1
|
Hirase Y, Arigami T, Matsushita D, Shimonosono M, Uenosono Y, Yanagita S, Tsuruda Y, Sasaki K, Baba K, Kawasaki Y, Ohtsuka T. Function-preserving gastrectomy based on the sentinel node concept prevents osteosarcopenia in patients with gastric cancer. Gastric Cancer 2025; 28:696-704. [PMID: 40287906 DOI: 10.1007/s10120-025-01617-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Gastric cancer remains a significant global challenge, with conventional surgery for early gastric cancer often leading to post-gastrectomy complications. Sentinel node navigation surgery is being developed to preserve quality of life without compromising radicality. Although osteosarcopenia is linked to gastrointestinal cancers and prognosis, its impact on bone and muscle mass after function-preserving surgery for gastric cancer remains underexplored. METHODS We analyzed the data of patients diagnosed with early gastric cancer and not eligible for endoscopic treatments, who underwent either distal gastrectomy or sentinel node navigation surgery at our hospital between 2010 and 2020. Skeletal muscle index and bone mineral density were measured preoperatively and 1, 3, and 5 years, postoperatively; rates of changes in these measures were assessed. RESULTS Among the 63 patients included, 42 (67%) underwent conventional surgery, and 21 (33%) underwent function-preserving gastrectomy using the sentinel node technique. No significant difference in postoperative survival rates was observed between the two groups (P = 0.97). The rate of change in the skeletal muscle index and bone mineral density decreased in both groups from 1 to 3 years postoperatively. At 5 years postoperatively, the sentinel node navigation surgery group showed an increase in skeletal muscle index and bone mineral density change rates, the difference observed between the two groups was significant (P < 0.05). CONCLUSION Sentinel node navigation surgery for early gastric cancer may help prevent decreases in bone and muscle mass. This suggests that its use has a potential role in preventing osteosarcopenia.
Collapse
Affiliation(s)
- Yuki Hirase
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Takaaki Arigami
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan.
| | - Daisuke Matsushita
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Masataka Shimonosono
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Yoshikazu Uenosono
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Shigehiro Yanagita
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Yusuke Tsuruda
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Ken Sasaki
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Kenji Baba
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Yota Kawasaki
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Takao Ohtsuka
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| |
Collapse
|
2
|
Sun F, Gao X, Li T, Zhao X, Zhu Y. Tumor immune microenvironment remodeling after neoadjuvant therapy in gastric cancer: Update and new challenges. Biochim Biophys Acta Rev Cancer 2025; 1880:189350. [PMID: 40355011 DOI: 10.1016/j.bbcan.2025.189350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Gastric cancer (GC) is a malignant tumor with one of the highest morbidity and death rates in the world. Neoadjuvant therapy, including neoadjuvant chemotherapy (NAC) and NAC combined with immunotherapy, can improve the resection and long-term survival rates. However, not all patients respond well to neoadjuvant therapy. It has been confirmed that immune cells in the tumor immune microenvironment, including T cells, B cells, and natural killer cells, can affect the efficacy of neoadjuvant therapy. This paper summarizes current preclinical and clinical evidence to more fully describe the effects of neoadjuvant therapy on the immune microenvironment of GC, to provide the impetus to identify biomarkers to predict the potency of neoadjuvant therapy, and to identify the mechanisms of drug resistance, which should promote the development of individualized and accurate treatments for GC patients.
Collapse
Affiliation(s)
- Fujing Sun
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Xiaozhuo Gao
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Tianming Li
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Xiaoyan Zhao
- Graduate School, Dalian Medical University, Dalian, China
| | - Yanmei Zhu
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China.
| |
Collapse
|
3
|
Wang J, Zhu Y, Li Q, Wang L, Bian H, Lu X, Ye Z. Spectral CT-based nomogram for evaluation of neoadjuvant chemotherapy response in esophageal squamous cell carcinoma. Eur Radiol 2025; 35:3800-3811. [PMID: 39729110 DOI: 10.1007/s00330-024-11294-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/15/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVES To establish a spectral CT-based nomogram for predicting the response to neoadjuvant chemotherapy (NAC) in patients with locally advanced esophageal squamous cell carcinoma (ESCC). METHODS This retrospective study included 172 patients with ESCC who underwent spectral CT scans before NAC followed by resection. Based on postoperative tumor regression grades (TRG), 34% (58) of patients were responsive (TRG1) and 66% (114) were non-responsive (TRG2-3). The data was divided into a primary set of 120 and a validation set of 52, maintaining a 7:3 random ratio. Measurements included iodine concentration (IC), normalized iodine concentration (nIC), CT40kev, CT70kev, spectral attenuation curve slope (λHU), and effective atomic number (Zeff) during non-contrast and venous phases (VP). Clinicopathologic characteristics were collected. Univariable and multivariable logistic regressions identified independent predictors of NAC response. The model was visualized using nomograms, and its efficacy was assessed via receiver operating characteristic (ROC) curves. RESULTS Multivariable logistic regression analysis identified the neutrophil-to-lymphocyte ratio (NLR), clinical stage, ZeffVP, and nICVP as independent predictors of NAC response. The nomogram incorporating all four independent predictors, outperformed spectral CT and the clinical model with the highest AUCs of 0.825 (95% CI: 0.746-0.895) for the primary set and 0.794 (95% CI: 0.635-0.918) for the validation set (DeLong test: all p < 0.05). CONCLUSIONS The spectral CT and clinical models were useful in predicting NAC response in ESCC patients. Combining spectral CT imaging parameters and clinicopathologic characteristics in a nomogram improved predictive accuracy. KEY POINTS Question Developing a non-invasive, practical tool to predict ESCC's response to chemotherapy is crucial and has not yet been done. Findings This nomogram, incorporating clinicopathologic characteristics and spectral CT-derived parameters, predicted NAC response in ESCC patients. Clinical relevance This spectral CT-based nomogram is a non-invasive and easily obtainable tool for accurately predicting ESCC response to NAC, aiding clinicians in personalized treatment planning.
Collapse
Affiliation(s)
- Jing Wang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yueqiang Zhu
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Qian Li
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lining Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haiman Bian
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiaomei Lu
- CT Clinical Science CT, Philips Healthcare, Beijing, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
4
|
Ma W, Guo H, Yang P, He J, Li T, Wu H, Wu J, Yang J, Tian Y, Guo S, Wang G, Wang H, Wang Q, Er L, Ding P, Zhao Q. Lymph node metastasis mapping and prognostic analysis of early gastric cancer. J Gastrointest Surg 2025; 29:102082. [PMID: 40381833 DOI: 10.1016/j.gassur.2025.102082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/01/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Currently, numerous studies focus on the analysis of risk factors for lymph node metastasis in early gastric cancer, but few studies analyze the drainage patterns of metastatic lymph nodes. METHODS Data were retrospectively analyzed from a database of gastric cancer resections from 2014 to 2018. The cohort included 786 pT1 patients with complete data. Outcomes evaluated were lymph node metastasis frequencies, survival analyses, and risk factors affecting prognosis. RESULTS The overall lymph node metastasis rate was 23.7%. The 5-year overall survival (OS) rate (54.8% vs 95.7%; P <.001) and disease-free survival (DFS) rate (48.4% vs 95.7%; P <.001) of patients with node-positive disease were significantly worse than those of patients with node-negative disease. Multivariable Cox regression identified tumor size of >2 cm (P =.007; P <.001), poor differentiation (P =.007; P <.001), T1b stage (all P <.001), lymph node metastasis (all P <.001), and vascular invasion (all P =.002; P =.016) as independent negative prognostic factors affecting 5-year OS and DFS in patients with early gastric cancer. Postoperative chemotherapy (P <.001; P =.019) was an independent positive prognostic factor. CONCLUSION This real-world observational study demonstrates that lymph node metastasis in early gastric cancer is widely and disorderly not depending on the location. Therefore, systematic lymph node dissection is necessary to cure early gastric cancer. Meanwhile, its prognosis is closely related to lymph node metastasis.
Collapse
Affiliation(s)
- Wenqian Ma
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China; Department of Endoscopy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Honghai Guo
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China
| | - Peigang Yang
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China
| | - Jinchen He
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China
| | - Tongkun Li
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China
| | - Haotian Wu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China
| | - Jiaxiang Wu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China
| | - Jiaxuan Yang
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China
| | - Yuan Tian
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China
| | - Shuo Guo
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China; Department of Endoscopy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gongning Wang
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China; Department of Endoscopy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongcai Wang
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China; Department of Endoscopy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qi Wang
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China; Department of Endoscopy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Limian Er
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China; Department of Endoscopy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ping'an Ding
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China.
| | - Qun Zhao
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China; Big data analysis and mining application for precise diagnosis and treatment of gastric cancer, Hebei Provincial Engineering Research Center, Shijiazhuang, China.
| |
Collapse
|
5
|
Hu Y, Wen T, Tuo B. The role of ICG NIRL fluorescence imaging in the surgical treatment of digestive system tumors (Review). Mol Med Rep 2025; 32:181. [PMID: 40280113 PMCID: PMC12059463 DOI: 10.3892/mmr.2025.13546] [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: 12/20/2024] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Indocyanine green (ICG) is a relatively non‑toxic fluorescent dye with a history of safe use, which has fueled the development of new applications for ICG. Research on the use of ICG near‑infrared light (NIRL) fluorescence imaging during oncologic surgery has increased, revealing its role in tumor identification and localization, lymph node navigational resection and blood perfusion assessment. The purpose of the present review was to provide a comprehensive overview of advances in the clinical application of ICG NIRL fluorescence imaging during gastrointestinal tumor surgery. The present review discusses the techniques, outcomes, limitations and key considerations necessary for clinical practice, aiming to provide a valuable resource for professionals in the field.
Collapse
Affiliation(s)
- Yanxia Hu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Tingyuan Wen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
6
|
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
|
7
|
Dawson-Gore CC, Evans L, Vincent S, Colbert MT, Harnke B, Moyer A, Alaniz VI, De la Torre L, Bischoff A. The need for screening recommendations after intestinal vaginal replacement: a systematic review of the literature. Pediatr Surg Int 2025; 41:172. [PMID: 40522524 DOI: 10.1007/s00383-025-06081-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/02/2025] [Indexed: 06/18/2025]
Abstract
PURPOSE To investigate the occurrence, presentation, management, and treatment of patients found to have a primary colorectal malignancy arising in the intestinal neovagina. METHODS A systematic review of primary research studies describing intestinal malignancy arising in an intestinal neovagina was conducted. Reports of extraintestinal malignancy and secondary research study designs were excluded. RESULTS Sixteen case reports were identified over 35 years. Patients commonly had sigmoid (n = 11, 69%) neovaginas. Indications for vaginal reconstruction included Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome (n = 5, 31%), gynecologic carcinoma (n = 3, 19%), vaginal agenesis (n = 5, 31%) and cloaca (n = 3, 19%). Symptom onset ranged from 7 to 53 years after reconstruction. Age at diagnosis ranged from 17 to 53 years. Bleeding was the most common presenting symptom (n = 12, 73%). Fifty percent of patients presented with local disease (n = 8), 31% with locoregional (n = 5) and 6% with metastasis (n = 1). Treatment varied with most patients undergoing surgical resection. Survival was reported for 10 patients, half of which experienced disease recurrence. Surveillance plans were rarely described. CONCLUSION Adenocarcinoma in intestinal neovaginas is a rare but life-threatening disease that lacks routine screening, management and surveillance guidelines. Our findings underscore the variability in management of these patients, and the morbidity of the disease. Work to develop formalized screening guidelines is ongoing.
Collapse
Affiliation(s)
- Catherine C Dawson-Gore
- Department of Pediatric Surgery, Children's Hospital Colorado, 13123 East 16th Avenue Box 323, Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado School of Medicine, 13001 E 17th Pl, Aurora, CO, 80045, USA
| | - Lauren Evans
- Department of Pediatric Surgery, Children's Hospital Colorado, 13123 East 16th Avenue Box 323, Anschutz Medical Campus, Aurora, CO, 80045, USA
- International Center for Colorectal and Urogenital Care, Children's Hospital Colorado, 13123 East 16th Avenue Box 323, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Sage Vincent
- Department of Pediatric Surgery, Children's Hospital Colorado, 13123 East 16th Avenue Box 323, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Marie-Teresa Colbert
- International Center for Colorectal and Urogenital Care, Children's Hospital Colorado, 13123 East 16th Avenue Box 323, Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pediatric and Adolescent Gynecology, Children's Hospital Colorado, 13123 E 16th Ave, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ben Harnke
- University of Colorado Strauss Health Sciences Library, 12950 E Montview Blvd, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Amber Moyer
- University of Colorado School of Medicine, 13001 E 17th Pl, Aurora, CO, 80045, USA
- Department of Surgery, University of Colorado School of Medicine, 12631 East 17th Ave, Aurora, CO, 80045, USA
| | - Veronica I Alaniz
- University of Colorado School of Medicine, 13001 E 17th Pl, Aurora, CO, 80045, USA
- International Center for Colorectal and Urogenital Care, Children's Hospital Colorado, 13123 East 16th Avenue Box 323, Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pediatric and Adolescent Gynecology, Children's Hospital Colorado, 13123 E 16th Ave, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Luis De la Torre
- Department of Pediatric Surgery, Children's Hospital Colorado, 13123 East 16th Avenue Box 323, Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado School of Medicine, 13001 E 17th Pl, Aurora, CO, 80045, USA
- International Center for Colorectal and Urogenital Care, Children's Hospital Colorado, 13123 East 16th Avenue Box 323, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Andrea Bischoff
- Department of Pediatric Surgery, Children's Hospital Colorado, 13123 East 16th Avenue Box 323, Anschutz Medical Campus, Aurora, CO, 80045, USA.
- University of Colorado School of Medicine, 13001 E 17th Pl, Aurora, CO, 80045, USA.
- International Center for Colorectal and Urogenital Care, Children's Hospital Colorado, 13123 East 16th Avenue Box 323, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
8
|
Yang C, Wang P, Yang M, Lu Q, Zhu Z, Lu H, Li H, Zhang Z, Li M, Zhao L, Li J, Ling B, Fu X, Tong A. A fc-engineered NKG2D × B7-H3 bispecific antibody enhances the antitumor activity by orchestrating cytotoxic lymphocytes. Int Immunopharmacol 2025; 161:115032. [PMID: 40516256 DOI: 10.1016/j.intimp.2025.115032] [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: 01/01/2025] [Revised: 05/31/2025] [Accepted: 06/03/2025] [Indexed: 06/16/2025]
Abstract
B7-H3, an immune-checkpoint molecule that is overexpressed in several cancer types, has been identified as a promising immunotherapy target. However, most immunotherapy approaches against B7-H3+ tumor cells focus on manipulating the T cells. Natural killer (NK) cells, another important part of the cellular immune system, also exhibit anti-tumor properties and play complementary roles in tumor eradication with T cells. In this study, natural killer group 2D (NKG2D), an activating receptor in most cytotoxic immune cells, was selected for engaging NK cells. We obtained specific anti-NKG2D nanobodies via phage display and developed a series of B7-H3 × NKG2D bispecific antibodies (bsAb) with different formats to fight against the B7-H3+ tumor cells. Through functional comparison of candidate antibodies in vitro, B1-C53 was selected and further modified with the optimized Fc fragment (known as FC-C53) to enhance anti-tumor immunity by antibody-dependent cell-mediated cytotoxicity (ADCC). Stronger tumor lysis mediated by FC-C53 was demonstrated both in vitro and in vivo when simultaneously directed at both NK cells and CD8+ T cells, particularly after the additional administration of a B7-H3 × CD3 bispecific T cell engager that targets B7-H3 with another epitope. Overall, we provided a strategy based on the B7-H3 × NKG2D antibody to improve the anti-B7-H3 immunotherapy approaches by orchestrating cytotoxic lymphocytes.
Collapse
Affiliation(s)
- Chen Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Wang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mingjun Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qizhong Lu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhixiong Zhu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huaqing Lu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hexian Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zongliang Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Li
- Blood Research Laboratory, Chengdu Blood Center, Gongtong Street, Jinjiang District, Chengdu 610041, China
| | - Lizhou Zhao
- Blood Research Laboratory, Chengdu Blood Center, Gongtong Street, Jinjiang District, Chengdu 610041, China
| | - Jia Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Ling
- Department of obstetrics and gynecology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China.
| | - Xuemei Fu
- Blood Research Laboratory, Chengdu Blood Center, Gongtong Street, Jinjiang District, Chengdu 610041, China.
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China.
| |
Collapse
|
9
|
Ahn JH, Yi JW. DNA methylation changes in thyroid cancer patients infected with SARS-CoV-2. Updates Surg 2025:10.1007/s13304-025-02233-x. [PMID: 40490622 DOI: 10.1007/s13304-025-02233-x] [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: 03/14/2025] [Accepted: 04/28/2025] [Indexed: 06/11/2025]
Abstract
The impact of SARS-CoV-2 infection on thyroid cancer at the genomic level remains poorly understood. The purpose of our study was to determine whether significant DNA methylation changes occur in thyroid cancer tissues from patients with recent SARS-CoV-2 infection. Surgically resected normal thyroid and Papillary (PTC) tissues from three COVID-19-infected PTC patients (Cases) and three prepandemic PTC patients (Controls) were analyzed using DNA methylation EPIC arrays. Differentially methylated probes (DMPs) and differentially methylated regions (DMRs) were identified in normal thyroid and PTC tissues. Functional enrichment analysis was subsequently performed to explore the affected pathways. COVID-19-infected PTC tissues presented distinct DNA methylation profiles, with 6,848 DMPs in PTC tissues compared with 140 in normal thyroid tissues. SARS-CoV-2 infection did not significantly affect normal thyroid tissue by methylation. SARS-CoV-2 infection in PTC tissues was associated with hypermethylation of tumor suppressor genes (RUNX3, PAOX), the Wnt signaling pathway, the HOX gene family, cell adhesion-related genes and hypomethylation in response to virus-related genes. The key DMRs identified in PTC included GPR75, CCDC80, and ENTPD3, suggesting altered cell adhesion, tumor proliferation, and immune evasion. SARS-CoV-2 infection is linked to significant DNA methylation alterations in PTC tissues, with potential implications for tumor progression and aggressiveness. These findings suggest that COVID-19 may influence thyroid cancer biology. Further research is needed to validate these epigenetic modifications, establish causal relationships and determine their clinical relevance.
Collapse
Affiliation(s)
- Jong-Hyuk Ahn
- Department of Surgery, Chung-Ang University Hospital, Seoul, Korea
| | - Jin Wook Yi
- Department of Surgery, Inha University College of Medicine, Incheon, Korea.
- Department of Surgery, Inha University Hospital, Incheon, Korea.
| |
Collapse
|
10
|
Wen J, Zheng B, Fu T. CXCL12 expression and the survival of patients with gastric cancer: a meta-analysis. Clin Exp Med 2025; 25:191. [PMID: 40481962 PMCID: PMC12145318 DOI: 10.1007/s10238-025-01674-3] [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: 03/02/2025] [Accepted: 04/05/2025] [Indexed: 06/11/2025]
Abstract
Gastric cancer (GC) remains a leading cause of cancer-related mortality worldwide. CXCL12, a chemokine involved in tumor progression and metastasis, has been inconsistently associated with GC survival. This meta-analysis aimed to evaluate the prognostic significance of CXCL12 expression in GC patients. A comprehensive literature search was conducted in PubMed, Embase, and Web of Science. Observational studies assessing tumor CXCL12 expression and survival outcomes in GC patients were included. Hazard ratios (HRs) with 95% confidence intervals (CIs) were pooled using a random-effects model by incorporating heterogeneity. Ten studies comprising 1361 GC patients were included. High CXCL12 expression was significantly associated with poorer overall survival (OS) (HR: 1.85, 95% CI 1.51-2.26, p < 0.001) with mild heterogeneity (I2 = 17%). Subgroup analyses revealed that the association between high CXCL12 expression and OS was stronger in studies defining high expression as above the median density value (HR: 2.63, 95% CI 1.79-3.86) than in those using any positive expression (HR: 1.61, 95% CI 1.30-2.00; p for subgroup difference = 0.03). Additionally, a more pronounced association was observed in studies with follow-up durations ≥ 36 months (HR: 2.42, 95% CI 1.84-3.18) compared to those with < 36 months (HR: 1.59, 95% CI 1.28-1.99; p = 0.03). The pooled results also indicated an association between high CXCL12 expression and worse progression-free survival (PFS) (HR: 1.52, 95% CI 1.05-2.20, p = 0.03). High CXCL12 expression is associated with poorer survival outcomes in GC patients.
Collapse
Affiliation(s)
- Jinxiu Wen
- Department of Gastrointestinal and Hernia Surgery, Heyuan People's Hospital, Guangdong Provincial People's Hospital Heyuan Hospital, No. 733, Wenxiang Road, Yuancheng District, Heyuan, 517000, China
| | - Bingbing Zheng
- Department of Gastrointestinal and Hernia Surgery, Heyuan People's Hospital, Guangdong Provincial People's Hospital Heyuan Hospital, No. 733, Wenxiang Road, Yuancheng District, Heyuan, 517000, China.
| | - Ting Fu
- Department of Gastrointestinal and Hernia Surgery, Heyuan People's Hospital, Guangdong Provincial People's Hospital Heyuan Hospital, No. 733, Wenxiang Road, Yuancheng District, Heyuan, 517000, China
| |
Collapse
|
11
|
Li M, Cao J, Wang Y, Zhao Z, Ai L, Zhang K. Predictive power of tertiary lymphoid structure for prognosis and neoadjuvant chemotherapy response in HER2-positive breast cancer. Medicine (Baltimore) 2025; 104:e42566. [PMID: 40489875 DOI: 10.1097/md.0000000000042566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/11/2025] Open
Abstract
This study evaluated the prognostic significance of tertiary lymphoid structures (TLS) in human epidermal growth factor receptor 2 (HER2)-positive breast cancer (BC), focusing on their associations with survival outcomes, response to neoadjuvant therapy, and potential as a biomarker for personalized treatment strategies. Data from patients with HER2-positive BC in the METABRIC and The Cancer Genome Atlas databases were analyzed. TLS expression scores were calculated using gene set variation analysis, and their associations with survival outcomes were assessed. Immune cell infiltration, immune checkpoint expression, tumor mutational burden, and pathway enrichment were also evaluated. Data from the I-SPY2 clinical trial and a clinicopathological cohort of 19 patients from Xiangya Hospital were used to assess the relationship between TLS expression and pathological complete response following neoadjuvant therapy. High TLS expression was associated with improved survival and increased infiltration of antitumor immune cells. TLS-high tumors were enriched in immune-related pathways, whereas TLS-low tumors showed activation of proliferation and metabolism pathways. Patients with high TLS expression had better responses to neoadjuvant therapy, while those with low TLS expression derived greater benefit from dual-targeted treatments. TLS represents a promising biomarker for predicting survival and response to neoadjuvant therapy in HER2-positive BC, with potential to support personalized treatment strategies.
Collapse
Affiliation(s)
- Mengxi Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan, China
| | - Jing Cao
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan, China
| | - Yueheng Wang
- Department of Clinical Medicine, Queen Marry School, Nanchang University, Nanchang, Jiangxi, China
| | - Ziru Zhao
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan, China
| | - Liqiang Ai
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Kejing Zhang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan, China
| |
Collapse
|
12
|
Wu Y, Zhang F, Du F, Huang J, Wei S. Combination of tumor organoids with advanced technologies: A powerful platform for tumor evolution and treatment response (Review). Mol Med Rep 2025; 31:140. [PMID: 40183402 PMCID: PMC11976518 DOI: 10.3892/mmr.2025.13505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Malignant tumors notably decrease life expectancy. Despite advances in cancer diagnosis and treatment, the mechanisms underlying tumorigenesis, progression and drug resistance have not been fully elucidated. An emerging method to study tumors is tumor organoids, which are a three‑dimensional miniature structure. These retain the patient‑specific tumor heterogeneity while demonstrating the histological, genetic and molecular features of original tumors. Compared with conventional cancer cell lines and animal models, patient‑derived tumor organoids are more advanced at physiological and clinical levels. Their synergistic combination with other technologies, such as organ‑on‑a‑chip, 3D‑bioprinting, tissue‑engineered cell scaffolds and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR‑associated protein 9, may overcome limitations of the conventional 3D organoid culture and result in the development of more appropriate model systems that preserve the complex tumor stroma, inter‑organ and intra‑organ communications. The present review summarizes the evolution of tumor organoids and their combination with advanced technologies, as well as the application of tumor organoids in basic and clinical research.
Collapse
Affiliation(s)
- Ying Wu
- Department of Obstetrics and Gynecology, The 920th Hospital of Joint Logistics Support Force, Kunming, Yunnan 650032, P.R. China
| | - Fan Zhang
- Department of Comprehensive Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, P.R. China
| | - Furong Du
- Department of Medicine, Kingbio Medical Co., Ltd., Chongqing 401123, P.R. China
| | - Juan Huang
- Department of Breast Surgery and Multidisciplinary Breast Cancer Center, Clinical Research Center of Breast Cancer in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Shuqing Wei
- Department of Comprehensive Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, P.R. China
| |
Collapse
|
13
|
Sonmezoz GB, Yilmaz M. Association of neutrophil-lymphocyte ratio, platelet-lymphocyte ratio and monocyte-to-high-density lipoprotein ratio with diabetic nephropathy. Ther Apher Dial 2025; 29:428-436. [PMID: 40119545 DOI: 10.1111/1744-9987.70008] [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: 01/13/2025] [Revised: 02/22/2025] [Accepted: 03/07/2025] [Indexed: 03/24/2025]
Abstract
INTRODUCTION The aim of this study was to determine the relationship between albuminuria and neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and monocyte-to-high density lipoprotein-cholesterol ratio (MHR). METHODS Patients with type 2 diabetes mellitus diagnosis, aged over 18, had estimated glomerular filtration rate (eGFR) ≥60 mL/dk/1.73 m2 included. Patients were divided into groups according to ACR values: <30 mg/g (group 1), 30-300 mg/g (group 2) and >300 mg/g (group 3). We examined whether there was a significant difference in NLR, PLR, and MHR among the three groups. RESULTS A total of 360 patients were included in the study. NLR was significantly higher in group 3 than in group 1 (p = 0.016). There was no significant difference in PLR or MHR among the three groups (p = 0.312 and p = 0.687, respectively). A significant difference was detected in NLR in comparison between the groups with and without diabetic nephropathy, but there was no significant difference in PLR or MHR (p = 0.028; p = 0.950 and p = 0.389, respectively). NLR correlated with creatinine and ACR (r: 0.166, p = 0.002; r: 0.144, p = 0.006, respectively). MHR correlated positively with creatinine (r: 0.25.3, p = 0.016, respectively). CONCLUSION NLR was significantly higher in the diabetic nephropathy group than in the non-diabetic nephropathy group. This may suggest that NLR can be used as a prognostic marker in diabetic nephropathy. Although there was no significant relationship between MHR and albuminuria, MHR positively correlated with creatinine and negatively correlated with eGFR. Therefore, MHR may be useful in monitoring the development and progression of chronic kidney disease in diabetic patients rather than in the early stages. However, further studies are needed.
Collapse
Affiliation(s)
- Gulru Birce Sonmezoz
- Taksim Training and Research Hospital, Department of Internal Medicine, Istanbul, Turkey
| | - Murvet Yilmaz
- Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Department of Nephrology, Istanbul, Turkey
| |
Collapse
|
14
|
Pierce K, Gaskins J, Martin Ii RCG. The Weight of Nutrition on Post-Resection Oncologic Morbidity and Mortality: A Systematic Review and Meta-Analysis of Nutritional Indices. Nutr Rev 2025; 83:988-1005. [PMID: 39405175 DOI: 10.1093/nutrit/nuae138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025] Open
Abstract
CONTEXT Nutritional status is a critical factor in the selection of patients for solid tumor resection. A variety of indices have been developed to quantify nutritional status, and they have differing degrees of predictive power for various postoperative outcomes. OBJECTIVE This study aimed to comprehensively evaluate the predictive ability of commonly used nutritional indices in relation to postoperative complications (POCs), recurrence-free survival (RFS), and OS. DATA SOURCES We performed a systematic review of 14 established nutritional indices from January 2015 to July 2022. DATA EXTRACTION The primary end point was OS, while the secondary end points were POCs and RFS. A subsequent meta-analysis was performed to further assess the predictive ability of these indices for OS based on general index type, primary tumor site, and the patient's index status. DATA ANALYSIS In this evaluation, 38 articles reporting data on 23 970 patients were analyzed, focusing on 14 nutritional indices. The indices were categorized into phenotypic, metabolic, immunologic, and combined types. Patients within the cut-off range of any index were predicted to have lower OS (hazard ratio [HR] 2.14, 95% CI 1.84-2.49, P < .01). Lower gastrointestinal (GI) and "other" sites were less predictive than upper GI primary tumors (HR 1.63, HR 1.82, and HR 2.54, respectively; all with P < .01). Phenotypic indices were less predictive than combined indices (HR 1.73 vs HR 2.47, P < .01). Within the combined category, there was no significant difference in the predictive ability of Prognostic Nutritional Index (PNI) vs Geriatric Nutritional Risk Index (GNRI) vs Controlling Nutritional Index (CONUT) (HR 2.63 vs HR 2.42 vs HR 2.07, P = .07). CONCLUSION The predictive efficacy of a nutritional index was found to be highly dependent on the index type, the primary tumor site, and the outcome of interest. In the context of upper GI resections, nutritional status appeared to be more of a significant predictor of OS, compared with cases involving lower GI and hepatic malignancies. Indices that integrate phenotypic, metabolic, and immunologic patient factors potentially offer greater clinical utility in forecasting OS.
Collapse
Affiliation(s)
- Katherine Pierce
- Division of Surgical Oncology, The Hiram C. Polk, Jr., M.D. Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, United States
| | - Jeremy Gaskins
- Department of Bioinformatics and Biostatistics, University of Louisville School of Medicine, Louisville, KY 40292, United States
| | - Robert C G Martin Ii
- Division of Surgical Oncology, The Hiram C. Polk, Jr., M.D. Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, United States
| |
Collapse
|
15
|
Mariani A, Triantafyllou E, Kepenekian V, Zaanan A, Glehen O, Karoui M. Management of peritoneal gastric metastasis: An update. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109731. [PMID: 40106892 DOI: 10.1016/j.ejso.2025.109731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/13/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
IMPORTANCE Peritoneal metastases from gastric cancer (PMGC) are associated with poorer median survival and systemic chemotherapy remains the standard of care. This narrative review summarizes the current evidence for medical and surgical treatment of PMGC. OBSERVATIONS Treatment is moving to local ways of administering chemotherapy, either intraperitoneal normothermic chemotherapy, laparoscopic hyperthermic intraperitoneal chemotherapy (HIPEC) or even pressurized intraperitoneal aerosol chemotherapy. Furthermore, cytoreductive surgery±HIPEC could also be an alternative in specific situations. CONCLUSIONS and relevance: This review provides an updated summary of existing strategies that can be discussed in the management of patients with PMGC.
Collapse
Affiliation(s)
- Antoine Mariani
- Department of Digestive Surgery, Université Paris Cité, Assistance Publique Hôpitaux de Paris, Georges Pompidou University Hospital, Paris, France.
| | - Evangelia Triantafyllou
- Department of Digestive Surgery, Université Paris Cité, Assistance Publique Hôpitaux de Paris, Georges Pompidou University Hospital, Paris, France
| | - Vahan Kepenekian
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Aziz Zaanan
- Department of Digestive Oncology, Université Paris Cité, Assistance Publique Hôpitaux de Paris, Georges Pompidou University Hospital, Paris, France
| | - Olivier Glehen
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Mehdi Karoui
- Department of Digestive Surgery, Université Paris Cité, Assistance Publique Hôpitaux de Paris, Georges Pompidou University Hospital, Paris, France
| |
Collapse
|
16
|
Richter M, Doll C, Hofmann E, Reds A, Mrosk F, Neumann K, Heiland M, Koerdt S, Raguse JD, Jöhrens K. The prevalence and prognostic value of CD168 in early-stage oral squamous cell carcinoma. Oral Oncol 2025; 165:107329. [PMID: 40294580 DOI: 10.1016/j.oraloncology.2025.107329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 03/19/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
OBJECTIVE CD168 has been established as a negative prognostic marker in various tumor entities leading to a poor prognosis. Regarding OSCC, there is a lack of comprehensive studies that examine the correlation between CD168 expression and clinical outcome, hindering the implementation of this prognostic factor into clinical practice. MATERIALS AND METHODS This retrospective analysis included all patients with primary pT1 and pT2 pN0 OSCC who received surgical therapy without the need for adjuvant therapy (pN0, M0, R0) over a seven-year long period. Immunohistochemical staining for CD168 and Mib/Ki67 was evaluated using tissue microarrays of primary tumors and correlated with clinical outcome. A cut-off value for CD168 expression of ≥10 % was considered as positive. RESULTS A total of 139 patients (male: 91 (65.5 %), female: 48 (34.5 %)) with a mean age of 61.2 years were included (mean follow-up: 62.6 months). CD168 expression was evident in 35 (25.2 %) tumors leading to higher levels of Mib/Ki67 positivity (p < 0.001). Tumor biopsies of stage T2 OSCC stained positive for CD168 more frequently when compared to T1 tumors (p = 0.002). Tumors with CD168 expression ≥ 10 % had a significantly lower OS (p < 0.001) and RFS (p = 0.011) compared to patients with lower expression. Multivariate Cox regression identified CD168 status as a risk factor for OS (HR 2.10, CI: 1.06-4.14; p = 0.033). CONCLUSION Our results demonstrate a significant proportion of CD168-positive tumors in OSCC and suggest an impact on prognosis, particularly with regard to OS.
Collapse
Affiliation(s)
- Maximilian Richter
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, Berlin 13353, Germany.
| | - Christian Doll
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, Berlin 13353, Germany
| | - Elena Hofmann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, Berlin 13353, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Charitéplatz 1, Berlin 10117, Germany
| | - Anna Reds
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, Berlin 13353, Germany
| | - Friedrich Mrosk
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, Berlin 13353, Germany
| | - Konrad Neumann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology, Charitéplatz 1, Berlin 10117, Germany
| | - Max Heiland
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, Berlin 13353, Germany
| | - Steffen Koerdt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, Berlin 13353, Germany
| | - Jan-Dirk Raguse
- Department of Oral and Maxillofacial Surgery, Fachklinik Hornheide, Dorbaumstrasse 300, Münster 48157, Germany
| | - Korinna Jöhrens
- Institute of Pathology, Klinikum Chemnitz gGmbH, Flemmingstraße 2, Chemnitz 09116, Germany
| |
Collapse
|
17
|
Mondeali M, Mozaffari F, Ghiasizadeh P, Bakhti S, Salehi M, Kamali N, Rafat M. miR-216 Is a Key Regulator and Potential Marker in Human Cancers. Adv Biomed Res 2025; 14:40. [PMID: 40519574 PMCID: PMC12165296 DOI: 10.4103/abr.abr_184_23] [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: 05/26/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 06/18/2025] Open
Abstract
MicroRNAs, a class of small noncoding RNAs, have been identified as promising biomarkers for cancer identification and management by regulating gene expression and other cellular biological pathways. This review gathers findings for understanding the molecular basis and clinical importance of microRNA-216 (miR-216) in several cancers. Increased or decreased expression of miR-216 has been observed in a variety of cancers, including esophageal cancer, breast cancer, colorectal cancer, gastric cancer, pancreatic cancer, cervical cancer, brain tumor (glioma), prostate cancer, and acute myeloid leukemia, indicating its activity as an oncogene or tumor suppressor. Through this study, we proposed that miR-216 can potentially be a candidate as a prognostic marker for early detection of tumor development, progression, as well as metastasis in cancer patients.
Collapse
Affiliation(s)
- Mozhgan Mondeali
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mozaffari
- Student Research Committee, Arak University of Medical Sciences, Arak, Iran
| | | | - Sanaz Bakhti
- Department of Biological Sciences, Islamic Azad University, North Tehran, Tehran, Iran
| | - Marjan Salehi
- Department of Microbiology, Islamic Azad University, Qom Branch, Qom, Iran
| | - Navid Kamali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Rafat
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
18
|
Xu Z, Lei Z, Cheng Q, Gao Y, Xiang Y. Biomedical applications of organoids derived from the digestive system. Front Cell Dev Biol 2025; 13:1599384. [PMID: 40519270 PMCID: PMC12162665 DOI: 10.3389/fcell.2025.1599384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 05/19/2025] [Indexed: 06/18/2025] Open
Abstract
The global incidence of digestive system diseases is increasing, posing a significant public health challenge and driving an escalating demand for research into the mechanisms underlying their onset and progression. Traditional cell models and xenotransplantation animal models have been widely used to simulate human digestive diseases, thereby enhancing our understanding of disease occurrence, progression, and drug resistance. However, these models fail to fully replicate the complex cellular microenvironment and spatial structure, and are further limited by individual and species differences. Organoid technology, as an emerging in vitro cell culture approach, enables the precise culturing and differentiation of human stem cells to generate highly tissue-specific and functionally intact organoids. This technology not only better recapitulates cell-to-cell interactions, extracellular matrix (ECM) microenvironment, and organ-specific physiological functions but also more closely mimics the human physiological state in vitro. Moreover, it reduces reliance on animal experiments, enhances the translatability of research findings, mitigates the limitations of animal models and two-dimensional cell models, and plays a pivotal role in simulating the physiological and pathological processes of the human digestive tract. Currently, common techniques for constructing organoids include embedding culture, rotating culture, magnetic suspension culture, organ-on-a-chip, three-dimensional (3D), and four-dimensional (4D) printing technologies. Seed cells are primarily derived from digestive system epithelial cells and pluripotent stem cells. This article reviews the construction methods of digestive system organoids, evaluates their applications in studying growth and development mechanisms, disease modeling and mechanism research, drug screening, regenerative medicine, and precision medicine, and identifies existing challenges and future research directions to provide a valuable reference for biomedical research.
Collapse
Affiliation(s)
- Zhensheng Xu
- Department of Oncologic Chemotheraphy, Haikou Affiliated Hospital of Central South University Xiangya School of Medcine, Haikou, China
| | - Zhongwen Lei
- Department of Hepatobiliary Surgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medcine, Haikou, China
| | - Qiuhua Cheng
- Department of Hepatobiliary Surgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medcine, Haikou, China
| | - Yuanhui Gao
- Central Laboratory, Haikou Affiliated Hospital of Central South University Xiangya School of Medcine, Haikou, China
| | - Yang Xiang
- Department of Hepatobiliary Surgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medcine, Haikou, China
| |
Collapse
|
19
|
Inoue T, Koyama F, Iwasa Y, Sho M. Combined Endoscopic Submucosal Dissection and Transanal Minimally Invasive Surgery for Rectal Laterally Spreading Tumor after Prior Transanal Resection: A Case Report. Surg Case Rep 2025; 11:24-0166. [PMID: 40443959 PMCID: PMC12122182 DOI: 10.70352/scrj.cr.24-0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 05/14/2025] [Indexed: 06/02/2025] Open
Abstract
INTRODUCTION Endoscopic submucosal dissection (ESD) is an effective procedure for resecting noninvasive colorectal neoplasms. However, submucosal fibrosis affects the technical difficulty of ESD. We experienced a combined ESD and transanal minimally invasive surgery (TAMIS) for a rectal neoplasm with submucosal fibrosis. CASE PRESENTATION We report our experience with a 75-year-old woman who had a rectal laterally spreading tumor with scarring from a prior transanal resection. She underwent combined ESD and TAMIS to overcome the procedural difficulty of ESD for submucosal fibrosis. The portion of the bowel without scarring was dissected using ESD, while the portion with scarring was dissected using TAMIS. A successful en bloc resection of the tumor was achieved, and there was no recurrence. CONCLUSIONS Based on the findings from this case, the combination of ESD and TAMIS may be particularly effective under conditions such as rectal neoplasms with submucosal fibrosis.
Collapse
Affiliation(s)
- Takashi Inoue
- Department of Surgery, Saiseikai Chuwa Hospital, Sakurai, Nara, Japan
- Department of Surgery, Nara Medical University, Kashihara, Nara, Japan
- Department of Endoscopy, Nara Medical University Hospital, Kashihara, Nara, Japan
| | - Fumikazu Koyama
- Department of Surgery, Nara Medical University, Kashihara, Nara, Japan
- Department of Endoscopy, Nara Medical University Hospital, Kashihara, Nara, Japan
| | - Yosuke Iwasa
- Department of Surgery, Nara Medical University, Kashihara, Nara, Japan
- Department of Endoscopy, Nara Medical University Hospital, Kashihara, Nara, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
20
|
Pal S, Shimshoni E, Torres SF, Kong M, Tai K, Sangwan V, Bertos N, Bailey SD, Bérubé J, Ingber DE, Ferri L. Patient-derived esophageal adenocarcinoma organ chip: a physiologically relevant platform for functional precision oncology. J Transl Med 2025; 23:577. [PMID: 40410759 PMCID: PMC12102830 DOI: 10.1186/s12967-025-06593-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 05/08/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Esophageal adenocarcinoma (EAC) is the sixth most deadly cancer worldwide, with increasing incidence in North America. As no targeted therapy or immunotherapy has revolutionized the management of EAC, chemotherapy is the only standard of care. Most patients with EAC experience poor outcomes because of the inherent or acquired resistance to chemotherapy. METHODS Adapting a patient-centered approach, we leveraged a microfluidic cell culture technology platform (Emulate), organoids derived from treatment-naive patient tumors or adjacent normal tissues, and patient-matched cancer-associated or normal fibroblasts respectively, to develop a novel, physiologically relevant, high-fidelity preclinical esophagus-on-a-chip model. H&E, immunofluorescence staining, live/dead assay, LDH assay, and ELISA-based detection of tumor biomarkers were used to assess treatment responses. RESULTS Each patient-specific stroma-inclusive microfluidic esophageal adenocarcinoma on-a-chip (EAC chip) faithfully recreates the tumor-stroma interface while preserving the full diversity of two cell types (epithelia and fibroblasts), genetic landscapes and histological architecture of the source tumors. EAC chips also accurately predict the response to neoadjuvant chemotherapy (NACT) within a clinically useful timeframe (approx. 12 days). A docetaxel-based triplet chemotherapy regimen matched with the treatment of the source patient was successfully perfused through the interstitial space within this model. Therefore, EAC chips more accurately recapitulate inpatient pathological and objective responses than the corresponding static 3D-organoid-only cultures. CONCLUSIONS Overall, this model is an effective tool for predicting patients' responses to chemotherapy and testing tumor- or stroma-targeted alternative therapies. Moreover, these high-fidelity, low-throughput EAC chips effectively complement high-throughput PDO culture-based drug testing and provide improved insights into drug efficacy before human studies.
Collapse
Affiliation(s)
- Sanjima Pal
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Elee Shimshoni
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, USA
| | | | - Mingyang Kong
- Department of Surgical and Interventional Sciences, McGill University, Montreal, Canada
| | - Kulsum Tai
- Department of Surgical and Interventional Sciences, McGill University, Montreal, Canada
| | - Veena Sangwan
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Surgical and Interventional Sciences, McGill University, Montreal, Canada
| | - Nicholas Bertos
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Swneke Donovan Bailey
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Surgical and Interventional Sciences, McGill University, Montreal, Canada
| | - Julie Bérubé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, USA
| | - Lorenzo Ferri
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Department of Surgical and Interventional Sciences, McGill University, Montreal, Canada.
| |
Collapse
|
21
|
Chopko TC, Maroun JW, Reisenauer JS, Tapias LF. Sentinel Lymph Node Mapping in Esophageal Cancer: Current Status and Future Directions. Ann Surg Oncol 2025:10.1245/s10434-025-17479-3. [PMID: 40402425 DOI: 10.1245/s10434-025-17479-3] [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: 02/24/2025] [Accepted: 04/28/2025] [Indexed: 05/23/2025]
Abstract
OBJECTIVE This review provides a comprehensive discussion about the importance of adequate lymphadenectomy, its anatomic and oncologic significance, principles and rationale of sentinel lymph node mapping, current evidence stratified by tracer substrate, challenges, and future directions. Esophageal cancer has one of the worst cancer-related survival rates, and nodal status is the single most significant prognostic factor. Submucosal penetration generally demands esophagectomy, often following neoadjuvant therapy in the presence of deeper extension. Guidelines recommend resecting ≥15 lymph nodes. Variability in surgical approach and dissection in concert with aberrant esophageal lymphatic anatomy make adequate lymphadenectomy difficult. METHODS A narrative review was conducted to explore existing literature regarding lymphadenectomy with its requisite anatomic and oncologic significance in esophageal cancer, as well as the rationale for and present state of sentinel lymph node mapping stratified by substrates. Tables and figures were constructed by the authors using Microsoft Office applications and Biorender software, respectively. RESULTS Sentinel lymph node mapping exploits the tumoral lymphatic network to identify the nodes most prone to metastasis, directing further dissection. Targeting sentinel lymph nodes with dyes, radiotracers, or hybrid tracers can assist surgeons with lymphadenectomy, potentially improving staging accuracy and personalizing care to individual anatomy. CONCLUSIONS While this approach would benefit from larger studies and long-term data, early evaluations suggest improved detection of metastases outside the en bloc field without significantly increasing morbidity.
Collapse
Affiliation(s)
| | | | - Janani S Reisenauer
- Division of Thoracic Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Luis F Tapias
- Division of Thoracic Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
22
|
Chuang CH, Chen CT, Hsu CH, Shao YY. Carry-over effect of immunotherapy in patients with advanced hepatocellular carcinoma. Cancer Immunol Immunother 2025; 74:208. [PMID: 40377687 DOI: 10.1007/s00262-025-04052-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] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 04/10/2025] [Indexed: 05/18/2025]
Abstract
BACKGROUND Combination immunotherapy is the current standard for treating advanced hepatocellular carcinoma (HCC). The response elicited by upfront immune checkpoint inhibitors (ICIs) might influence the efficacy of salvage therapy, a phenomenon known as the carry-over effect. This effect is thought to stem from immune memory and sustained immune activation, providing extended protection against tumor progression and resulting in a durable response even after discontinuation of ICI. This study aimed to investigate the carry-over effect of first-line ICI therapy in patients with advanced HCC. METHODS Patients who received first-line ICI therapy for advanced HCC from December 2017 to December 2021 were included if they exhibited disease progression and received second-line systemic therapy. We analyzed the associations between clinical benefit (classified as complete, partial response and stable disease) of first-line ICI therapy, post-progression survival (PPS) and second-line progression-free survival (PFS). We used a historical cohort of patients receiving first-line multikinase inhibitor (MKI) for comparison. RESULTS A total of 137 patients were analyzed. We included 60 patients who received first-line ICI therapy, of which clinical benefit was detected in 46 (76.7%). Compared with patients without clinical benefit of first-line ICI therapy, patients with clinical benefit exhibited significantly longer PPS (median: 14.6 vs. 4.9 months, P = 0.024) and second-line PFS (median: 3.6 vs. 1.6 months, P = 0.027). In multivariate analysis, clinical benefit of first-line ICI therapy remained an independent predictor of PPS [hazard ratio (HR): 0.295, P = 0.005] and second-line PFS (HR: 0.484, P = 0.047). Conversely, clinical benefit was not associated with PPS among patients receiving first-line MKI therapy in both univariate and multivariate analysis in historical MKI cohort. CONCLUSIONS Clinical benefit of first-line ICI therapy was associated with PPS and second-line PFS in patients with advanced HCC, suggestive of the carry-over effect of ICI.
Collapse
Affiliation(s)
- Chien-Huai Chuang
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 10002, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Tso Chen
- Department of Oncology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 10002, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Chih-Hung Hsu
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 10002, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Yun Shao
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan.
- Department of Oncology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 10002, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
23
|
Seiki Y, Ikezawa K, Kai Y, Takada R, Kawabata M, Kishimoto H, Hosokawa K, Watsuji K, Kozumi K, Urabe M, Mukai K, Nakabori T, Ohkawa K. Impact of zinc deficiency on the prognosis of unresectable pancreatic cancer. Pancreatology 2025:S1424-3903(25)00092-4. [PMID: 40410049 DOI: 10.1016/j.pan.2025.05.007] [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: 03/04/2025] [Revised: 04/28/2025] [Accepted: 05/15/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND/OBJECTIVES Pancreatic cancer (PC) is an aggressive malignancy with a poor prognosis. Zinc deficiency (ZD) is prevalent among patients with cancer, especially those with PC, owing to factors such as malabsorption and systemic inflammation. Although ZD is associated with poor nutritional status and cancer cachexia, its impact on overall survival (OS) in unresectable PCs remains unclear. This study aimed to evaluate the prevalence of ZD in patients with unresectable PC and its influence on prognosis. METHODS A retrospective study was conducted on 272 patients with unresectable pancreatic cancer whose serum zinc levels measured before starting chemotherapy between April 2016 and March 2024. ZD was defined as serum zinc level below 60 μg/dL. Patient characteristics, nutritional indices (modified Glasgow prognostic score [mGPS], neutrophil-to-lymphocyte ratio [NLR], prognostic nutritional index [PNI], controlling nutritional status [CONUT] score), and survival outcomes were analyzed. RESULTS ZD was identified in 19.4 % of patients. Patients with ZD were significantly older and exhibited lower PNI, indicating poorer nutritional status. Regarding OS, univariate analysis showed that the ZD group had significantly lower survival rates than non-ZD group. Multivariate analysis revealed that ZD was an independent predictor of poor prognosis. CONCLUSIONS This study demonstrates that ZD before chemotherapy is an independent prognostic factor in patients with unresectable PC undergoing chemotherapy. Considering its association with poor nutritional status, serum zinc levels may serve as a valuable prognostic biomarker. Further research is warranted to explore the potential therapeutic benefits of zinc supplementation therapy.
Collapse
Affiliation(s)
- Yusuke Seiki
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan.
| | - Yugo Kai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Ryoji Takada
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Masaki Kawabata
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroki Kishimoto
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kana Hosokawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Ko Watsuji
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuhiro Kozumi
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Makiko Urabe
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kaori Mukai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Tasuku Nakabori
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
24
|
Wu J, Du X, He Y, Xian S. Comparison of outcomes between surgery and non-surgery after conversion therapy for advanced gastric cancer with unresectable factors: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:371. [PMID: 40369412 PMCID: PMC12080213 DOI: 10.1186/s12876-025-03969-x] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Advanced gastric cancer (AGC) with unresectable factors presents a significant treatment challenge. Conventional treatments such as systemic chemotherapy, radiotherapy, and immunotherapy can delay disease progression but often yield limited outcomes. For stage III-IV gastric cancer with unresectable factors, conversion therapy based on chemotherapy can achieve tumor downstaging, providing a subset of patients with the opportunity for curative surgery. However, the efficacy of multimodal approaches combining chemotherapy, with or without immunotherapy, and conversion surgery compared to chemotherapy alone remains controversial. METHODS We conducted a systematic review and meta-analysis of high-quality studies published between January 2014 and November 2024, assessing the role of surgery following conversion therapy in advanced gastric cancer. Relevant studies were retrieved from PubMed, Embase, and Web of Science databases. All included studies were observational; no randomized trials were available. Clinical data, including overall survival (OS), progression-free survival (PFS), objective response rate (ORR) and adverse event (AE) rates, were analyzed using RevMan 5.4. RESULTS Twelve observational cohort studies were included. Conversion surgery(CS) was associated with improved 1-year, 3-year, and 5-year OS rates (RR 0.38, 95% CI: 0.31-0.47; RR 0.64, 95% CI: 0.54-0.76; RR 0.77, 95% CI: 0.65-0.91, respectively) and increased 1-year and 3-year PFS rates (RR 0.57, 95% CI: 0.49-0.99; RR 0.67, 95% CI: 0.57-0.78, respectively). No significant difference in AE rates was observed between groups. CONCLUSIONS Conversion surgery following chemotherapy in stage III-IV gastric cancer is associated with improved OS and PFS in observational studies. However, these findings may reflect inherent prognostic differences between groups, as surgery was only feasible for chemotherapy responders. Prospective trials are needed to validate causality.
Collapse
Affiliation(s)
- Jiaheng Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xuetian Du
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yiqiang He
- Department of Gastrointestinal Surgery, Guangxi Nationalities Hospital, Guangxi, China
| | - Shulin Xian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
25
|
Numata Y, Fujii T, Toda C, Okumura T, Manabe T, Takeda N, Shimizu T, Tabuchi Y, Fujii T, Sakai H. Digoxin promotes anoikis of circulating cancer cells by targeting Na +/K +-ATPase α3-isoform. Cell Death Dis 2025; 16:373. [PMID: 40350473 PMCID: PMC12066707 DOI: 10.1038/s41419-025-07703-z] [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: 11/13/2024] [Revised: 04/19/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
Circulating cancer cells (CCCs) are closely related to the process of distant metastasis. In early step of the metastasis cascade, CCCs must evade the detachment-induced cell death (anoikis) for their survival. Here, we examined whether Na+/K+-ATPase α3-isoform (α3NaK) in CCCs contributes to avoidance of anoikis. In CCCs isolated from gastric cancer patients, α3NaK was predominantly localized in the plasma membrane (PM), but it moved to the cytoplasm when the CCCs were attached to culture dishes. The CCCs showed significant expression of integrin α5 but not fibronectin, one of components of the extracellular matrix (ECM). In human gastric cancer MKN45 cells, digoxin (20 and 50 nM), a cardiac glycoside, significantly inhibited the enzyme activity and translocation (from cytoplasm to PM) of α3NaK, while they had no significant effect on ubiquitous Na+/K+-ATPase α1-isoform (α1NaK) in the PM. The translocation of α3NaK required the loss of ECM components from the cells. Additionally, digoxin significantly enhanced caspase 3/7 activity, as well as the expression of cleaved caspase 3, while reducing the viability of detached (floating) cells. In the MKN45 xenograft mouse model, intraperitoneal administration of digoxin (2 mg/kg/day) significantly decreased the number of CCCs and suppressed their liver metastasis. Our results suggest that α3NaK plays an essential role in the survival of CCCs in gastric cancer, and that digoxin enhances anoikis in detached (metastatic) gastric cancer cells by inhibiting the α3NaK translocation from cytoplasm to PM, thereby reducing CCCs. Targeting α3NaK may be a promising therapeutic strategy against CCC survival.
Collapse
Affiliation(s)
- Yoshihisa Numata
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Takuto Fujii
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| | - Chihiro Toda
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Tomoyuki Okumura
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Takahiro Manabe
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Naoya Takeda
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan
| | - Takahiro Shimizu
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
| | - Tsutomu Fujii
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, 930-0194, Japan.
| | - Hideki Sakai
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| |
Collapse
|
26
|
Meng ZN, Chen JY, Yu C, Zheng AH, Reddy OS, Liu KY, Su YR, Zhang ST, Wang YS, Gu HY, Wang FW, Xu SC, Sun LT, Chen BC, Lai WF, Wu GQ, Zhang DH. A gelable polymer loaded with curcumin and apatinib absorbed in gelatin sponge delays postoperative residual tumor growth. Sci Rep 2025; 15:16375. [PMID: 40350449 PMCID: PMC12066724 DOI: 10.1038/s41598-025-97732-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Surgical resection of the tumor remains the preferred treatment for most solid tumors at an early stage, however, residual tumor cells after surgical resection poses a considerable obstacle in cancer treatment. Here, we developed a gel carrier using a cellulose-based gel-forming polymer (CT) combined with gelatin sponge (GS) to fill the resection cavity and delay postoperative residual tumor growth. The fabricated gel exhibited a porous nature along with gradual swelling and erosion over time. Curcumin (Cur) and apatinib (Apa) were loaded into CT gel (CT-CA), and a sustained release behavior was observed at pH 7.4 and 6.4 at 37 °C. The preclinical studies indicated that the mouse weight and tissue exhibited no apparent change after administration of the GS-CT compared with the control. The in vivo fluorescence images showed that GS-CT has the capability to regulate the release of Cur and Apa, facilitating the accumulation of these two agents at the surgical tumor site. Moreover, GS-CT loaded Cur and Apa (GS-CT-CA) delayed postoperative residual tumor growth in intraperitoneal and subcutaneous postoperative mouse models. These findings demonstrated that our gel carrier system significantly prevents postoperative residual tumor growth because of enhanced drug accumulation and sustained drug release at the tumor site.
Collapse
Affiliation(s)
- Zhuo-Nan Meng
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jian-Yuan Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chong Yu
- Department of Urology, Urology & Nephrology Center, Zhejiang Provincial People'sHospital, Affiliated People's Hospital, Hangzhou Medical College, No. 138 ShangTang Road, Hangzhou, 310014, China
| | - Ai-Hong Zheng
- Department of Medical Oncology, Cancer Center, Zhejiang Provincial People's Hospital(Affiliated People's Hospital, Hangzhou Medical College), No. 138 ShangTang Road, Hangzhou, 310014, China
| | - O Sreekanth Reddy
- Department of Urology, Urology & Nephrology Center, Zhejiang Provincial People'sHospital, Affiliated People's Hospital, Hangzhou Medical College, No. 138 ShangTang Road, Hangzhou, 310014, China
| | - Kai-Yan Liu
- Department of Urology, Urology & Nephrology Center, Zhejiang Provincial People'sHospital, Affiliated People's Hospital, Hangzhou Medical College, No. 138 ShangTang Road, Hangzhou, 310014, China
| | - Yong-Rui Su
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shi-Tai Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yin-Shuang Wang
- Department of Ultrasound Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang Province, China
| | - Hang-Yu Gu
- Department of Oncology and Hematology, Beilun District People's Hospital, Ningbo, China
| | - Fu-Wei Wang
- Department of Medical Oncology, Cancer Center, Zhejiang Provincial People's Hospital(Affiliated People's Hospital, Hangzhou Medical College), No. 138 ShangTang Road, Hangzhou, 310014, China
| | - Song-Cheng Xu
- Department of Ultrasound, Zhejiang Provincial People's Hospital (Affiliated People'sHospital, Hangzhou Medical College), No. 138 ShangTang Road, Hangzhou, 310014, China
| | - Li-Tao Sun
- Department of Ultrasound, Zhejiang Provincial People's Hospital (Affiliated People'sHospital, Hangzhou Medical College), No. 138 ShangTang Road, Hangzhou, 310014, China
| | - Bing-Chen Chen
- Department of Anal Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Wing-Fu Lai
- Department of Urology, Urology & Nephrology Center, Zhejiang Provincial People'sHospital, Affiliated People's Hospital, Hangzhou Medical College, No. 138 ShangTang Road, Hangzhou, 310014, China.
- School of Food Science and Nutrition, University of Leeds, Leeds, LS29JT, UK.
| | - Guo-Qing Wu
- Department of Medical Oncology, Cancer Center, Zhejiang Provincial People's Hospital(Affiliated People's Hospital, Hangzhou Medical College), No. 138 ShangTang Road, Hangzhou, 310014, China.
| | - Da-Hong Zhang
- Department of Urology, Urology & Nephrology Center, Zhejiang Provincial People'sHospital, Affiliated People's Hospital, Hangzhou Medical College, No. 138 ShangTang Road, Hangzhou, 310014, China.
| |
Collapse
|
27
|
Huang J, Chen C, Shen YM, Luo YF, Sun ZM, Chen J, Xu SJ, Lin JH, Chen SC. Preoperative immune prognostic index predicts the prognosis and postoperative adjuvant chemotherapy benefits of esophageal squamous cell carcinoma after minimally invasive esophagectomy. BMC Gastroenterol 2025; 25:344. [PMID: 40340583 PMCID: PMC12060512 DOI: 10.1186/s12876-025-03959-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND The utility of the immune prognostic index (IPI) for esophageal squamous cell carcinoma (ESCC) has yet to be established after minimally invasive esophagectomy (MIE). The purpose of this study was to investigate the value of IPI in predicting the prognosis and postoperative adjuvant chemotherapy (AC) benefits of ESCC patients. METHODS Between January 2011 and December 2018, 613 ESCC patients underwent MIE at our center and were divided into two groups: low IPI and high IPI.Log-rank tests were used to compare the overall survival (OS) and disease-free survival (DFS) of patients in different groups based on Kaplan-Meier survival analysis. Differences in clinical characteristics between groups were eliminated by propensity score matching (PSM) analysis. To identify independent risk factors influencing OS and DFS, the Cox proportional risk model was used. RESULTS In comparison to the high IPI group, the low IPI group had a better 5-year OS and DFS in both the entire and matched cohorts (P < 0.05). IPI was found to be an independent prognostic factor for OS and DFS in a multivariate analysis of the entire cohort and the matched cohort (P < 0.05). In subgroup analyses of most clinicopathological factors, high IPI was associated with a higher risk of death or recurrence in the matched cohorts. When combined with 8th TNM staging, the 5-year OS and DFS of stage II or III patients with low IPI in the AC group were not different from those in the non-AC group (P > 0.05), and AC of stage III patients with high IPI significantly prolonged 5-year OS and DFS (OS: 37.4% vs 26.2%, P = 0.018; DFS: 33.6% vs 19.8%, P = 0.042). CONCLUSION Preoperative IPI is a promising predictor of ESCC after MIE. For stage III ESCC patients with high IPI, AC can significantly reduce the risk of death or recurrence.
Collapse
Affiliation(s)
- Jin Huang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Chao Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Yan-Ming Shen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Yun-Fan Luo
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Zhao-Min Sun
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Jie Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Shao-Jun Xu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, Fuzhou, 350001, Fujian Province, China.
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian Province, China.
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, 350001, Fujian Province, China.
| | - Ji-Hong Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, Fuzhou, 350001, Fujian Province, China.
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian Province, China.
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, 350001, Fujian Province, China.
| | - Shu-Chen Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No.29 Xin Quan Road, Fuzhou, 350001, Fujian Province, China.
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian Province, China.
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, 350001, Fujian Province, China.
| |
Collapse
|
28
|
Zou X, Wang Y. Predictive value of osteopenia as prognostic marker for survival and recurrence in patients with gastrointestinal cancers: a systematic review and meta-analysis. Front Med (Lausanne) 2025; 12:1527829. [PMID: 40375934 PMCID: PMC12078326 DOI: 10.3389/fmed.2025.1527829] [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: 11/13/2024] [Accepted: 03/10/2025] [Indexed: 05/18/2025] Open
Abstract
Background Early detection, systematic prevention, and personalized therapy are crucial to reduce mortality in patients with gastrointestinal (GI) cancers. This systematic review and meta-analysis aimed to clarify the predictive value of osteopenia and osteosarcopenia as prognostic markers of survival and recurrence in patients with GI cancers. Methods Medline, Google Scholar, and Science Direct databases were searched for English-language studies that included patients who underwent surgical resection following a pathologically diagnosed GI cancer and reported the association between osteopenia and osteosarcopenia on the overall survival (OS) and recurrence-free survival (RFS). Meta-analysis was done using STATA 14.2, and the results were reported as pooled hazard ratios (HR) with 95% confidence intervals (CI). Heterogeneity was assessed using the I2 statistic and the Chi-square test. Study quality was evaluated using the Newcastle Ottawa Scale (NOS). Results A comprehensive literature search yielded 23 eligible studies, primarily from Japan. Osteopenia emerged as a significant risk factor for both OS (pooled HR 2.20, 95% CI: 1.74-2.79) and RFS (pooled HR 2.15, 95% CI: 1.60-2.89). Patients with osteosarcopenia exhibited threefold higher mortality rates (pooled HR 2.96, 95% CI: 1.99-4.40) and heightened risk of recurrence (pooled HR 2.75, 95% CI: 1.79-4.24). Subgroup analyses underscored the consistency of these associations across diverse contexts. Conclusion This meta-analysis establishes osteopenia and osteosarcopenia as robust prognostic indicators for survival and recurrence in GI cancers. Integrating musculoskeletal assessments into routine oncological care is imperative for timely interventions and optimized patient outcomes.
Collapse
Affiliation(s)
| | - Yang Wang
- Ward 13 (Respiratory Digestive Geriatrics), Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| |
Collapse
|
29
|
Seo WJ, Lee S, Jang YJ, Choi SI, Kim JH. Lymphatic invasion as the solitary risk factor for extraperigastric lymph node metastasis in early gastric cancer. Surgery 2025; 181:109157. [PMID: 39919694 DOI: 10.1016/j.surg.2025.109157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Lymph node metastasis, particularly to extraperigastric lymph nodes, is an important issue in determining the suitability of minimally invasive treatment for early gastric cancer. However, there is a paucity of research focused on identifying risk factors for extraperigastric lymph node metastasis, which is crucial for optimizing treatment selection. METHODS Patients who underwent radical gastrectomy for early gastric cancer between 2007 and 2023 at Korea University Guro Hospital and Kyung Hee University Hospital at Gandong in Seoul, Korea, were reviewed retrospectively. We compared clinicopathologic characteristics to find out risk factors for lymph node metastasis between the no lymph node metastasis, perigastric lymph node metastasis, and extraperigastric lymph node metastasis groups. RESULTS A total of 1,021 patients were included in the study, with 90.0% in the lymph node metastasis, 7.94% in perigastric lymph node metastasis, and 2.06% in extraperigastric lymph node metastasis groups. Clinicopathologic analysis revealed the lymph node metastasis group exhibited a greater prevalence of mucosal lesions (55.2%), whereas the perigastric lymph node metastasis and extraperigastric lymph node metastasis groups demonstrated a greater incidence of SM3 invasion (55.6% and 52.4%, respectively; P < .001). Lymphatic invasion were found to be significantly different among the groups (P < .001). Multinomial logistic regression identified lymphatic invasion as a sole significant risk factor for the perigastric lymph node metastasis and extraperigastric lymph node metastasis groups. The 5-year relapse-free survival rates were 99.0% (lymph node metastasis), 94.5% (perigastric lymph node metastasis), and 100% (extraperigastric lymph node metastasis) (P = .011). CONCLUSION This multicenter retrospective analysis indicates that lymphatic invasion is the primary independent predictor of extraperigastric lymph node metastasis in patients with early gastric cancer. Future prospective research is warranted to validate this result and establish guidelines for treatment decision-making for early gastric cancer.
Collapse
Affiliation(s)
- Won Jun Seo
- Division of Foregut Surgery, Department of Surgery, Korea University College of Medicine, Seoul, South Korea
| | - Sangjun Lee
- Department of Surgery, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| | - You-Jin Jang
- Division of Foregut Surgery, Department of Surgery, Korea University College of Medicine, Seoul, South Korea
| | - Sung Il Choi
- Department of Surgery, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| | - Jong-Han Kim
- Division of Foregut Surgery, Department of Surgery, Korea University College of Medicine, Seoul, South Korea.
| |
Collapse
|
30
|
Weng CM, Zhong Q, Sun YQ, Liu ZY, Ma YB, Zhang ZQ, Zhang HX, Zhu JY, Ye W, Wu J, Du H, Zheng CH, Li P, Chen QY, Huang CM, Xie JW. A novel ypN-TRG staging system for gastric cancer patients after neoadjuvant therapy based on the metro-ticket paradigm: a multicenter and large sample retrospective analysis. Gastric Cancer 2025; 28:465-477. [PMID: 39918688 DOI: 10.1007/s10120-025-01586-x] [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: 10/14/2024] [Accepted: 01/15/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Conventional ypT category evaluates the depth of invasion after neoadjuvant therapy (NAT) for gastric cancer (GC) and has limited prognostic value. Tumor regression grade (TRG) measures the extent of tumor response to treatment, and when combined with the ypN category, it may enhance the prediction of patient outcomes. This study aims to develop a new staging system by integrating TRG and ypN category to better evaluate the prognosis of GC patients receiving NAT. METHODS This retrospective analysis included 962 patients who underwent radical gastrectomy after NAT, with 513 in the development cohort (from one center) and 449 in the external validation cohort (from five centers). The ypN-TRG staging system was established by calculating the distance from the origin on a cartesian plane incorporating the ypN (x-axis) stage and TRG (y-axis) grade, and five sub-stages were delineated. RESULTS In the development cohort, 3-year overall survival rates according to ypN-TRG stage I, IIA, IIB, IIIA, IIB were 87.6%, 80.2%, 70.7%, 47.3%, 21.5%, p < 0.01. Compared with ypTNM, the ypN-TRG staging system performed better in terms of the prognostic discrimination power (C-index), goodness-of-fit (AIC, BIC), model improvement (NRI, IDI), and model stability (time-AUC). Multivariate Cox regression analysis confirmed the superiority of ypN-TRG over ypTNM staging. In the external validation cohort, ypN-TRG staging was a better predictor of OS and DFS in patients with GC. CONCLUSIONS The ypN-TRG staging system is superior to the AJCC eighth edition ypTNM staging system in accurately assessing the prognosis of patients with GC after NAT.
Collapse
Affiliation(s)
- Cai-Ming Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Yu-Qin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, Fujian, China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai, China
| | - Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Hao-Xiang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Ji-Yun Zhu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Wen Ye
- Department of Gastrointestinal Surgery, Longyan First Hospital Affiliated to, Fujian Medical University, Longyan, 364000, Fujian, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116000, Liaoning, China
| | - He Du
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
31
|
Lewis KA, Diggs LP, Badgwell BD. Educational Review: Updates on Therapeutic Strategies for Gastric Cancer with Peritoneal Metastasis. Ann Surg Oncol 2025; 32:3672-3687. [PMID: 40016614 DOI: 10.1245/s10434-025-17069-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 02/09/2025] [Indexed: 03/01/2025]
Abstract
Gastric cancer (GC) commonly presents in advanced stages with metastatic spread to the peritoneal cavity, and outcomes associated with gastric cancer with peritoneal metastasis (GCPM) continue to carry a dismal prognosis. Persistent challenges in the detection of peritoneal metastasis (PM) have resulted in a relative paucity of high-quality data to inform management decisions. Several consensus groups have published recommendations to guide management, including most recently the National Comprehensive Cancer Network guidelines, which now include cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) as a potential treatment modality in select patients with GCPM. Multiple clinical trials have investigated the use of CRS/HIPEC and other peritoneal-directed therapies, such as intraperitoneal chemotherapy (IPC) and pressurized intraperitoneal aerosolized chemotherapy (PIPAC). As high-volume centers work to incorporate such therapies into their practice, ongoing clinical trials are aimed at understanding their efficacy. Recent findings have improved understanding of the mechanisms and pathophysiology underlying GCPM while the discovery of novel targets offers potential for drug development and therapeutic strategies to overcome treatment resistance. This review highlights recent advancements and addresses the persistent challenges in managing GCPM while also offering a comprehensive summary of current guidelines and treatment strategies.
Collapse
Affiliation(s)
- Kever A Lewis
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Laurence P Diggs
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian D Badgwell
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
32
|
Zhao Z, Huang W, Fu Y, Ji W, Xu J, Chen F, Chen Z, Huang Z. Comparative analysis of cis-cutting and retro-cutting techniques in gastric tube fabrication for preventing postoperative anastomotic leakage after esophagectomy. Surgery 2025; 181:109124. [PMID: 39884217 DOI: 10.1016/j.surg.2024.109124] [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: 08/06/2024] [Revised: 11/13/2024] [Accepted: 12/25/2024] [Indexed: 02/01/2025]
Abstract
OBJECTIVE The construction of a gastric-tube-substitute esophagus is a common method for digestive tract reconstruction after esophagectomy. However, the incidence of postoperative anastomotic leakage remains high. This study aims to compare the effectiveness of 2 gastric tube fabrication methods-cis-cutting and retro-cutting-in reducing postoperative anastomotic leakage. METHODS We retrospectively analyzed 253 patients who underwent McKeown radical esophagectomy for esophageal cancer at the Second Affiliated Hospital of Fujian Medical University from February 2021 to February 2024. Patients were divided into cis-cut (n = 126) and retro-cut (n = 127) groups on the basis of the surgical technique used. The incidence of anastomotic leakage was compared, and logistic regression was used to identify risk factors. RESULTS Anastomotic leakage (16.7%, 21/126 vs 3.9%, 5/127; P < .001) and stricture (28.6%, 36/126 vs 15%, 19/127; P = .009) rates were lower in the retro-cut group than the cis-cut group. Multivariable analysis identified the gastric tube fabrication method in the cis-cut group as an independent risk factor (odds ratio, 3.390; 95% confidence interval, 1.147-10.018; P = .027) for postoperative anastomotic leakage. CONCLUSION Retro-cut gastric tube fabrication significantly reduces the incidence of anastomotic leakage and anastomotic stricture, suggesting its suitability as a standard technique for esophageal reconstruction.
Collapse
Affiliation(s)
- Zhihuang Zhao
- The Second Clinical College of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Wenbo Huang
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - YuXiang Fu
- The Second Clinical College of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Wei Ji
- The Second Clinical College of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jie Xu
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Feng Chen
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Zhiyao Chen
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - ZhiJun Huang
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| |
Collapse
|
33
|
Marzioni M, Maroni L, Aabakken L, Carpino G, Groot Koerkamp B, Heimbach J, Khan S, Lamarca A, Saborowski A, Vilgrain V, Nault JC. EASL Clinical Practice Guidelines on the management of extrahepatic cholangiocarcinoma. J Hepatol 2025:S0168-8278(25)00162-X. [PMID: 40348685 DOI: 10.1016/j.jhep.2025.03.007] [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: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 05/14/2025]
Abstract
Recent years have witnessed significant advances in the imaging, molecular profiling, and systemic treatment of cholangiocarcinoma (CCA). Despite this progress, the early detection, precise classification, and effective management of CCA remain challenging. Owing to recent developments and the significant differences in CCA subtypes, EASL commissioned a panel of experts to draft evidence-based recommendations on the management of extrahepatic CCA, comprising distal and perihilar CCA. Particular attention is given to the need for accurate classification systems, the integration of emerging molecular insights, and practical strategies for diagnosis and treatment that reflect real-world clinical scenarios.
Collapse
|
34
|
Du WF, Liang TS, Guo ZF, Li JJ, Yang CG. Comparison of outcomes of laparoscopic-assisted and total laparoscopic right hemicolectomy for right-sided colon cancer. World J Gastrointest Surg 2025; 17:100476. [PMID: 40291870 PMCID: PMC12019061 DOI: 10.4240/wjgs.v17.i4.100476] [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: 08/18/2024] [Revised: 01/13/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Colon cancer is a significant health issue in China, with high incidence and mortality rates. Surgical resection remains the primary treatment, with the introduction of complete mesocolic excision in 2009 improving precision and outcomes. Laparoscopic techniques, including laparoscopic-assisted right hemicolectomy (LARH) and total laparoscopic right hemicolectomy (TLRH), have further advanced colon cancer treatment by reducing trauma, blood loss, and recovery time. While TLRH offers additional benefits such as faster recovery and fewer complications, its adoption has been limited by longer operative times and technical challenges. AIM To compare the short-term outcomes of TLRH and LARH for the treatment of right -sided colon cancer and explore the advantages and feasibility of TLRH. METHODS Clinical data from 109 right-sided colon cancer patients admitted between January 2019 and May 2021 were retrospectively analyzed. Patients were divided into an observation group (TLRH, n = 50) and a control group (LARH, n = 59). Study variables were operation time, intraoperative bleeding volume, postoperative hospital stays, length of surgical specimen, number of lymph nodes dissected, and postoperative inflammatory factor levels of the two groups of patients. The postoperative complications were analyzed and compared, and survival, recurrence, and remote metastasis rates of the two groups were compared during a 2-year follow-up period. RESULTS The TLRH group showed the advantages of reduced intraoperative bleeding, shorter hospital stays, and quicker recovery. Lymph node dissection outcomes were comparable, and postoperative inflammatory markers were lower in the TLRH group. Complication rates were similar. Short-term follow-up (2 years) revealed no significant differences in recurrence, metastasis, or survival rates. CONCLUSION Compared to LARH, TLRH offers significant advantages in terms of reducing surgical trauma, lowering postoperative inflammatory factor levels, and mitigating the impact on intestinal function. This approach contributes to a shorter hospital stay and promotes postoperative recovery in patients. The study suggests that TLRH may offer favorable outcomes for colorectal cancer patients.
Collapse
Affiliation(s)
- Wen-Feng Du
- Department of Gastrointestinal Surgery, Liaocheng People’s Hospital, Liaocheng 252000, Shandong Province, China
| | - Tang-Shuai Liang
- Department of Gastrointestinal Surgery, Liaocheng People’s Hospital, Liaocheng 252000, Shandong Province, China
| | - Zong-Fei Guo
- College of Clinical and Basic Medicine, Shandong First Medical University, Liaocheng 252000, Shandong Province, China
| | - Jian-Jun Li
- Department of Gastrointestinal Surgery, Liaocheng People’s Hospital, Liaocheng 252000, Shandong Province, China
| | - Cheng-Gang Yang
- Department of Gastrointestinal Surgery, Liaocheng People’s Hospital, Liaocheng 252000, Shandong Province, China
| |
Collapse
|
35
|
Kijima Y, Hirata M, Nakazawa Y, Shinmura K, Hayashi N, Kijima R, Zaha H. Oncoplastic breast-conserving surgery using a modified Grisotti's flap for a patient with central breast cancer in a non-ptotic breast. Surg Today 2025:10.1007/s00595-025-03047-5. [PMID: 40278865 DOI: 10.1007/s00595-025-03047-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025]
Abstract
The treatment of patients with early breast cancer using breast-conserving surgery (BCS) commonly leads to local control and acceptable cosmetic results. We herein report a useful technique to achieve symmetry of the breast shape and size for a central lesion of the breast. A Japanese patient with early breast cancer located in the central area of the breast was enrolled in this study. Intraductal spread to the nipple was suspected. We resected the cylindrically shaped breast tissue with the nipple-areola complex and repaired the resected area using a modified Grisotti technique, a volume displacement technique using breast tissue with extra-breast tissue. The modified Grisotti flap technique during oncoplastic breast-conserving surgery (OPBCS) may thus be useful for patients who desire a symmetrical breast shape and size.
Collapse
Affiliation(s)
- Yuko Kijima
- Department of Breast Surgery, Fujita Health University, School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan.
- Nakagami Hospital, 610 Noborikawa, Okinawa, Okinawa, 904-2192, Japan.
| | - Munetsugu Hirata
- Department of Breast Surgery, Fujita Health University, School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
- Nakagami Hospital, 610 Noborikawa, Okinawa, Okinawa, 904-2192, Japan
| | - Yumika Nakazawa
- Department of Breast Surgery, Fujita Health University, School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
- Nakagami Hospital, 610 Noborikawa, Okinawa, Okinawa, 904-2192, Japan
| | - Kazuya Shinmura
- Department of Breast Surgery, Fujita Health University, School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
- Nakagami Hospital, 610 Noborikawa, Okinawa, Okinawa, 904-2192, Japan
| | - Naoki Hayashi
- Department of Breast Surgery, Fujita Health University, School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
- Nakagami Hospital, 610 Noborikawa, Okinawa, Okinawa, 904-2192, Japan
| | - Ryunosuke Kijima
- Department of Breast Surgery, Fujita Health University, School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
- Nakagami Hospital, 610 Noborikawa, Okinawa, Okinawa, 904-2192, Japan
| | - Hisamitsu Zaha
- Department of Breast Surgery, Fujita Health University, School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
- Nakagami Hospital, 610 Noborikawa, Okinawa, Okinawa, 904-2192, Japan
| |
Collapse
|
36
|
Fukuda K, Osumi H, Shimozaki K, Chin K, Ogura M, Fukuoka S, Udagawa S, Yoshino K, Tamba M, Wakatsuki T, Shinozaki E, Yamaguchi K, Ooki A. Impact of early tumor shrinkage on survival outcomes in patients with HER2-positive advanced gastric cancer treated with trastuzumab deruxtecan in third- or later-line settings. Therap Adv Gastroenterol 2025; 18:17562848251333538. [PMID: 40297207 PMCID: PMC12035244 DOI: 10.1177/17562848251333538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background Trastuzumab deruxtecan (T-DXd) has been approved for a third- or later-line treatment of HER2-positive advanced gastric cancer (AGC) in Japan. However, clinical data on the use of T-DXd in real-world practice remain insufficient. Although early tumor shrinkage (ETS) serves as an early on-treatment indicator of high treatment sensitivity, the use of ETS in predicting T-DXd efficacy remains unclear. Objectives This study aimed to evaluate the clinical efficacy and safety of T-DXd and investigate the clinical utility of ETS as a predictor of long-term efficacy and survival. Design Single-center retrospective cohort study. Methods This study consecutively enrolled patients with HER2-positive AGC who received T-DXd as a third- or later-line treatment between March 2018 and December 2023. Data on patient characteristics, adverse events (AEs), and clinical outcomes were obtained from electronic medical records. Clinical efficacy was assessed using progression-free survival (PFS) and overall survival (OS). In patients with measurable lesions, the overall response rate (ORR), ETS, and depth of response (DpR) were evaluated. Prognostic outcomes were assessed using the log-rank test and the Cox proportional hazards model. Results A total of 65 patients received T-DXd, with a median age of 66 years (range, 31-82 years); 77% had HER2 immunohistochemistry score of 3+, 71% received T-DXd as a third-line treatment, and 32% required initial dose reduction. At a median follow-up of 33.6 months, the median PFS and OS were 4.5 months and 7.7 months, respectively. Among the 47 patients with measurable lesions, the ORR was 36%. A median DpR of 15.8% was observed, with higher DpR correlating with longer OS. ETS was achieved in 38% of the patients and was an independent predictor of favorable PFS (hazard ratio (HR), 0.21; 95% confidence interval (CI), 0.09-0.49; p < 0.01) and OS (HR, 0.23; 95% CI, 0.10-0.52; p < 0.01). Longer second-line treatment duration was independently associated with improved OS. Overall, grade ⩾ 3 AEs occurred in 37% of the patients. Initial dose reduction reduced AE-induced discontinuation of treatment without compromising efficacy. Conclusion T-DXd demonstrated notable efficacy and a manageable safety profile in patients with HER2-positive AGC. Rapid and deep tumor shrinkage may have a significant impact on survival.
Collapse
Affiliation(s)
- Koshiro Fukuda
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroki Osumi
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Keitaro Shimozaki
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Keisho Chin
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Mariko Ogura
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Shota Fukuoka
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Shohei Udagawa
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Koichiro Yoshino
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Mikako Tamba
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Takeru Wakatsuki
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Eiji Shinozaki
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, Koto-ku, Tokyo, Japan
| | - Akira Ooki
- Department of Gastroenterological Chemotherapy, Japanese Foundation for Cancer Research, Ganken Ariake Byoin Cancer Institute Hospital, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan
| |
Collapse
|
37
|
Juthani R, Manne A. Blood-based biomarkers in pancreatic ductal adenocarcinoma: developments over the last decade and what holds for the future- a review. Front Oncol 2025; 15:1555963. [PMID: 40330826 PMCID: PMC12052548 DOI: 10.3389/fonc.2025.1555963] [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: 01/06/2025] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) accounts for a significant burden of global cancer deaths worldwide. The dismal outcomes associated with PDAC can be overcome by detecting the disease early and developing tools that predict response to treatment, allowing the selection of the most optimal treatment. Over the last couple of years, significant progress has been made in the development of novel biomarkers that aid in diagnosis, prognosis, treatment selection, and monitoring response. Blood-based biomarkers offer an alternative to tissue-based diagnosis and offer immense potential in managing PDAC. In this review, we have discussed the advances in blood-based biomarkers in PDAC, such as DNA (mutations and methylations), RNA, protein biomarkers and circulating tumor cells (CTC) over the last decade and also elucidated all aspects of practical implementation of these biomarkers in clinical practice. We have also discussed implementing multiomics utilizing more than one biomarker and targeted therapies that have been developed using these biomarkers.
Collapse
Affiliation(s)
- Ronit Juthani
- Department of Medicine, Saint Vincent Hospital, Worcester, MA, United States
| | - Ashish Manne
- Department of Internal Medicine, Division of Medical Oncology at the Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
38
|
Peng Y, Shi M, Xiong D, Lu S, Miao Y, Yuan H, Yan C, Zhu Z, Li B, Yang Z, Hu J. Preoperative assessment and prognostic prediction of gastric cancer patients with peritoneal metastasis using 18F-FDG PET/CT before conversion surgery. EJNMMI Res 2025; 15:46. [PMID: 40257553 PMCID: PMC12011703 DOI: 10.1186/s13550-025-01244-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/10/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Conversion therapy followed by conversion surgery (CS) can improve the prognosis of gastric cancer (GC) patients with peritoneal metastasis (PM). However, patients benefit differently. There is no way to confirm the prognostic benefit non-invasively and early. This retrospective study assessed the value of 18F-FDG PET/CT after conversion therapy in preoperative assessment and prognostic prediction of GC patients with PM. RESULTS Fifty-one GC patients with PM were enrolled. 18F-FDG PET/CT after conversion therapy helped in preoperative assessment. Its diagnostic accuracy for residual peritoneal lesions was slightly better than contrast-enhanced CT (72.5% vs. 61.2%, P = 0.229), although the difference was not statistically significant. TBR of peritoneal lesions could help preoperative assessment, with TBR of peritoneal lesions to the mediastinal blood pool SUVmax (TBRAmaxp) as the best predictor (cutoff = 0.705, specificity 80%, sensitivity 80%, AUC 0.825, P < 0.001). Additionally, PET/CT could predict prognosis and assess surgical benefit. SUVmax of peritoneal lesions (SUVmaxp) was the best predictor of 24 months survival (cutoff = 1.466, AUC 0.870, P = 0.002, Specificity 77.8%, Sensitivity 83.3%) and metabolic parameters of peritoneal lesions could predict OS and the prognosis of patients who underwent CS. CONCLUSION 18F-FDG PET/CT provides quantitative imaging indicators for preoperative evaluation and prognostic prediction in GC patients with PM.
Collapse
Affiliation(s)
- Yao Peng
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Daxi Xiong
- Department of Nuclear Medicine, Baoshan People's Hospital, Yunnan, China
| | - Sheng Lu
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Miao
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Yuan
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Yan
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenggang Zhu
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhongyin Yang
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiajia Hu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
39
|
He WZ, Yang YZ, Yin CX, Xian XY, Gu JM, Yi JH, Xue J, Zhao Y, Wang F, Hu WM, Xia LP. Evolution of HER2-low expression from primary to paired metastatic gastric cancer lesions. NPJ Precis Oncol 2025; 9:108. [PMID: 40234621 PMCID: PMC12000306 DOI: 10.1038/s41698-025-00881-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/15/2025] [Indexed: 04/17/2025] Open
Abstract
HER2-low expression has recently gained considerable attention as an actionable biomarker in gastric cancer. However, changes in HER2-low expression between primary and metastatic gastric cancers remain inadequately explored. This study included consecutive patients diagnosed with metastatic gastric cancer with both primary and metastatic tumors, between January 2014 and December 2023. HER2 status was evaluated in both primary and matched metastatic tumors. A total of 332 patients were enrolled, with HER2-negative, HER2-low, and HER2-positive statuses were observed in 226, 81, and 25 primary tumors, respectively, and in 175, 104, and 53 metastatic tumors, respectively. Among the 226 patients with HER2-negative primary tumors, 74 and 23 developed HER2-low and HER2-positive metastatic tumors, respectively. Conversion from HER2-negative primary to HER2-low metastatic gastric cancer was associated with metachronous and non-peritoneal metastasis. Overall, re-biopsy to evaluate HER2 status may be necessary, potentially broadening the patient population eligible for targeted HER2 therapy.
Collapse
Affiliation(s)
- Wen-Zhuo He
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Yuan-Zhong Yang
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Chen-Xi Yin
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Xin-Yi Xian
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Jia-Mei Gu
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Jia-Hong Yi
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Ju Xue
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Yue Zhao
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Fang Wang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| | - Wan-Ming Hu
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| | - Liang-Ping Xia
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| |
Collapse
|
40
|
Suzuki M, Sakurazawa N, Hagiwara N, Kogo H, Haruna T, Ohashi R, Yoshida H. Usefulness of shear-wave elastography for detection of lymph node metastasis in esophageal and gastric cancer. World J Gastrointest Oncol 2025; 17:101925. [PMID: 40235886 PMCID: PMC11995353 DOI: 10.4251/wjgo.v17.i4.101925] [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: 10/01/2024] [Revised: 01/17/2025] [Accepted: 02/05/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Tissue hardness is closely related to disease pathophysiology. Shear-wave elastography (SWE) is a simple and noninvasive ultrasound technique that has been used to evaluate the presence of lymph node metastases and differentiate between benign and malignant tumors. AIM To investigate SWE usefulness in measuring lymph node hardness to predict metastasis presence or absence in surgically removed lymph nodes. METHODS This observational study obtained data from patients who underwent surgery for esophageal or gastric cancer at Nippon Medical School Hospital. The hardness of the surgically removed lymph nodes was measured using SWE. The lymph nodes with hardness values ≥ 2.2 m/s were considered clinically positive for metastasis, whereas those with lower hardness values were considered clinically negative. The lymph nodes subsequently underwent pathological examination to determine the presence of metastasis, and the SWE results and pathological assessments were compared. RESULTS A total of 1077 lymph nodes were evaluated; 18 and 15 cases of esophageal and gastric cancer were identified, respectively. The optimal cutoff value for lymph node size was calculated to be 5.1 mm, and the area under the curve value was 0.74 (95% confidence interval: 0.69-0.84). When limited to a lymph node larger than the cut off value, the SWE sensitivity and specificity for metastasis identification were 0.76 and 0.82, respectively. CONCLUSION SWE was useful in detecting lymph node metastases in the upper gastrointestinal tract.
Collapse
Affiliation(s)
- Mikito Suzuki
- Department of Gastroenterological Surgery, Nippon Medical School, Tokyo 113-8603, Japan
| | - Nobuyuki Sakurazawa
- Department of Gastroenterological Surgery, Nippon Medical School, Tokyo 113-8603, Japan
| | - Nobutoshi Hagiwara
- Department of Gastroenterological Surgery, Nippon Medical School, Tokyo 113-8603, Japan
| | - Hideki Kogo
- Department of Gastroenterological Surgery, Nippon Medical School, Tokyo 113-8603, Japan
| | - Takahiro Haruna
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo 113-8603, Japan
| | - Ryuji Ohashi
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo 113-8603, Japan
| | - Hiroshi Yoshida
- Department of Gastroenterological Surgery, Nippon Medical School, Tokyo 113-8603, Japan
| |
Collapse
|
41
|
He WZ, Yang YZ, Yin CX, Xian XY, Gu JM, Yi JH, Xue J, Zhao Y, Wang F, Hu WM, Xia LP. Evolution of HER2-low expression from primary to paired metastatic gastric cancer lesions. NPJ Precis Oncol 2025; 9:108. [DOI: 40234621 10.1038/s41698-025-00881-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/15/2025] [Indexed: 05/20/2025] Open
|
42
|
Tamba M, Osumi H, Ogura M, Fukuoka S, Okamura A, Kanamori J, Imamura Y, Yoshino K, Udagawa S, Wakatsuki T, Shinozaki E, Watanabe M, Yamaguchi K, Chin K, Ooki A. Real-world safety and efficacy of neoadjuvant docetaxel, cisplatin, and 5-fluorouracil therapy for locally advanced esophageal squamous cell carcinoma. BMC Cancer 2025; 25:636. [PMID: 40200210 PMCID: PMC11980314 DOI: 10.1186/s12885-025-14011-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy with docetaxel, cisplatin plus 5-FU (DCF) has become the new standard of care for locally advanced esophageal squamous cell carcinoma (ESCC). In a real-world setting, the efficacy, recurrence, and adverse events (AEs) remain unclear. METHODS This retrospective cohort study included 86 patients who received neoadjuvant DCF followed by esophagectomy for resectable ESCC. RESULTS Following neoadjuvant DCF treatment, 75 patients underwent R0 curative resection. At the median follow-up of 19.2 months, the median disease-free survival (DFS)/recurrence-free survival (RFS) was not yet reached, with estimated 3-year DFS/RFS rates of 65.2%, respectively. The incidence of primary tumor regression grading (TRG) grade 1a and pathological complete response (pCR) were 21.3% (16/75) and 14.7% (11/75), respectively. The estimated 1-year DFS/RFS rates were 93.8% for primary TRG grade 1a and 100% for pCR. Baseline elevated serum SCC-antigen levels were inversely associated with achieving primary TRG grade 1a or pCR. In 64 patients who did not achieve pCR, residual tumor cells in the lymph nodes (ypN; HR, 16.96; 95% CI, 2.11-136.12; P < 0.01) and Glasgow prognostic score (GPS; HR, 8.34; 95% CI, 1.73-40.31; P < 0.01) were independent predictors of shorter DFS/RFS. The most common grade ≥ 3 AEs were neutropenia (61.6%) and febrile neutropenia (26.7%), which were not associated with clinicopathological factors. The most common non-hematological AEs were appetite loss (9.3%), pulmonary embolism (8.1%), diarrhea (7.0%), and nausea (2.3%). Nine patients discontinued neoadjuvant DCF due to toxicities. CONCLUSIONS Neoadjuvant DCF was effective and well-tolerated in real-world ESCC patients. Primary TRG grade 1a or pCR showed a favorable DFS/RFS, while positive ypN and GPS were independent risk factors for worse DFS/RFS.
Collapse
Affiliation(s)
- Mikako Tamba
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Hiroki Osumi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Mariko Ogura
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Shota Fukuoka
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Akihiko Okamura
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Jun Kanamori
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yu Imamura
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Koichiro Yoshino
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Shohei Udagawa
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Takeru Wakatsuki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Eiji Shinozaki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Keisho Chin
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Akira Ooki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| |
Collapse
|
43
|
Mielcarska S, Kot A, Kula A, Dawidowicz M, Sobków P, Kłaczka D, Waniczek D, Świętochowska E. B7H3 in Gastrointestinal Tumors: Role in Immune Modulation and Cancer Progression: A Review of the Literature. Cells 2025; 14:530. [PMID: 40214484 PMCID: PMC11988818 DOI: 10.3390/cells14070530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
B7-H3 (CD276), a member of the B7 immune checkpoint family, plays a critical role in modulating immune responses and has emerged as a promising target in cancer therapy. It is highly expressed in various malignancies, where it promotes tumor evasion from T cell surveillance and contributes to cancer progression, metastasis, and therapeutic resistance, showing a correlation with the poor prognosis of patients. Although its receptors were not fully identified, B7-H3 signaling involves key intracellular pathways, including JAK/STAT, NF-κB, PI3K/Akt, and MAPK, driving processes crucial for supporting tumor growth such as cell proliferation, invasion, and apoptosis inhibition. Beyond immune modulation, B7-H3 influences cancer cell metabolism, angiogenesis, and epithelial-to-mesenchymal transition, further exacerbating tumor aggressiveness. The development of B7-H3-targeting therapies, including monoclonal antibodies, antibody-drug conjugates, and CAR-T cells, offers promising avenues for treatment. This review provides an up-to-date summary of the B7H3 mechanisms of action, putative receptors, and ongoing clinical trials evaluating therapies targeting B7H3, focusing on the molecule's role in gastrointestinal tumors.
Collapse
Affiliation(s)
- Sylwia Mielcarska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Anna Kot
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Agnieszka Kula
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Miriam Dawidowicz
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Piotr Sobków
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Daria Kłaczka
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| |
Collapse
|
44
|
Oh SE, Suh YS, An JY, Ryu KW, Cho I, Kim SG, Park JH, Hur H, Kim HH, Ahn SH, Hwang SH, Yoon HM, Park KB, Kim HI, Kwon IG, Yang HK, Suh BJ, Jeong SH, Kim TH, Kwon OK, Ahn HS, Park JY, Yoon KY, Son MW, Kong SH, Son YG, Song GJ, Yun JH, Bae JM, Park DJ, Lee S, Yang JY, Seo KW, Jang YJ, Kang SH, Eom BW, Lee J, Lee HJ. Prospective Multicenter Observational Study on Postoperative Quality of Life According to Type of Gastrectomy for Gastric Cancer. J Gastric Cancer 2025; 25:382-399. [PMID: 40200880 PMCID: PMC11982510 DOI: 10.5230/jgc.2025.25.e26] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/10/2025] Open
Abstract
PURPOSE This study evaluated the postoperative quality of life (QoL) after various types of gastrectomy for gastric cancer. MATERIALS AND METHODS A multicenter prospective observational study was conducted in Korea using the Korean Quality of Life in Stomach Cancer Patients Study (KOQUSS)-40, a new QoL assessment tool focusing on postgastrectomy syndrome. Overall, 496 patients with gastric cancer were enrolled, and QoL was assessed at 5 time points: preoperatively and at 1, 3, 6, and 12 months after surgery. RESULTS Distal gastrectomy (DG) and pylorus-preserving gastrectomy (PPG) showed significantly better outcomes than total gastrectomy (TG) and proximal gastrectomy (PG) with regard to total score, indigestion, and dysphagia. DG, PPG, and TG also showed significantly better outcomes than PG in terms of dumping syndrome and worry about cancer. Postoperative QoL did not differ significantly according to anastomosis type in DG, except for Billroth I anastomosis, which achieved better bowel habit change scores than the others. No domains differed significantly when comparing double tract reconstruction and esophagogastrostomy after PG. The total QoL score correlated significantly with postoperative body weight loss (more than 10%) and extent of resection (P<0.05 for both). Reflux as assessed by KOQUSS-40 did not correlate significantly with reflux observed on gastroscopy 1 year postoperatively (P=0.064). CONCLUSIONS Our prospective observation using KOQUSS-40 revealed that DG and PPG lead to better QoL than TG and PG. Further study is needed to compare postoperative QoL according to anastomosis type in DG and PG.
Collapse
Affiliation(s)
- Sung Eun Oh
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji Yeong An
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keun Won Ryu
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - In Cho
- Department of Surgery, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Sung Geun Kim
- Department of Surgery, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji-Ho Park
- Department of Surgery, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, Korea
| | - Hyung-Ho Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sang-Hoon Ahn
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sun-Hwi Hwang
- Department of Surgery, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Hong Man Yoon
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - Ki Bum Park
- Department of Surgery, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Hyoung-Il Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - In Gyu Kwon
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Han-Kwang Yang
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Byoung-Jo Suh
- Department of Surgery, Inje University Haeundae Paik Hospital, Busan, Korea
| | - Sang-Ho Jeong
- Department of Surgery, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Tae-Han Kim
- Department of Surgery, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Oh Kyoung Kwon
- Department of Surgery, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Hye Seong Ahn
- Department of Surgery, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Ji Yeon Park
- Department of Surgery, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Ki Young Yoon
- Department of Surgery, Kosin University College of Medicine, Busan, Korea
| | - Myoung Won Son
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Seong-Ho Kong
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Gil Son
- Department of Surgery, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Geum Jong Song
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Jong Hyuk Yun
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Jung-Min Bae
- Department of Surgery, Yeungnam University College of Medicine, Daegu, Korea
| | - Do Joong Park
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sol Lee
- Department of Surgery, Seoul Medical Center, Seoul, Korea
| | - Jun-Young Yang
- Department of Surgery, Gachon University Gil Medical Center, Incheon, Korea
| | - Kyung Won Seo
- Department of Surgery, Kosin University College of Medicine, Busan, Korea
| | - You-Jin Jang
- Department of Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - So Hyun Kang
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Bang Wool Eom
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - Joongyub Lee
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Hyuk-Joon Lee
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
45
|
Kumar R, Shalaby A, Narra LR, Gokhale S, Deek MP, Jabbour SK. Updates in the Role of Positron Emission Tomography/Computed Tomography in Radiation Oncology in Gastrointestinal Malignancies. PET Clin 2025; 20:219-229. [PMID: 39952884 PMCID: PMC12037145 DOI: 10.1016/j.cpet.2025.01.004] [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] [Indexed: 02/17/2025]
Abstract
Positron Emission Tomography/Computed Tomography (PET/CT) plays a critical role in managing gastrointestinal (GI) cancers within radiation oncology. It enhances tumor detection, staging, and lymph node involvement assessment, leading to better-targeted radiation treatment. PET/CT also aids in delineating tumor volumes to minimize geographic misses, enabling precise dose escalation to metabolically active regions. Despite its benefits, PET/CT has limitations such as false positives and dependency on complementary imaging. Emerging technologies offer real-time adjustments and personalized treatments, advancing precision medicine in GI radiation oncology. Further research is needed to refine PET/CT integration for improved treatment outcomes and cost-effectiveness.
Collapse
Affiliation(s)
- Ritesh Kumar
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Ahmed Shalaby
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Lakshmi Rekha Narra
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Shivani Gokhale
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Matthew P Deek
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA.
| |
Collapse
|
46
|
Amini N, Kinoshita T, Arrieta M, Yoshida M, Nagata H, Habu T, Komatsu M, Yura M. Novel Robotic Valvuloplastic Esophagogastrostomy Technique After Proximal Gastrectomy: A Safety and Feasibility Study. Surg Laparosc Endosc Percutan Tech 2025; 35:e1322. [PMID: 39895544 DOI: 10.1097/sle.0000000000001322] [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: 07/26/2024] [Accepted: 08/09/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND Esophagogastrostomy is one of the reconstruction techniques after proximal gastrectomy, but reflux and esophagitis are significant concerns. We introduced a new robotic valvuloplasty technique (single-flap), taking advantage of robotic surgery to address these issues and simplify the technique, especially for tumors with esophageal invasion. METHODS Between March 2022 and March 2024, patients who underwent robotic proximal gastrectomy with the single-flap technique were included. Based on the difficulty of the surgery, patients were divided into 2 groups: one with esophageal invasion requiring anastomosis in the mediastinum and the second group with tumors in the upper third of the stomach requiring anastomosis in the abdomen. RESULTS A total of 22 patients were included: 13 in the esophageal invasion group and 9 in the upper stomach group. The median size of esophageal invasion was 2 cm (1 to 3 cm). The median operative time was 320 minutes (esophageal invasion 326 vs. upper stomach 280 min, P =0.51), with a median blood loss of 35 g (31 vs. 38 g, P =0.19). No postoperative mortality, anastomotic leaks, reflux symptoms, or pancreatic fistulas were observed. Eighteen patients underwent endoscopic evaluation, and no sign of esophagitis was detected. Five patients (22.7%) developed grade III strictures requiring endoscopic balloon dilation (esophageal invasion 32.1% vs. upper stomach 22.2%; P =0.96). CONCLUSIONS Robotic proximal gastrectomy with single-flap valvuloplastic esophagogastrostomy is a safe and feasible option for gastroesophageal junction tumors with up to 3 cm of esophageal invasion.
Collapse
Affiliation(s)
- Neda Amini
- Department of Gastric Surgery, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Surgical Oncology, North Shore/Long Island Jewish, Northwell Health, New Hyde Park, NY
| | - Takahiro Kinoshita
- Department of Gastric Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Manuel Arrieta
- Department of Gastric Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Mitsumasa Yoshida
- Department of Gastric Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiromi Nagata
- Department of Gastric Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takumi Habu
- Department of Gastric Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masaru Komatsu
- Department of Gastric Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masahiro Yura
- Department of Gastric Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
47
|
Li F, Deng H, Hu Z, Chen Z, Zhang H, He J, Wang X, Liu Y. Immunohistochemical-Based Molecular Typing of ACRG Combined With Immune-Associated PD-L1 Expression Can Predict the Prognosis of Gastric Cancer. Cancer Med 2025; 14:e70863. [PMID: 40202155 PMCID: PMC11979789 DOI: 10.1002/cam4.70863] [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: 11/11/2024] [Revised: 03/29/2025] [Accepted: 03/30/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a molecularly heterogeneous disease with diverse clinical outcomes. Traditional classifications lack predictive accuracy, necessitating alternative molecular subtyping approaches for effective prognosis prediction. The Asian Cancer Research Group (ACRG) molecular subtypes, combined with immune-associated PD-L1 expression, offer a promising framework to predict patient outcomes and potentially guide treatment strategies in GC. METHODS This study retrospectively analyzed 1007 primary GC patients who underwent surgical resection between January 2017 and June 2019 at the Fourth Hospital of Hebei Medical University. Comprehensive immunohistochemical and fluorescent PCR-capillary electrophoresis analyses were conducted to determine ACRG molecular subtypes (microsatellite instability [MSI], microsatellite stability with epithelial-mesenchymal transition [MSS/EMT], MSS/TP53+, and MSS/TP53-) and PD-L1 expression. We assessed the relationship between these classifications and various clinicopathological parameters, including survival outcomes, using Cox regression and Kaplan-Meier analysis. RESULTS The ACRG subtypes showed significant associations with clinicopathological features, including tumor invasion depth, Lauren classification, and HER2 status. The MSI subtype (6.7% of cases) was associated with higher PD-L1 positivity and a favorable prognosis, whereas the EMT subtype had the lowest 5-year survival rate (34.55%) and was predominantly linked to diffuse-type histology. PD-L1 positivity correlated with worse survival outcomes, with independent predictive value alongside ACRG subtypes (HR for PD-L1 = 1.759, p = 0.001; HR for ACRG = 5.144, p < 0.001). CONCLUSION The combination of ACRG molecular subtyping and PD-L1 expression serves as an effective predictor of GC prognosis, facilitating tailored clinical decision-making. The ACRG-PD-L1 classification system offers a practical, cost-effective approach for routine clinical application, providing critical insight into GC heterogeneity. Further multicenter studies are needed to validate these findings and explore the impact of ACRG subtypes on therapy responses, particularly in immunotherapy settings.
Collapse
Affiliation(s)
- Fang Li
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
- Department of Graduate SchoolHebei Medical UniversityShijiazhuangChina
| | - Huiyan Deng
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Zeqing Hu
- Department of EmergencyPingxiang General HospitalXingtaiChina
| | - Zihao Chen
- Department of OncologyFirst Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Huirui Zhang
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Jiankun He
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Xiaoxiao Wang
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yueping Liu
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| |
Collapse
|
48
|
Luo G, Zhu S, He L, Liu Q, Xu C, Yao Q, Hu H, Wang Q, Zou S. Platelets promote metastasis of intrahepatic cholangiocarcinoma through activation of TGF-β/Smad2 pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167734. [PMID: 39978442 DOI: 10.1016/j.bbadis.2025.167734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 01/11/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Intrahepatic cholangiocarcinoma (ICC), an aggressive liver cancer, lacks simple and accurate clinical tests, which poses challenges to postoperative diagnosis and treatment. Recent studies have indicated that platelet levels might be relevant to the postoperative prognosis of ICC. However, their prognostic significance in ICC remains unclarified. This study included 218 ICC patients who underwent hepatic resection. Comprehensive analyses of patients' postoperative prognosis were conducted primarily focusing on their platelet levels associated with prognostic traits. To further investigate the underlying mechanism between platelet levels and patients' postoperative prognosis, we elucidated the association between platelets and tumor metastasis using HCCC-9810 and HUCC-T1 cells as well as mouse models. In the retrospective cohort study, elevated serum platelet levels (≥300 × 109/L) or tumoral platelet levels (≥0.23) individually indicated an unfavorable postoperative prognosis in individuals with ICC. Multivariate analysis showed that tumoral platelet levels can be an independent prognostic factor, while the loss of prognostic superiority of serum platelet levels in the analysis may be attributed to the influence of confounding inclusion variables. Epithelial/mesenchymal transition (EMT) marker expression changes in HCCC-9810 and HUCC-T1 cells with platelet treatment were analyzed to understand how platelets contribute to ICC malignant recurring progression. The significant role of the TGF-β/Smad2 pathway in ICC metastasis was identified. In addition, aspirin was found to have the potential to reduce ICC metastasis by inhibiting platelet function. In conclusion, this study indicated that ICC patients with postoperative serum platelet levels ≥300 × 109/L or tumoral platelet levels ≥0.23 have significantly higher risk of poor postoperative prognosis. This is due to platelet-derived TGFβ1 leading to EMT in ICC cells, thus promoting tumor metastasis.
Collapse
Affiliation(s)
- Guijuan Luo
- Department of Hepatobiliary Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Shuyang Zhu
- Department of Hepatobiliary Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Liujie He
- Department of Hepatobiliary Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Qiang Liu
- Department of Hepatobiliary Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Chao Xu
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou 310005, China; Integrated Traditional Chinese and Western Medicine Oncology Laboratory, Key Laboratory of Traditional Chinese Medicine of Zhejiang Province, Hangzhou 310022, China
| | - Qinghua Yao
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou 310005, China; Integrated Traditional Chinese and Western Medicine Oncology Laboratory, Key Laboratory of Traditional Chinese Medicine of Zhejiang Province, Hangzhou 310022, China
| | - Heping Hu
- Department of Hepatobiliary Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China.
| | - Qiang Wang
- Department of Urology, Peking University People's Hospital, Beijing 100044, China.
| | - Shanshan Zou
- Department of Hepatobiliary Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China.
| |
Collapse
|
49
|
Nakagawa S, Sato T, Ohashi E, Kajita M, Miya F, Yamamoto K, Yotsumata H, Yamaguchi K, Nakajima Y, Miura A, Kinugasa Y, Ohteki T. An organoid library of human esophageal squamous cell carcinomas (ESCCs) uncovers the chemotherapy-resistant ESCC features. Commun Biol 2025; 8:507. [PMID: 40169778 PMCID: PMC11961618 DOI: 10.1038/s42003-025-07869-4] [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: 07/31/2024] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a deadly cancer with a poor prognosis and a high recurrence rate after chemotherapy, posing a significant clinical challenge. To elucidate the molecular basis of chemotherapy (chemo)-resistance and to develop methods to effectively eliminate chemo-resistant tumor clones, we established an ESCC organoid (ESCCO) library from 24 ESCC patients of various stages, ages, and treatments. These ESCCOs faithfully recapitulate the oncogenic mutations observed in the original ESCC tissues and manifest tumorigenic properties when xenografted. The ESCCOs respond differently to cisplatin and 5-fluorouracil, chemotherapeutic agents commonly used to treat ESCC patients, with 7 ESCCOs exhibiting potent chemo-resistance. Notably, the chemo-resistant ESCCOs show higher genes involved in antioxidant stress response pathways and more accessible chromatin at their loci than the sensitive ESCCOs. These genes can serve as valuable biomarkers to stratify chemo-resistant ESCCs in histopathological specimens. Through drug screening using the ESCCO library, we reveal that fedratinib effectively induces cell death in chemo-resistant ESCCOs. Collectively, our human ESCCO model offers novel insights into the mechanism of chemo-resistance in ESCCs, which is critical for developing effective therapeutic approaches to eradicate the recurrence of ESCCs.
Collapse
Affiliation(s)
- Shunsaku Nakagawa
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University (TMDU)), Tokyo, Japan
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Taku Sato
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University (TMDU)), Tokyo, Japan.
- Department of Biochemistry and Molecular Biology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan.
| | - Eriko Ohashi
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University (TMDU)), Tokyo, Japan
| | - Mihoko Kajita
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University (TMDU)), Tokyo, Japan
| | - Fuyuki Miya
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Hiroki Yotsumata
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University (TMDU)), Tokyo, Japan
| | - Kazuya Yamaguchi
- Department of Esophageal Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Yasuaki Nakajima
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Akinori Miura
- Department of Esophageal Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Yusuke Kinugasa
- Department of Gastrointestinal Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Toshiaki Ohteki
- Department of Biodefense Research, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo (formerly Medical Research Institute, Tokyo Medical and Dental University (TMDU)), Tokyo, Japan.
| |
Collapse
|
50
|
Kong S, Cai X, Cai B, Xian Y, Zhou Z, Cai D, Yang X, Lin D, Nie Q. Genomic and transcriptomic analyses unveil the genetic basis of green shank trait in small white-feather chickens. Poult Sci 2025; 104:104912. [PMID: 39985900 PMCID: PMC11904536 DOI: 10.1016/j.psj.2025.104912] [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: 12/10/2024] [Revised: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025] Open
Abstract
Small white-feather chickens (SWFC) have become popular as a hybrid strain recently. Shank color is a notable economic trait in this strain. Despite numerous studies on the green shank trait from both physiological and genetic perspectives, research focusing specifically on the green shank trait in hybrid chickens (HC) remains limited. In this study, to investigate the genetic mechanisms and molecular basis of the green shank trait in HC, we created a population by intercrossing white-feathered and yellow-feathered broilers, both with yellow shanks. Physiological analysis confirmed that melanin deposition in the shank dermis is the primary cause of the green shank trait in HC. By combining genome-wide association studies (GWAS) and population genomics analysis, the 83.20-85.68 Mb region on the Z chromosome was identified as a candidate region for the green shank trait in HC. Transcriptome sequencing revealed differentially expressed genes (DEGs) between green shank and yellow shank individuals, with MTAP and CDKN1A identified as candidate genes in the genomic region associated with the green shank trait. Notably, the green shank trait includes a light green phenotype. Our study is the first to identify genes associated with different color depths of the green shank. The candidate genes influence both the biosynthesis and deposition of pigments.
Collapse
Affiliation(s)
- Shaofen Kong
- College of Animal Science, South China Agricultural University, Guangzhou, China; State Key Laboratory of Swine and Poultry Breeding Industry, Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Xiaodian Cai
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Bolin Cai
- College of Animal Science, South China Agricultural University, Guangzhou, China; State Key Laboratory of Swine and Poultry Breeding Industry, Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Yuanrong Xian
- College of Animal Science, South China Agricultural University, Guangzhou, China; State Key Laboratory of Swine and Poultry Breeding Industry, Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Zhen Zhou
- College of Animal Science, South China Agricultural University, Guangzhou, China; State Key Laboratory of Swine and Poultry Breeding Industry, Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Danfeng Cai
- College of Animal Science, South China Agricultural University, Guangzhou, China; State Key Laboratory of Swine and Poultry Breeding Industry, Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Xin Yang
- College of Animal Science, South China Agricultural University, Guangzhou, China; State Key Laboratory of Swine and Poultry Breeding Industry, Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Duo Lin
- College of Animal Science, South China Agricultural University, Guangzhou, China; State Key Laboratory of Swine and Poultry Breeding Industry, Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Qinghua Nie
- College of Animal Science, South China Agricultural University, Guangzhou, China; State Key Laboratory of Swine and Poultry Breeding Industry, Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China.
| |
Collapse
|