epidemiology, disease biology, screening, diagnosis, and treatment landscape of esophageal cancer, focusing on the ongoing debate surrounding multimodality therapy. Despite significant advancements, EC remains a deadly disease, underscoring the need for continued research into early detection methods, understanding the molecular mechanisms, and developing effective treatments.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Radiation OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenGuangdongP. R. China
| | - Bo Zhang
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Jiaqi Xu
- Department of PathologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Liyan Xue
- Department of PathologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Luhua Wang
- Department of Radiation OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenGuangdongP. R. China
| |
Collapse
|
43
|
Raimondi A, Lonardi S, Murgioni S, Cardellino GG, Tamberi S, Strippoli A, Palermo F, De Manzoni G, Bencivenga M, Bittoni A, Chiodoni C, Lorenzini D, Todoerti K, Manca P, Sangaletti S, Prisciandaro M, Randon G, Nichetti F, Bergamo F, Brich S, Belfiore A, Bertolotti A, Stetco D, Guidi A, Torelli T, Vingiani A, Joshi RP, Khoshdeli M, Beaubier N, Stumpe MC, Nappo F, Leone AG, Pircher CC, Leoncini G, Sabella G, Airo' Farulla L, Alessi A, Morano F, Martinetti A, Niger M, Fassan M, Di Maio M, Kaneva K, Milione M, Nimeiri H, Sposito C, Agnelli L, Mazzaferro V, Di Bartolomeo M, Pietrantonio F. Tremelimumab and durvalumab as neoadjuvant or non-operative management strategy of patients with microsatellite instability-high resectable gastric or gastroesophageal junction adenocarcinoma: the INFINITY study by GONO. Ann Oncol 2025; 36:285-296. [PMID: 39637944 DOI: 10.1016/j.annonc.2024.11.016] [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: 10/15/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND In resectable gastric/gastroesophageal junction adenocarcinoma, microsatellite instability-high (MSI-H) confers improved survival, but limited benefit from chemotherapy. Immunotherapy may eliminate the need for chemotherapy or surgery. PATIENTS AND METHODS INFINITY is a multicenter, multicohort phase II trial (NCT04817826) investigating in cohort 1 the activity and safety of tremelimumab + durvalumab (T300/D) as neoadjuvant treatment of mismatch repair deficient/MSI-H, resectable gastric/gastroesophageal junction adenocarcinoma. Primary endpoint was pathologic complete response (pCR) rate; Secondary endpoints: progression-free survival (PFS), overall survival (OS), quality of life, and translational analyses. In cohort 2, the activity and safety of T300/D was explored as definitive treatment in patients achieving clinical complete response (cCR). Primary endpoint was 2-year cCR rate, and secondary endpoints were PFS, OS, quality of life, gastrectomy-free survival and translational analyses. RESULTS In cohort 1, 18 patients were recruited and 15 evaluable. pCR and major pathologic response-pCR were 60% and 80%, respectively. Since pCR rate in T4 tumors was 17%, this subgroup of patients was excluded from enrollment in cohort 2. At 28.1 months median follow-up, 24-month gastric cancer-specific PFS and OS rates were 85% and 92%, respectively. In cohort 2, 18 patients were enrolled and 17 assessable, and 13 had cCR and started non-operative management. At 11.5 months median follow-up, one patient had local regrowth and underwent salvage surgery; 12-month gastrectomy-free survival was 64.2%. CONCLUSIONS The INFINITY study provided promising activity results of a chemo-free T300/D combination regimen as preoperative treatment in mismatch repair deficient/MSI gastric/gastroesophageal junction adenocarcinoma and the first available feasibility results of a non-operative management strategy in this disease setting, worthy of further validation in larger cohorts.
Collapse
Affiliation(s)
- A Raimondi
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - S Lonardi
- Medical Oncology 3, Veneto Institute of Oncology IOV - IRCCS, Padua, USA
| | - S Murgioni
- Medical Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, USA
| | - G G Cardellino
- Department of Oncology, Presidio Ospedaliero "Santa Maria della Misericordia"-ASUFC, Udine, USA
| | - S Tamberi
- Oncology Unit, Ravenna Hospital, AUSL Romagna, Ravenna, USA
| | - A Strippoli
- Medical Oncology Unit, Policlinico Universitario A. Gemelli, Rome, USA
| | - F Palermo
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - G De Manzoni
- Department of Surgery, Azienda Ospedaliero Universitaria Integrata di Verona-Borgo Trento, Verona, USA
| | - M Bencivenga
- Department of Surgery, Azienda Ospedaliero Universitaria Integrata di Verona-Borgo Trento, Verona, USA
| | - A Bittoni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, USA
| | - C Chiodoni
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, USA
| | - D Lorenzini
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - K Todoerti
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - P Manca
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - S Sangaletti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, USA
| | - M Prisciandaro
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - G Randon
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - F Nichetti
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - F Bergamo
- Medical Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, USA
| | - S Brich
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - A Belfiore
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - A Bertolotti
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - D Stetco
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - A Guidi
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - T Torelli
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - A Vingiani
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | | | | | | | | | - F Nappo
- Medical Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, USA
| | - A G Leone
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - C C Pircher
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - G Leoncini
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - G Sabella
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - L Airo' Farulla
- Division of Nuclear Medicine, Istituto Nazionale Tumori IRCCS Milan, Milan, Italy
| | - A Alessi
- Division of Nuclear Medicine, Istituto Nazionale Tumori IRCCS Milan, Milan, Italy
| | - F Morano
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - A Martinetti
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - M Niger
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - M Fassan
- School of Medicine and Surgery, University of Padua, Padua, Italy
| | - M Di Maio
- Department of Oncology, University of Turin, at Le Molinette Hospital, Turin, Italy
| | | | - M Milione
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | | | - C Sposito
- Department of Oncology, University of Milan and G.I. Surgery, Istituto Nazionale Tumori IRCCS Milan, Milan, Italy
| | - L Agnelli
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA; Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - V Mazzaferro
- Department of Oncology, University of Milan and G.I. Surgery, Istituto Nazionale Tumori IRCCS Milan, Milan, Italy
| | - M Di Bartolomeo
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - F Pietrantonio
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA.
| |
Collapse
|
44
|
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
|
45
|
Su R, Sun X, Luo Y, Gu L, Wang F, Dong A, Yamamoto M, Tsukamoto T, Nomura S, Zhao Z, Dai C, Deng G, Zhuang B, He Y, Zhang C, Yin S. SUSD2 + cancer-associated fibroblasts in gastric cancer mediate the effect of immunosuppression and predict overall survival and the effectiveness of neoadjuvant immunotherapy. Gastric Cancer 2025; 28:245-263. [PMID: 39656339 DOI: 10.1007/s10120-024-01572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/25/2024] [Indexed: 02/21/2025]
Abstract
BACKGROUND The expression patterns and functions of Sushi Domain Containing 2 (SUSD2) differ among various malignancies. This research aims to investigate the expression of SUSD2 and the role of the SUSD2+ cancer-associated fibroblasts (CAFs) for immunotherapy in gastric cancer. METHODS The expression of SUSD2 and specific markers (CD4, CD8, PD-1, TIGIT, TIM-3 and CD163) was determined using immunohistochemistry and multiplex immunofluorescence (mIHC) on paraffin sections. Flow cytometry and western blot were used to assess the expression of SUSD2 in fibroblasts from fresh samples. Also, analysis of single-cell and bulk RNA sequencing was employed to confirm the presence and characterize the function of SUSD2+ CAFs. The predictive power of indicators for neoadjuvant immunotherapy was evaluated via ROC curve analysis. Animal experiment was employed to validate the immunosuppressive effect of SUSD2+ CAFs. RESULTS SUSD2 is mainly expressed on fibroblasts within the tumors and the high infiltration of SUSD2+ CAFs went together with a poor survival and a more advanced tumor stage. Significantly, the joint use of SUSD2+ CAFs and CD8+ T cells demonstrated a remarkable ability to predict the efficacy of neoadjuvant immunotherapy superior to PD-L1 combined positive score. High SUSD2+ CAFs was correlated with resistance to immunotherapy as well as low CD8+ T infiltration and high exhausted T cell infiltration. CONCLUSIONS We have identified a novel subset of CAFs that could predict the survival and response to neoadjuvant immunotherapy of patients. The SUSD2+ CAFs have the potential to serve as a predictive biomarker and a promising target for immunotherapy.
Collapse
Affiliation(s)
- Rishun Su
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Xuezeng Sun
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Yusheng Luo
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Liang Gu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Fulin Wang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Aoran Dong
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Masami Yamamoto
- Laboratory of Physiological Pathology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Sachiyo Nomura
- Department of Clinical Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
| | - Zhenzhen Zhao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Chen Dai
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Guofei Deng
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Baoding Zhuang
- Hepatic-Biliary-Pancreatic Surgery, The Second People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Songcheng Yin
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China.
| |
Collapse
|
46
|
Malekzada F, Vladimiriov M, Leitz M, Michel J, Nimzewski F, Hoeppner J. Neoadjuvant treatment of esophageal cancer: chemotherapy, chemoradiation, immunotherapy, and future trends of therapy. Innov Surg Sci 2025; 10:3-9. [PMID: 40144785 PMCID: PMC11934940 DOI: 10.1515/iss-2023-0005] [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: 01/25/2023] [Accepted: 10/07/2024] [Indexed: 03/28/2025] Open
Abstract
In the Western hemisphere, nonmetastatic locally advanced esophageal carcinoma is mostly treated in multimodal therapy protocols according to current therapy guidelines. In squamous cell carcinoma of the esophagus, neoadjuvant chemoradiation is the favorable option. Unimodal surgical and chemoradiation treatment alternatives show inferior results on this entity. For locally advanced adenocarcinoma of the esophagus perioperative chemotherapy and neoadjuvant chemoradiation have been competing treatment approaches in the recent past. Both are evidence based (class I evidence) and superior compared to unimodal surgery. However, the latest results of head-to-head comparative therapy studies show superior overall survival results for the FLOT regimen of perioperative chemotherapy. Furthermore, immunotherapy and targeted therapy with monoclonal antibodies have become a strong focus of current clinical research. Nivolumab as well as trastuzumab are already an important part of the current standard therapies. In both entities - SCC and AC - a significant quota of patients shows a locoregional complete remission of the tumor in the specimen after modern neoadjuvant therapy and surgical resection. The addition of immunotherapy and targeted therapy to neoadjuvant therapy further increases complete remission rates in defined subgroups according to the results of current studies. Currently, three prospective randomized trials are ongoing on the subject of future possibilities for organ-preserving concepts in case of complete clinical remission ("surgery as needed," "watch and wait"). It is to be expected for the future that curative short-term and long-term treatment results in locally advanced esophageal carcinoma will significantly improve, particularly due to the additional possibilities of immunotherapy and organ-preserving therapy concepts in postneoadjuvant complete remission.
Collapse
Affiliation(s)
- Freschta Malekzada
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Miljana Vladimiriov
- Department of Surgery, Bielefeld University, Medical School and University Medical Center OWL, Klinikum Lippe, Detmold, Germany
| | - Michael Leitz
- Department of Surgery, Bielefeld University, Medical School and University Medical Center OWL, Klinikum Lippe, Detmold, Germany
| | - Julia Michel
- Department of Surgery, Bielefeld University, Medical School and University Medical Center OWL, Klinikum Lippe, Detmold, Germany
| | - Fabian Nimzewski
- Department of Surgery, Bielefeld University, Medical School and University Medical Center OWL, Klinikum Lippe, Detmold, Germany
| | - Jens Hoeppner
- Department of Surgery, Bielefeld University, Medical School and University Medical Center OWL, Klinikum Lippe, Detmold, Germany
| |
Collapse
|
47
|
Rencuzogullari A, Karahan SN, Selcukbiricik F, Lacin S, Taskin OC, Saka B, Karahacioglu D, Gurses B, Ozoran E, Uymaz DS, Ozata IH, Saglam S, Bugra D, Balik E. The New Era of Total Neoadjuvant FLOT Therapy for Locally Advanced, Resectable Gastric Cancer: A Propensity-Matched Comparison With Standard Perioperative Therapy. J Surg Oncol 2025; 131:417-426. [PMID: 39400342 PMCID: PMC12044282 DOI: 10.1002/jso.27934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/28/2024] [Accepted: 09/17/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND The FLOT 4-AIO trial established the docetaxel-based regimen's superiority over epirubicin-based triplet therapy in terms of survival rates and acceptable toxicity for locally advanced resectable gastric (LARGC). Yet, fewer than half of the patients achieved completion of eight prescribed FLOT cycles. We proposed that administering all FLOT cycles in the form of total neoadjuvant therapy may improve completion rates and downstaging. This study contrasted total neoadjuvant therapy (FLOT x8) with standard neoadjuvant therapy (FLOT 4+4) for patients LARGC adenocarcinoma who underwent curative resection with routine D2 lymphadenectomy, focusing on histopathological outcomes, toxicity, and survival outcomes. METHODS We reviewed patients with histologically confirmed advanced clinical stage cT2 or higher, nodal positive stage (cN+), or both, with resectable gastric tumors and no distant metastases (January 2017 to July 2023). We divided patients into two groups, FLOT 4+4 and FLOT x8; FLOT 4+4 patients underwent four preoperative and four postoperative bi-weekly cycles of docetaxel, oxaliplatin, leucovorin, and fluorouracil, while FLOT x8 patients received all eight cycles preoperatively after a gradual practice change starting from January 2020. Propensity score matching adjusted for age, clinical stage, tumor location, and histology. RESULTS Of the 77 patients in the FLOT x8 group, 37 were propensity-matched to an equal number in the FLOT 4+4 group. Demographics, duration of surgery, and hospital stay showed no significant differences between the groups. The FLOT x8 group exhibited a significantly higher all-cycle completion rate at 89.1% compared to FLOT 4+4's 67.6% (p < 0.01). Both groups demonstrated comparable hematological and non-hematological toxicity rates, Clavien-Dindo ≥ 3 complications, and CAP tumor regression grades. The mean number of harvested lymph nodes was 42.5 and 41.2 in the FLOT 4+4 and FLOT x8 groups, respectively. Similar rates of disease-free survival and overall survival were noted in both groups, despite a trend toward a higher pathological complete response rate, albeit not statistically significant (8.1% vs. 18.9%, p = 0.29), in the FLOT x8 group at a median follow-up of 36 months. CONCLUSION Total neoadjuvant therapy with the FLOT x8 protocol corresponds to higher treatment completion rates, a safety profile similar to standard perioperative therapy, and a twofold increase in complete pathological response. Further research on long-term oncological outcomes is needed to confirm the effectiveness of total neoadjuvant therapy.
Collapse
Affiliation(s)
| | - Salih Nafiz Karahan
- Department of General Surgery, School of MedicineKoç UniversityIstanbulTurkey
| | - Fatih Selcukbiricik
- Department of Medical Oncology, School of MedicineKoç UniversityIstanbulTurkey
| | - Sahin Lacin
- Department of Medical Oncology, School of MedicineKoç UniversityIstanbulTurkey
| | - Orhun Cig Taskin
- Department of Pathology, School of MedicineKoç UniversityIstanbulTurkey
| | - Burcu Saka
- Department of Pathology, School of MedicineKoç UniversityIstanbulTurkey
| | | | - Bengi Gurses
- Department of Radiology, School of MedicineKoç UniversityIstanbulTurkey
| | - Emre Ozoran
- Department of General Surgery, School of MedicineKoç UniversityIstanbulTurkey
| | - Derya Salim Uymaz
- Department of General Surgery, School of MedicineKoç UniversityIstanbulTurkey
| | - Ibrahim Halil Ozata
- Department of General Surgery, School of MedicineKoç UniversityIstanbulTurkey
| | - Sezer Saglam
- Department of Medical OncologyDemiroglu Bilim UniversityIstanbulTurkey
| | - Dursun Bugra
- Department of General Surgery, School of MedicineKoç UniversityIstanbulTurkey
- Department of General SurgeryAmerican HospitalIstanbulTurkey
| | - Emre Balik
- Department of General Surgery, School of MedicineKoç UniversityIstanbulTurkey
| |
Collapse
|
48
|
Semenova Y, Kerimkulov A, Uskenbayev T, Zharlyganova D, Shatkovskaya O, Sarina T, Manatova A, Yessenbayeva G, Adylkhanov T. Chemotherapy Options for Locally Advanced Gastric Cancer: A Review. Cancers (Basel) 2025; 17:809. [PMID: 40075656 PMCID: PMC11899121 DOI: 10.3390/cancers17050809] [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: 01/24/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Cancers represent a significant global health burden, affecting millions of individuals each year [...].
Collapse
Affiliation(s)
- Yuliya Semenova
- Department of Surgery, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan;
| | - Altay Kerimkulov
- Department of Multidisciplinary Surgery, National Research Oncology Center, Astana 020000, Kazakhstan; (A.K.); (T.U.); (T.S.); (T.A.)
| | - Talgat Uskenbayev
- Department of Multidisciplinary Surgery, National Research Oncology Center, Astana 020000, Kazakhstan; (A.K.); (T.U.); (T.S.); (T.A.)
| | - Dinara Zharlyganova
- Department of Scientific Management, National Research Oncology Center, Astana 020000, Kazakhstan; (D.Z.); (G.Y.)
| | - Oxana Shatkovskaya
- Board for Strategic Development, Scientific and Educational Activities, National Research Oncology Center, Astana 020000, Kazakhstan;
| | - Tomiris Sarina
- Department of Multidisciplinary Surgery, National Research Oncology Center, Astana 020000, Kazakhstan; (A.K.); (T.U.); (T.S.); (T.A.)
| | - Almira Manatova
- Department of Scientific Management, National Research Oncology Center, Astana 020000, Kazakhstan; (D.Z.); (G.Y.)
| | - Gulfairus Yessenbayeva
- Department of Scientific Management, National Research Oncology Center, Astana 020000, Kazakhstan; (D.Z.); (G.Y.)
| | - Tasbolat Adylkhanov
- Department of Multidisciplinary Surgery, National Research Oncology Center, Astana 020000, Kazakhstan; (A.K.); (T.U.); (T.S.); (T.A.)
| |
Collapse
|
49
|
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
|
50
|
Li Y, Cao Y, Wang X, Li C, Zhao L, Li H. Effects of perioperative treatment of resectable adenocarcinoma of esophagogastric junction by immunotherapy (Adebrelimab) combined with chemotherapy (XELOX): protocol for a single-center, open-labeled study (AEGIS trial, neoadjuvant immunochemotherapy). BMC Cancer 2025; 25:198. [PMID: 39905364 PMCID: PMC11792203 DOI: 10.1186/s12885-025-13589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND For resectable adenocarcinoma of the esophagogastric junction (AEG), current treatment exploration primarily focuses on perioperative chemotherapy regimens combined with PD-1/PD-L1 inhibitors, but the long-term survival benefits of still require further investigation, and the use of upfront immunotherapy is typically restricted to patients with metastatic MSI-H (M1 MSI-H) disease due to their potential responsiveness to immunological agents. Adebrelimab, as a novel PD-L1 antibody, has not yet been proven for its efficacy and safety in adenocarcinoma of the esophagogastric junction. METHODS The AEGIS study is a prospective, open-labeled, single-arm, phase II clinical trial. A total of 26 patients with AEG will be enrolled. The primary endpoint is the pathologic complete response (pCR) rate after perioperative neoadjuvant immunochemotherapy. Secondary outcomes of the study include the objective response rate (ORR), R0 resection rate, major pathological response (MPR) rate, and pCR rate in combined positive score(CPS) ≥ 5 and MSI-H populations, event-free survival (EFS), and overall survival (OS). The exploratory outcomes are the biomarkers related to therapeutic efficacy, such as PD-L1 expression, microsatellite instability (MSI), tumor mutational burden(TMB), Epstein-Barr virus(EBV) infection, and circulating tumor DNA(ctDNA). DISCUSSION This trail aims to verify the efficacy and safety of the perioperative treatment regimen of anti-PD-L1 (Adebrelimab) combined with chemotherapy (capecitabine plus oxaliplatin, XELOX) for patients with resectable AEG. Considering the differences in chemotherapy regimen tolerance between Asian and Western populations, this study intends to evaluate the suitability of Adebrelimab combined with XELOX chemotherapy for the Asian population. TRIAL REGISTRATION ClinicalTrials.gov: NCT06482788. The trial was prospectively registered on 22 May 2024, https://clinicaltrials.gov/study/NCT06482788 .
Collapse
Affiliation(s)
- Yingyi Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuqin Cao
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xipeng Wang
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chengqiang Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liqin Zhao
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|