151
|
Baba SK, Alblooshi SSE, Yaqoob R, Behl S, Al Saleem M, Rakha EA, Malik F, Singh M, Macha MA, Akhtar MK, Houry WA, Bhat AA, Al Menhali A, Zheng ZM, Mirza S. Human papilloma virus (HPV) mediated cancers: an insightful update. J Transl Med 2025; 23:483. [PMID: 40301924 PMCID: PMC12039116 DOI: 10.1186/s12967-025-06470-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] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/07/2025] [Indexed: 05/01/2025] Open
Abstract
Human papillomavirus (HPV), a DNA virus, is a well-documented causative agent of several cancers, including cervical, vulvar, vaginal, penile, anal, and head & neck cancers. Major factors contributing to HPV-related cancers include persistent infection and the oncogenic potential of particular HPV genotypes. High-risk HPV strains, particularly HPV-16 and HPV-18, are responsible for over 70% of cervical cancer cases worldwide, as well as a significant proportion of other genital and head and neck cancers. At the molecular level, the oncogenic activity of these viruses is driven by the overexpression of E6 and E7 oncoproteins. These oncoproteins dysregulate the cell cycle, inhibit apoptosis, and promote the accumulation of DNA damage, ultimately transforming normal cells into cancerous ones. This review aims to provide a comprehensive overview of the recent advances in HPV-related cancer biology and epidemiology. The review highlights the molecular pathways of HPV-driven carcinogenesis, focusing on the role of viral oncoproteins in altering host cell targets and disrupting cellular signalling pathways. The review explores the therapeutic potential of these viral proteins, and discusses current diagnostic and treatment strategies for HPV-associated cancers. Furthermore, the review highlights the critical role of HPV in the development of various malignancies, emphasizing the persistent challenges in combating these cancers despite advancements in vaccination and therapeutic strategies. We also emphasize recent breakthroughs in utilizing biomarkers to monitor cancer therapy responses, such as mRNAs, miRNAs, lncRNAs, proteins, and genetic markers. We hope this review will serve as a valuable resource for researchers working on HPV, providing insights that can guide future investigations into this complex virus, which continues to be a major contributor to global morbidity and mortality.
Collapse
Affiliation(s)
- Sadaf Khursheed Baba
- Department of Chemistry, College of Science (COS), United Arab Emirates University (UAEU), P.O. Box 15551, Al Ain, United Arab Emirates
| | | | - Reem Yaqoob
- Department of Chemistry, College of Science (COS), United Arab Emirates University (UAEU), P.O. Box 15551, Al Ain, United Arab Emirates
| | - Shalini Behl
- Omics Centre of Excellence, M42 Health, Abu Dhabi, United Arab Emirates
| | - Mansour Al Saleem
- Department of Applied Medical Sciences, Applied College, Qassim University, Qassim, Saudi Arabia
| | - Emad A Rakha
- Histopathology Department, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Pathology, Hamad Medical Corporation, Doha, Qatar
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar, Jammu and Kashmir, 190005, India
| | - Mayank Singh
- Department of Medical Oncology (Lab), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, 192122, India
| | - Mohammed Kalim Akhtar
- Department of Chemistry, College of Science (COS), United Arab Emirates University (UAEU), P.O. Box 15551, Al Ain, United Arab Emirates
| | - Walid A Houry
- Department of Biochemistry, University of Toronto, Toronto, ON, M5G 1M1, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
| | - Ajaz A Bhat
- Metabolic and Mendelian Disorders Clinical Research Program, Precision OMICs Research & Translational Science, Sidra Medicine, Doha, Qatar
| | - Asma Al Menhali
- Department of Biology, College of Science (COS), United Arab Emirates University (UAEU), Al Ain, United Arab Emirates
| | - Zhi-Ming Zheng
- Tumor Virus RNA Biology Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Sameer Mirza
- Department of Chemistry, College of Science (COS), United Arab Emirates University (UAEU), P.O. Box 15551, Al Ain, United Arab Emirates.
- Zayed Bin Sultan Centre for Health Sciences, United Arab Emirates University (UAEU), Al Ain, United Arab Emirates.
| |
Collapse
|
152
|
Zhang W, Zeng M, Ma X, Chen J, Qiao J, He Z, Zhong G, Li Y, Yu L. CLDN18.2-targeting STAR-T cell therapy for pancreatic cancer: a strategy to minimize gastric off-tumor toxicity compared to CLDN18.2 CAR-T. Oncogene 2025:10.1038/s41388-025-03414-z. [PMID: 40301544 DOI: 10.1038/s41388-025-03414-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025]
Abstract
Claudin18 isoform 2 (CLDN18.2), primarily expressed in gastric tissue and upregulated in pancreatic cancer (PC), is a key target for innovative treatments like chimeric antigen receptor T (CAR-T) cell therapy. However, CAR-T's effectiveness comes with a significant risk of on-target, off-tumor (OTOT) toxicity due to CLDN18.2's presence in normal gastric mucosa. To address this, we developed CLDN18.2-specific synthetic T cell receptor and antigen receptor T (STAR-T) cells. Our research shows that STAR-T and CAR-T cells have comparable in vitro cytotoxicity, but STAR-T cells cause less gastric damage in vivo despite having weaker antitumor effects than CAR-T cells. Clinical tests with gastroscopes confirmed the gastric safety of STAR-T cell therapy, which effectively controlled the disease. Additionally, incorporating the IL12β p40 subunit into STAR-T cells enhanced their function in both lab and animal studies. This evidence suggests that CLDN18.2 STAR-T cell could be a safer alternative to CAR-T cell therapy for PC, meriting further clinical trials.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
| | - Miao Zeng
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
| | - Xingyu Ma
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, China
| | - Jinghong Chen
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, China
| | - Jingqiao Qiao
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, China
| | - Ziqian He
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, China
| | - Guocheng Zhong
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, China.
| | - Yisheng Li
- Shenzhen Haoshi Biotechnology Co. Ltd, Shenzhen, 518125, China.
| | - Li Yu
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, China.
| |
Collapse
|
153
|
Norouzkhani N, Mobaraki H, Varmazyar S, Zaboli H, Mohamadi Z, Nikeghbali G, Bagheri K, Marivany N, Najafi M, Nozad Varjovi M, Abouzeid M, Zeidi Baghrabad H, Eini P, Azhdarimoghaddam A, Khosravi F, Asadi Anar M. Artificial intelligence networks for assessing the prognosis of gastrointestinal cancer to immunotherapy based on genetic mutation features: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:310. [PMID: 40301768 PMCID: PMC12039286 DOI: 10.1186/s12876-025-03884-1] [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: 01/18/2025] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND AND AIM Artificial intelligence (AI) networks offer significant potential for predicting immunotherapy outcomes in gastrointestinal cancers by analyzing genetic mutation profiles. Their application in prognosis remains underexplored. This systematic review and meta-analysis aim to evaluate the effectiveness of AI-based models, which refers to systems utilizing artificial intelligence to analyze data and make predictions, in predicting immunotherapy responses in gastrointestinal cancers using genetic mutation features. METHODS This study, adhering to PRISMA guidelines, aimed to evaluate AI networks for predicting gastrointestinal cancer prognosis in response to immunotherapy using genetic mutation features. A search in PubMed, WOS, and Scopus identified relevant studies. Data extraction and quality assessment were conducted, and statistical analysis included pooled estimates for sensitivity, specificity, accuracy, and AUC. Regression models and imputation methods addressed missing values, ensuring accurate and robust results. STATA version 18 was used to analyze the data. RESULT A total of 45 studies, all published in 2024, involving 14,047 participants in training sets and 10,885 participants in test sets, were included. The pooled results of AI model performance for gastrointestinal cancers based on genetic mutation features were: AUC = 0.86 (95% CI: 0.86-0.87), Sensitivity = 83% (95% CI: 83%-84%), Specificity = 72% (95% CI: 72%-73%), and Accuracy = 82% (95% CI: 82%-83%). Heterogeneity was low to moderate, and no publication bias was detected. Subgroup analysis showed higher AUC for gastric cancer models (AUC: 0.87) and lower for pancreatic cancer models (AUC: 0.52). CONCLUSION AI networks demonstrate promising potential in predicting immunotherapy outcomes for gastrointestinal cancers based on genetic mutation features. This systematic review highlights their effectiveness in stratifying patients and optimizing treatment decisions. However, further large-scale studies are needed to validate AI models and integrate them into clinical practice for improved precision in cancer immunotherapy.
Collapse
Affiliation(s)
- Narges Norouzkhani
- Department of Medical Informatics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hesam Mobaraki
- Faculty of Medicine, İstanbul Yeniyuzyil University, Istanbul, Turkey
| | | | - Hadis Zaboli
- Student Research Committee, Babol University Medical Science, Babol, Iran
| | - Zhina Mohamadi
- Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Golnaz Nikeghbali
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamyar Bagheri
- Student Research Committee, Abadan University of Medical Sciences, Abadan, Iran
| | - Newsha Marivany
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mirmehdi Najafi
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahdiyeh Nozad Varjovi
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohamed Abouzeid
- Mersey & West Lancashire Teaching Hospitals NHS Trust, Southport Hospital, Lancashire, UK.
| | - Hanieh Zeidi Baghrabad
- Tabriz University of Medical Sciences, Gastroenterology and Hepatology Research Center, Tabriz, Iran
| | - Pooya Eini
- Toxicological Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aida Azhdarimoghaddam
- Student Research Committee, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Farbod Khosravi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
154
|
Yang T, Wang X, Jin Y, Yao X, Sun Z, Chen P, Zhou S, Zhu W, Chen W. Deep learning radiopathomics predicts targeted therapy sensitivity in EGFR-mutant lung adenocarcinoma. J Transl Med 2025; 23:482. [PMID: 40301933 PMCID: PMC12039126 DOI: 10.1186/s12967-025-06480-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 04/11/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Ttyrosine kinase inhibitors (TKIs) represent the standard first-line treatment for patients with epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma. However, not all patients with EGFR mutations respond to TKIs. This study aims to develop a deep learning radiological-pathological-clinical (DLRPC) model that integrates computed tomography (CT) images, hematoxylin and eosin (H&E)-stained aspiration biopsy samples, and clinical data to predict the response in EGFR-mutant lung adenocarcinoma patients undergoing TKIs treatment. METHODS We retrospectively analyzed data from 214 lung adenocarcinoma patients who received TKIs treatment from two medical centers between September 2013 and June 2023. The DLRPC model leverages paired CT, pathological images and clinical data, incorporating a clinical-based attention mask to further explore the cross-modality associations. To evaluate its diagnostic performance, we compared the DLRPC model against single-modality models and a decision level fusion model based on Dempster-Shafer theory. Model performances metrics, including area under the curve (AUC), accuracy, sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV), were used for evaluation. The Delong test assessed statistically significantly differences in AUC among models. RESULTS The DLRPC model demonstrated strong performance, achieving an AUC value of 0.8424. It outperformed the single-modality models (AUC = 0.6894, 0.7753, 0.8052 for CT model, pathology model and clinical model, respectively. P < 0.05). Additionally, the DLRPC model surpassed the decision level fusion model (AUC = 0.8132, P < 0.05). CONCLUSION The DLRPC model effectively predicts the response of EGFR-mutant lung adenocarcinoma patients to TKIs, providing a promising tool for personalized treatment decisions in lung cancer management.
Collapse
Affiliation(s)
- Taotao Yang
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China
| | - Xianqi Wang
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China
| | - Yuan Jin
- Zhejiang Lab, Hangzhou, 311121, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, (Third Military Medical University), Chongqing, 400038, China
| | - Zhiyuan Sun
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Pinzhen Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Suyi Zhou
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | | | - Wei Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China.
| |
Collapse
|
155
|
Liu W, Li L, Guo L, Li H, Tang Z, Wang X, Huang L, Sun Y. Dasatinib demonstrates efficacy in organoid derived paclitaxel-resistant Trp53/Cdh1-deficient mouse gastric adenocarcinoma with peritoneal metastasis. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:16. [PMID: 40299206 PMCID: PMC12040775 DOI: 10.1186/s13619-025-00232-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025]
Abstract
Gastric cancer peritoneal metastasis (GCPM) typically indicates a poor clinical prognosis and is frequently observed in diffuse gastric cancer (GC) patients with CDH1 loss of function. GCPM characterized for its aggressiveness and resistance to chemotherapy, most notably paclitaxel (PTX), poses significant treatment challenges. Previously, no mouse gastric adenocarcinoma (MGA) cell lines with Trp53 (encoding mouse p53) and Cdh1 (encoding mouse E-cadherin) mutations and a high potential for peritoneal metastasis in mice have been established. Here, we derived a mouse GC cell line, called MTC, from subcutaneously transplanted mouse Trp53-/-Cdh1-/- GC organoids. Through matching the short tandem repeat profile of MTC with those in current cell banks, we verified the uniqueness of MTC. Furtherly, we confirmed the features of MTC by detecting the expression of p53, E-cadherin, and pan-CK. After long-term exposure of the original MTC line to PTX, we developed a more aggressive, PTX-resistant cell line, termed MTC-R. Compared with MTC, MTC-R demonstrated enhanced tumorigenicity and high potential for peritoneal metastasis in subcutaneous and intraperitoneal tumour models both in BALB/c nude mice and C57BL/6 J mice. Transcriptome analysis revealed the ECM‒receptor interaction pathway activation during the development of PTX resistance, and dasatinib (DASA) was identified as a potential drug targeting this pathway. DASA showed promise in ameliorating disease progression and improving overall survival in MTC-R GCPM model in C57BL/6 J mice. Overall, we established a novel MGA cell line with Trp53 and Cdh1 mutations and its PTX-resistant variant and demonstrated the efficacy of DASA in treating PTX-resistant GCPM.
Collapse
Affiliation(s)
- Wenshuai Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Retroperitoneal Sarcoma Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lingmeng Li
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Leilei Guo
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haojie Li
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhaoqing Tang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xuefei Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Liyu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
156
|
Lin H, Hua J, Gong Z, Chen M, Qiu B, Wu Y, He W, Wang Y, Feng Z, Liang Y, Long W, Li R, Kuang Q, Chen Y, Lu J, Luo S, Zhao W, Yan L, Chen X, Shi Z, Xu Z, Mo Z, Liu E, Han C, Cui Y, Yang X, Chen X, Liu J, Pan X, Madabhushi A, Lu C, Liu Z. Multimodal radiopathological integration for prognosis and prediction of adjuvant chemotherapy benefit in resectable lung adenocarcinoma: A multicentre study. Cancer Lett 2025; 616:217557. [PMID: 39954935 DOI: 10.1016/j.canlet.2025.217557] [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: 11/30/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Lung adenocarcinoma (LUAD) has a heterogeneous prognosis and controversial postoperative treatment protocols. We aim to develop and validate a multimodal analysis framework that integrates CT images with H&E-stained whole-slide images (WSIs) to enhance risk stratification and predict adjuvant chemotherapy benefit in LUAD patients. We retrospectively collected data from 1039 resectable LUAD patients (stage I-III) across four centres, forming a training dataset (n = 303), two testing datasets (n = 197 and n = 228) for survival analysis, and a feature testing dataset (n = 311) for interpretability analysis. We extracted 487 tumour/peritumour radiomics features from CT images and 783 multiscale pathomics features from WSIs, characterising the shape of tumour (CT) and cancer nuclei (WSIs), as well as the intensity and texture of tumour/peritumour regions (CT) and tumour regions/epithelium/stroma (WSIs). A survival support vector machine (SVM) was employed to establish a radiopathomics signature using the optimal set of multimodal features, including 2 tumour radiomics features, 3 peritumour radiomics features, and 4 nuclei heterogeneity pathomics features. The radiopathomics signature outperformed both radiomics and pathomics signatures in predicting disease-free survival (DFS) (C-index: training dataset, 0.744 vs. 0.734 and 0.692; testing dataset 1, 0.719 vs. 0.701 and 0.638; testing dataset 2, 0.711 vs. 0.689 and 0.684), demonstrating greater robustness compared to the state-of-the-art deep learning integration approaches. It provided additional prognostic information beyond clinical risk factors (C-index of clinical plus radiopathomics vs. clinical models: training dataset, 0.763 vs. 0.676; testing dataset 1, 0.739 vs. 0.676; testing dataset 2, 0.711 vs. 0.699, p < 0.001). Compared to low-risk patients categorised by the radiopathomics signature, high-risk patients achieved comparable DFS when receiving adjuvant chemotherapy (training dataset, HR = 1.53, 95 % CI 0.85-2.73, p = 0.153; testing dataset 1 and 2, HR = 1.62, 95 % CI 0.92-2.85, p = 0.096), but had significantly worse DFS when only observed after surgery (training dataset, HR = 4.46, 95 % CI 2.82-7.05, p < 0.001; testing datasets 1 and 2, HR = 3.52, 95 % CI 2.26-5.49, p < 0.001), indicating the predictive value of the radiopathomics signature for adjuvant chemotherapy benefit (interaction p < 0.05). Further interpretability analysis revealed that the radiopathomics signature was associated with various prognostic/treatment-related biomarkers, including differentiation, immune phenotypes, and EGFR status. The multimodal integration framework offered a cost-effective approach for LUAD characterisation by leveraging complementary information from radiological and histopathological imaging. The radiopathomics signature demonstrated robust prognostic capabilities, providing valuable insights for postoperative treatment decisions.
Collapse
Affiliation(s)
- Huan Lin
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Junjie Hua
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhengze Gong
- Information and Data Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Mingwei Chen
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Bingjiang Qiu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Sciences, Guangzhou, 510080, China
| | - Yuxin Wu
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Wenfeng He
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Yumeng Wang
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Zhengyun Feng
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Yanting Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Wansheng Long
- Department of Radiology, Jiangmen Central Hospital, Jiangmen, 529030, China
| | - Ronggang Li
- Department of Pathology, Jiangmen Central Hospital, Jiangmen, 529030, China
| | - Qionglian Kuang
- Department of Radiology, Jiangmen Central Hospital, Jiangmen, 529030, China
| | - Yingxin Chen
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Jiawei Lu
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Shiwei Luo
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Wei Zhao
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Lixu Yan
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Zhenwei Shi
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zeyan Xu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Ziyang Mo
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Entao Liu
- WeiLun PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Chu Han
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yanfen Cui
- Department of Radiology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Xiaotang Yang
- Department of Radiology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China.
| | - Xiangmeng Chen
- Department of Radiology, Jiangmen Central Hospital, Jiangmen, 529030, China.
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Xipeng Pan
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China.
| | - Anant Madabhushi
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| | - Cheng Lu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China.
| |
Collapse
|
157
|
Kim HD, Kim SY, Lee H, Lee Y, Hyung J, Moon M, Shin J, Park YS, Ryu MH. Predictive value of EBV-positivity in patients with gastric cancer treated with first-line nivolumab plus chemotherapy. Gastric Cancer 2025:10.1007/s10120-025-01618-6. [PMID: 40295365 DOI: 10.1007/s10120-025-01618-6] [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: 10/27/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Epstein-Barr virus (EBV) positivity is a potential predictive biomarker for immune checkpoint inhibitors (ICIs) in gastric cancer patients, but its value in first-line ICI-based chemotherapy remains unclear. This study aimed to evaluate the predictive value of EBV positivity in patients treated with first-line nivolumab plus chemotherapy. METHODS This single-center study included advanced gastric cancer patients treated with first-line nivolumab plus chemotherapy (n = 293). Patients with EBV positivity treated with chemotherapy alone (n = 12) served as the control group. EBV positivity was confirmed by in situ hybridization. RESULTS Among patients treated with nivolumab plus chemotherapy, 18 (6.1%) had EBV-positive tumors, and these were associated with high PD-L1 combined positive score (CPS) expression levels. Progression-free survival (PFS) and overall survival (OS) tended to be more favorable in those with EBV-positive tumors. Multivariate analysis of patients treated with nivolumab-chemotherapy revealed that EBV positivity, combined with PD-L1 CPS ≥ 5, was an independent factor for PFS. In patients with EBV-positive tumors, nivolumab-chemotherapy was associated with significantly favorable PFS and OS compared to chemotherapy alone. Similar results were observed in the subgroup with PD-L1 CPS ≥ 5. However, survival outcomes did not differ between patients treated with nivolumab plus chemotherapy versus chemotherapy alone in the subgroup with PD-L1 CPS < 5. CONCLUSION EBV positivity predicts favorable survival outcomes in patients with gastric cancer treated with nivolumab plus chemotherapy. The benefit of nivolumab plus chemotherapy over chemotherapy alone for patients with EBV positivity appears to be associated with high PD-L1 expression levels.
Collapse
Affiliation(s)
- Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - So-Yeon Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Hyungeun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Yuna Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Jaewon Hyung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Meesun Moon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Jinho Shin
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
158
|
Avsec E, Blatnik A, Krajc M. Secondary findings in hereditary cancer genes after germline genetic testing - systematic review of literature. Hum Genet 2025:10.1007/s00439-025-02746-w. [PMID: 40293485 DOI: 10.1007/s00439-025-02746-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
In the last decade the increasing use of germline genetic testing has led to frequent discoveries of secondary findings (SF) in hereditary cancer (HC) genes. Disclosure and clinical management of such findings are still not clearly defined and raise many ethical, clinical, and practical questions. This systematic review is focused on frequency of reported SF in HC genes across different populations as well as summarizing current guidelines, recommendations, and actual clinical practice about reporting and managing SF in HC genes. A systematic literature search according to the PRISMA guidelines was performed on the electronic database PubMed from inception to June 2024. 30 research papers involving almost 150,000 patients were reviewed. The reported frequencies of SF in HC genes varied between 0.4 and 3.1%. The majority of patients agreed to receive SF for medically actionable genes. Management and surveillance of patients after disclosure of SF in HC genes were rarely reported, but the limited data show no regret of receiving such results as well as diagnoses of early-stage cancer in patients participating in recommended surveillance programs related to SF. A substantial number of carriers of highly penetrant pathogenic variants in HC genes is discovered by reporting SF after germline genetic testing with next-generation sequencing. Additional information about the impact of SF disclosure on individuals and health care systems is needed to optimize the integration of SF into clinical care.
Collapse
Affiliation(s)
- Eva Avsec
- Department of Clinical Cancer Genetics, Institute of Oncology Ljubljana, Zaloška 2, Ljubljana, 1000, Slovenia
- Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, 1000, Slovenia
| | - Ana Blatnik
- Department of Clinical Cancer Genetics, Institute of Oncology Ljubljana, Zaloška 2, Ljubljana, 1000, Slovenia
- Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, 1000, Slovenia
| | - Mateja Krajc
- Department of Clinical Cancer Genetics, Institute of Oncology Ljubljana, Zaloška 2, Ljubljana, 1000, Slovenia.
- Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, 1000, Slovenia.
| |
Collapse
|
159
|
Li J, Li S, Zhang Y, Ye S, Liu R, Shi W. The Efficacy and Safety of Nivolumab Combined with Nab-Paclitaxel or Oxaliplatin as a First-Line Treatment for Advanced or Metastatic Gastric Cancer and Gastroesophageal Junction Cancer. J Gastrointest Cancer 2025; 56:109. [PMID: 40293495 PMCID: PMC12037671 DOI: 10.1007/s12029-025-01211-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2025] [Indexed: 04/30/2025]
Abstract
OBJECTIVE This study aims to assess the therapeutic efficacy and safety of nivolumab combined with chemotherapy as a first-line treatment for advanced or metastatic gastric cancer, specifically comparing the outcomes of oxaliplatin-based versus albumin-bound paclitaxel (nab-paclitaxel)-based therapies. METHODS We retrospectively analyzed 93 patients with advanced gastric cancer or gastroesophageal junction adenocarcinoma treated at the First Medical Center of Chinese PLA General Hospital from September 2017 to November 2022. Patients were categorized into the nivolumab + oxaliplatin (N-OX group) or nivolumab + nab-paclitaxel (N-AP group) based on the chemotherapy regimen. Progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and safety were evaluated as endpoints. RESULTS At the end of the follow-up period on September 31, 2023, we reported an ORR of 65.6% and DCR of 95.7% across all patients. The median PFS was 8.4 months, with no significant difference between the N-OX and N-AP groups (median, 7.8 vs 9.5 months; P = 0.450). Notably, patients with diffuse gastric cancer in N-AP group showed a 44.7% reduction in tumor progression risk compared with the N-OX group (P = 0.046). The overall safety profile was acceptable in two groups. CONCLUSIONS Our study suggested that nivolumab combined with chemotherapy was effective and safe as a first-line intervention for advanced gastric cancer. While both oxaliplatin and nab-paclitaxel regimens showed similar efficacy, the nab-paclitaxel may offer additional benefits for patients with diffuse gastric cancer. Further research is encouraged to confirm these findings and refine treatment strategies.
Collapse
Affiliation(s)
- Juan Li
- Department of Oncology, First Medical Center, General Hospital of the People'S Liberation Army, Beijing, 100089, China
| | - Shuman Li
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Ying Zhang
- Department of Oncology, First Medical Center, General Hospital of the People'S Liberation Army, Beijing, 100089, China
| | - Sisi Ye
- Department of Oncology, First Medical Center, General Hospital of the People'S Liberation Army, Beijing, 100089, China
| | - Rongrui Liu
- Department of Oncology, First Medical Center, General Hospital of the People'S Liberation Army, Beijing, 100089, China
| | - Weiwei Shi
- Department of Oncology, First Medical Center, General Hospital of the People'S Liberation Army, Beijing, 100089, China.
| |
Collapse
|
160
|
Qi TT, Zhou SJ, Yu Z, Li Y, Chen JQ. Unveiling the heterogeneity and immunotherapy potency of tumor-associated neutrophils in the tumor microenvironment of gastric cancer. BMC Gastroenterol 2025; 25:303. [PMID: 40295944 PMCID: PMC12036284 DOI: 10.1186/s12876-025-03920-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The differentiation characteristics of neutrophils within the gastric cancer (GC) tumor microenvironment (TME) and their interactions with malignant gastric epithelial cells require further investigation. Furthermore, the therapeutic potential of tumor-associated neutrophils (TANs) in immunotherapy remains inadequately explored. METHODS We integrated two single-cell transcriptome datasets comprising 12 samples, including gastric primary tumors, non-tumor tissues, and metastatic tumors, to profile the epithelial cells and TANs atlas within the TME and examine their interaction modules. In addition, these data were integrated with the bulk transcriptomic including the Cancer Genome Atlas - Stomach Adenocarcinoma (TCGA-STAD) and Asian Cancer Research Group (ACRG) datasets to analyze the expression levels of neutrophil-associated genes across the tumor-associated neutrophil subsets. RESULTS We analyzed 3,118 gastric epithelial cells and 2,365 TANs from all samples. Epithelial cells were classified into ten subclusters, while TANs were grouped into five subclusters. In gastric primary tumors, epithelial cell subtypes included primarily MUC16 + and stem-like populations. In metastatic tumors, the epithelial cell subset with high CXCL5 expression was a characteristic subtype. TANs mainly interacted with epithelial cells via the LGALS9-CD45 and CD46-JAG1 pathways. And RGS2 was highly expressed in N4, a tumor-associated neutrophils subcluster characterized by high MMP9 expression, highlighting its potential as an immunotherapy target. CONCLUSION TANs exhibit robust interactions with gastric malignant epithelial cell subsets. Furthermore, RGS2, which is highly expressed in N4, could serve as a promising target for immunotherapy.
Collapse
Affiliation(s)
- Tong-Tong Qi
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery After Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
| | - Si-Jiang Zhou
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery After Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
| | - Zhu Yu
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery After Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
| | - Yong Li
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Guangxi Clinical Research Center for Enhanced Recovery After Surgery, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China
- Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun-Qiang Chen
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China.
- Guangxi Clinical Research Center for Enhanced Recovery After Surgery, Nanning, China.
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, Nanning, China.
| |
Collapse
|
161
|
Aoyama T, Ooki A, Oba K, Nishikawa K, Kawabata R, Honda M, Maeda H, Kanda M, Sugiyama K, Makiyama A, Segami K, Takahashi M, Shindo Y, Namikawa T, Oshima T, Katayama A, Shiosakai K, Sakamoto J. A multicenter randomized open-label phase 2 study investigating optimal antiemetic therapy for patients with advanced/recurrent gastric cancer treated with trastuzumab deruxtecan: the EN-hance study. Int J Clin Oncol 2025:10.1007/s10147-025-02748-8. [PMID: 40293658 DOI: 10.1007/s10147-025-02748-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/14/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Trastuzumab deruxtecan (T-DXd) has been approved for the treatment of human epidermal growth factor receptor-2 (HER2)-positive gastric cancer and other indications in several countries and is considered moderately or highly emetogenic. The management of nausea and vomiting associated with T-DXd treatment has not been fully evaluated and the effectiveness of conventional prophylaxis remains unknown. METHODS This open-label, randomized, multicenter, phase 2 study aimed to investigate the optimal antiemetic therapy for Japanese patients with gastric cancer undergoing T-DXd treatment. Patients were randomized to a doublet regimen group (dexamethasone and palonosetron) or triplet regimen group (aprepitant, dexamethasone, and palonosetron) at a ratio of one to one, stratified by sex, gastrectomy status, and study institution. Both antiemetic treatments were administered from day 1 before T-DXd administration, and emetic events and nausea were observed for 21 days. The primary endpoint was the antiemetic complete response (CR) rate to assess control for emetic events based on voluntary patient-reported outcomes (PROs) during cycle 1 (1-21 days). RESULTS Of the 60 enrolled patients, 58 were eligible for inclusion in this analysis (29 patients in each regimen group). The overall CR rates for the doublet and triplet regimens were 41.4% (12/29 patients) and 37.9% (11/29 patients), respectively, and neither regimen met the pre-specified threshold (> 18/29 patients). The CR rate in the acute phase (0-24 h) was 86.2% (25/29 patients) for both regimens, and the CR rates in the delayed phase (2-21 days) were 41.4% (12/29 patients) and 37.9% (11/29 patients) for the doublet and triplet regimens, respectively. CONCLUSIONS Given that the primary endpoint was not met, further research is needed to better characterize nausea and vomiting with T-DXd to tailor an anti-emetic regimen that suits the needs of the patients.
Collapse
Affiliation(s)
- Toru Aoyama
- Department of Surgery, Yokohama City University Hospital, Yokohama, Japan.
- Department of Gastric Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan.
| | - Akira Ooki
- The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto, Japan
| | - Koji Oba
- The University of Tokyo Graduate School of Medicine, Bunkyō, Japan
| | | | | | | | | | - Mitsuro Kanda
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | - Kenki Segami
- Saiseikai Yokohama-Shi Nanbu Hospital, Yokohama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
162
|
Cercek A, Foote MB, Rousseau B, Smith JJ, Shia J, Sinopoli J, Weiss J, Lumish M, Temple L, Patel M, Wilde C, Saltz LB, Argiles G, Stadler Z, Artz O, Maron S, Ku G, Gu P, Janjigian YY, Molena D, Iyer G, Coleman J, Abida W, Cohen S, Soares K, Schattner M, Strong VE, Yaeger R, Paty P, Shcherba M, Sugarman R, Romesser PB, Zervoudakis A, Desai A, Segal NH, El Dika I, Widmar M, Wei I, Pappou E, Fumo G, Aparo S, Gonen M, Gollub M, Jayaprakasham VS, Kim TH, Garcia Aguilar J, Weiser M, Diaz LA. Nonoperative Management of Mismatch Repair-Deficient Tumors. N Engl J Med 2025. [PMID: 40293177 DOI: 10.1056/nejmoa2404512] [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] [Indexed: 04/30/2025]
Abstract
BACKGROUND Among patients with mismatch repair-deficient (dMMR), locally advanced rectal cancer, neoadjuvant checkpoint blockade eliminated the need for surgery in a high proportion of patients. Whether this approach can be extended to all early-stage dMMR solid tumors, regardless of tumor site, is unknown. METHODS We conducted a phase 2 study in which patients with stage I, II, or III dMMR solid tumors that were amenable to curative-intent surgery were treated with neoadjuvant dostarlimab, a programmed cell death 1 (PD-1) blocking agent, for 6 months. The response to treatment was assessed in two cohorts: patients in cohort 1 had dMMR, locally advanced rectal cancer, and patients in cohort 2 had dMMR nonrectal solid tumors. Patients with a clinical complete response could elect to proceed with nonoperative management; those with residual disease were to undergo resection. In this analysis, the primary end point, assessed in cohort 1, was a sustained clinical complete response at 12 months. Recurrence-free survival and safety were evaluated. RESULTS A total of 117 patients were included in the analysis. In cohort 1, all 49 patients who completed treatment had a clinical complete response and elected to proceed with nonoperative management. A total of 37 patients had a sustained clinical complete response at 12 months, a finding that met the criterion for efficacy. In cohort 2, a total of 35 of 54 patients who completed treatment had a clinical complete response, and 33 elected to proceed with nonoperative management. Among the 103 patients who completed treatment across both cohorts, 84 had a clinical complete response, and 82 did not undergo surgery. Among the 117 total patients, recurrence-free survival at 2 years was 92% (95% confidence interval, 86 to 99); the median follow-up for recurrence was 20.0 months (range, 0 to 60.8). The majority of patients (95%) had reversible, grade 1 or 2 adverse events (60%) or had no adverse events (35%). The option for curative resection was not compromised during or after treatment in any of the patients. CONCLUSIONS Among patients with early-stage dMMR solid tumors that were amenable to curative-intent surgery, neoadjuvant PD-1 blockade led to organ preservation in a high proportion of patients. (Funded by Swim Across America and others; ClinicalTrials.gov number, NCT04165772.).
Collapse
Affiliation(s)
- Andrea Cercek
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Michael B Foote
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Benoit Rousseau
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - J Joshua Smith
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - Jenna Sinopoli
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Jill Weiss
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Melissa Lumish
- Department of Oncology, Case Comprehensive Cancer Center, Cleveland
| | - Lindsay Temple
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Miteshkumar Patel
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Callahan Wilde
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Leonard B Saltz
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Guillem Argiles
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Zsofia Stadler
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Oliver Artz
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Steven Maron
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Geoffrey Ku
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Ping Gu
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Yelena Y Janjigian
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Daniela Molena
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Gopa Iyer
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Jonathan Coleman
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Wassim Abida
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Seth Cohen
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Kevin Soares
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Mark Schattner
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Vivian E Strong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Rona Yaeger
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Philip Paty
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Marina Shcherba
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Ryan Sugarman
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Alice Zervoudakis
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Avni Desai
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Neil H Segal
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Imane El Dika
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Maria Widmar
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Iris Wei
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Emmanouil Pappou
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Gerard Fumo
- Department of Oncology, Hartford HealthCare, Hartford, CT
| | - Santiago Aparo
- Department of Oncology, Baptist Health Miami Cancer Institute, Miami
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Marc Gollub
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York
| | | | - Tae-Hyung Kim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York
| | | | - Martin Weiser
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Luis A Diaz
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| |
Collapse
|
163
|
Kret ZS, Sweder RJ, Pollock R, Tinoco G. Potential Mechanisms for Immunotherapy Resistance in Adult Soft-Tissue Sarcoma. Target Oncol 2025:10.1007/s11523-025-01145-5. [PMID: 40289241 DOI: 10.1007/s11523-025-01145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Soft-tissue sarcomas represent a diverse group of rare malignancies originating from mesenchymal tissue, accounting for less than 1% of adult cancers in the USA. With over 13,000 new cases and around 5350 deaths annually, patients with metastatic soft-tissue sarcomas face limited therapeutic options and an estimated median overall survival of 18 months. While immunotherapy has demonstrated effectiveness in several cancers, its application in soft-tissue sarcomas remains challenging owing to the tumors' largely "cold" immunological environment, characterized by low levels of tumor-infiltrating lymphocytes and a lack of soft-tissue sarcoma-specific biomarkers. This review examines potential mechanisms underlying immunotherapy resistance in soft-tissue sarcomas, including the complex interplay between innate and adaptive immunity, the tumor microenvironment, and the role of immune-related genes. Despite preliminary findings suggesting correlations between immune profiles and histological subtypes, consistent biomarkers for predicting immunotherapeutic responses across soft-tissue sarcoma types are absent. Emerging strategies focus on converting "cold" tumors to "hot" tumors, enhancing their susceptibility to immunologic activation. While research is ongoing, personalized treatment approaches may offer hope for overcoming the inherent heterogeneity and resistance seen in soft-tissue sarcomas, ultimately aiming to improve outcomes for affected patients.
Collapse
Affiliation(s)
- Zaina S Kret
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ryan J Sweder
- The Ohio State University College of Arts and Sciences and College of Medicine, Columbus, OH, USA
| | - Raphael Pollock
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gabriel Tinoco
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 1800 Cannon Drive, 1240 Lincoln Tower, Columbus, OH, 43210, USA.
| |
Collapse
|
164
|
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 2025:10.1007/s10120-025-01617-7. [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] [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
|
165
|
Jiménez-Labaig P, Lorini L, Gurizzan C, Kinloch E, Burton S, Forster MD, Metcalf R, Ferrarotto R, Bossi P, O Leary B, Hanna G, Felip E, Garcia IB, Harrington KJ. Clinical trials for patients with salivary gland cancers: A systematic review of worldwide registers and an evaluation of current challenges. Crit Rev Oncol Hematol 2025; 211:104747. [PMID: 40294876 DOI: 10.1016/j.critrevonc.2025.104747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Clinical trials (CT) are crucial for generating scientific evidence and improving clinical outcomes, but they can be challenging in the context of rare cancers. Salivary gland cancers (SGC) are rare and heterogeneous tumors, without standard-of-care approved systemic therapies. We analyzed completed and ongoing CTs to assess the current state of clinical research activity in the field. METHODS ClinicalTrials.gov, WHO-ICTRP, HealthCanadaCT were searched for antineoplastic pharmacological and interventional CT involving patients with SGC from the trials database creation until August 6th, 2024. CT characteristics and status were collected. RESULTS 134 clinical trials met inclusion criteria. Of these, 78 % were sponsored by non-industry entities. 49 % were conducted at only one site, and 61 % at up to five centers. Only 25 trials (19 %) were multinational, being 15 industry-sponsored, a significantly higher proportion compared to non-industry-sponsored trials(p < 0.01). 16 % CTs were umbrella or basket, and 6 % were randomized, again predominantly industry-sponsored(p < 0.01). Regarding SGC-specific trials, 32 % were open to all patients with SGC, regardless of specific histology. Patients with adenoid cystic, salivary duct, and mucoepidermoid carcinoma had access to 92 %, 66 % and 62 % of trials, respectively. 88 % CT targeted palliative setting, and 38 % incorporated predictive biomarkers. Tyrosine kinase inhibitors were the most studied therapy(26 %), followed by immunotherapy(15 %), chemotherapy and antibody-drug conjugate(12 % each) and androgen-blockade(8 %), among others. CONCLUSION Clinical research for patients with SGC relies mainly in non-industry organisations, most of them limited to run trials in one to five sites, in a single country. Further collaboration between investigators is needed, as well as reconsidering inclusion criteria and trial designs.
Collapse
Affiliation(s)
- Pablo Jiménez-Labaig
- Head and Neck Unit. The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom; Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Luigi Lorini
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Cristina Gurizzan
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Emma Kinloch
- Salivary Gland Cancer UK, London, United Kingdom
| | - Sarah Burton
- Head and Neck Unit. The Royal Marsden NHS Foundation Trust, London, United Kingdom; The International Centre for Recurrent Head and Neck Cancer, United Kingdom
| | - Martin D Forster
- UCL Cancer Institute, London, United Kingdom; University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Robert Metcalf
- Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom; The Cancer Research UK Manchester Institute, United Kingdom
| | - Renata Ferrarotto
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, United States
| | - Paolo Bossi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, MI, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan 20072, Italy
| | - Ben O Leary
- Head and Neck Unit. The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom
| | - Glenn Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, United States
| | - Enriqueta Felip
- Lung and Head & Neck Tumors Unit. Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Irene Braña Garcia
- Lung and Head & Neck Tumors Unit. Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Kevin J Harrington
- Head and Neck Unit. The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom
| |
Collapse
|
166
|
Doi T, Ishikawa T, Moriguchi M, Itoh Y. Current status of cancer genome medicine for pancreatic ductal adenocarcinoma. Jpn J Clin Oncol 2025; 55:443-452. [PMID: 39893577 DOI: 10.1093/jjco/hyaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis; however, advancements in cancer genome profiling using next-generation sequencing have provided new perspectives. KRAS mutations are the most frequently observed genomic alterations in patients with PDAC. However, until recently, it was not considered a viable therapeutic target. Although KRAS G12C mutations for which targeted therapies are already available are infrequent in PDAC, treatments targeting KRAS G12D and pan-KRAS are still under development. Similarly, new treatment methods for KRAS, such as chimeric antigen receptor T-cell therapy, have been developed. Several other potential therapeutic targets have been identified for KRAS wild-type PDAC. For instance, immune checkpoint inhibitors have demonstrated efficacy in PDAC treatment with microsatellite instability-high/deficient mismatch repair and tumor mutation burden-high profiles. However, for other PDAC cases with low immunogenicity, combination therapies that enhance the effectiveness of immune checkpoint inhibitors are being considered. Additionally, homologous recombination repair deficiencies, including BRCA1/2 mutations, are prevalent in PDAC and serve as important biomarkers for therapies involving poly (adenosine diphosphate-ribose) polymerase inhibitors and platinum-based therapies. Currently, olaparib is available for maintenance therapy of BRCA1/2 mutation-positive PDAC. Further therapeutic developments are ongoing for genetic abnormalities involving BRAF V600E and the fusion genes RET, NTRK, NRG, ALK, FGFR2, and ROS1. Overcoming advanced PDAC remains a formidable challenge; however, this review outlines the latest therapeutic strategies that are expected to lead to significant advancements.
Collapse
Affiliation(s)
- Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Medical Oncology Unit, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michihisa Moriguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
167
|
Kan L, Yu Y, Wang Y, Shi L, Fan T, Chen H, Ren C. The application of organoids in investigating immune evasion in the microenvironment of gastric cancer and screening novel drug candidates. Mol Cancer 2025; 24:125. [PMID: 40287758 PMCID: PMC12032790 DOI: 10.1186/s12943-025-02328-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Gastric cancer (GC) is a prevalent digestive system tumor, the fifth most diagnosed cancer worldwide, and a leading cause of cancer deaths. GC is distinguished by its pronounced heterogeneity and a dynamically evolving tumor microenvironment (TME). The lack of accurate disease models complicates the understanding of its mechanisms and impedes the discovery of novel drugs. A growing body of evidence suggests that GC organoids, developed using organoid culture technology, preserve the genetic, phenotypic, and behavioral characteristics. GC organoids hold significant potential for predicting treatment responses in individual patients. This review provides a comprehensive overview of the current clinical treatment strategies for GC, as well as the history, construction and clinical applications of organoids. The focus is on the role of organoids in simulating the TME to explore mechanisms of immune evasion and intratumoral microbiota in GC, as well as their applications in guiding clinical drug therapy and facilitating novel drug screening. Furthermore, we summarize the limitations of GC organoid models and underscore the need for continued technological advancements to benefit both basic and translational oncological research.
Collapse
Affiliation(s)
- Liuyue Kan
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Ying Yu
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yaxue Wang
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Lei Shi
- Department of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98 Western Nantong Road, Yangzhou, 225001, China
| | - Tingyuan Fan
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Hui Chen
- Department of Geriatrics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98 Western Nantong Road, Yangzhou, 225001, China.
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98, Western Nantong Road, Yangzhou, 225001, China.
| | - Chuanli Ren
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
- Department of Laboratory Medicine, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China.
- The Yangzhou Clinical Medical College of Xuzhou Medical University, No. 98, Western Nantong Road, Yangzhou, 225001, China.
| |
Collapse
|
168
|
Zhong C, Yuan Y, Jiang Y, Qiao G, Deng Z, Liu Z, Yu L, Lin H, Mao J, Ma L, Zhang J. Predictive role of inflammatory markers for the efficacy of first-line immunotherapy plus chemotherapy in advanced gastric cancer. Discov Oncol 2025; 16:618. [PMID: 40285934 PMCID: PMC12033139 DOI: 10.1007/s12672-025-01857-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/28/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) plus chemotherapy have become a new first-line treatment option for patients with locally advanced or metastatic gastric cancer. However, it is still controversial whether to choose chemotherapy alone or ICIs plus chemotherapy as the first-line treatment option due to a lack of ideal predictive biomarkers for the efficacy. This study intended to explore the predictive value of inflammatory markers for the efficacy of first-line ICIs plus chemotherapy in this disease. METHODS This retrospective study included 131 patients with locally advanced or metastatic gastric cancer who received first-line treatment between July, 2020 and June, 2023. Among them, 76 received first-line ICIs plus chemotherapy and 55 received chemotherapy alone. Firstly, Kaplan-Meier and Cox regression analyses were used to explore the correlation between inflammatory markers and efficacy of first-line ICIs plus chemotherapy. Subsequently, the predictive value of combined baseline and dynamic changes in inflammatory markers was explored. Moreover, the predictive value of baseline inflammatory markers was further verified by comparing efficacy of ICIs plus chemotherapy with that of chemotherapy alone. RESULTS In patients receiving first-line ICIs plus chemotherapy, low baseline monocyte-to-lymphocyte ratio (MLR) in peripheral blood was significantly associated with better progression-free survival (PFS) and overall survival (OS), and was an independent prognostic factor for OS. In addition, dynamic early changes of MLR also played predictive role. Patients whose MLR was lower at baseline and after two cycles of treatment had better OS (P = 0.009). Furthermore, compared to chemotherapy alone, patients with a lower baseline MLR were more likely to benefit from first-line ICIs plus chemotherapy. CONCLUSION MLR could serve as a new biomarker to predict the efficacy of first-line ICIs plus chemotherapy in patients with locally advanced or metastatic gastric cancer. And it is helpful to select the candidates for first-line ICIs plus chemotherapy, which is worthy of further study.
Collapse
Affiliation(s)
- Chenming Zhong
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Yuan
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Jiang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanglei Qiao
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhoufeng Deng
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zimei Liu
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Yu
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjian Lin
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiuang Mao
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijun Ma
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianjun Zhang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
169
|
Yamazawa S, Fukasawa-Hokazono M, Takase A, Kondo A, Matsubara J, Shinozaki-Ushiku A, Seto Y, Ushiku T. Immune evasion strategies in AFP-producing gastric carcinoma: characterized by HLA-G expression and HLA class I deficiency. Virchows Arch 2025:10.1007/s00428-025-04108-3. [PMID: 40278871 DOI: 10.1007/s00428-025-04108-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/28/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
Alpha-fetoprotein-producing gastric carcinoma (AFPGC) is an aggressive subtype of gastric cancer characterized by a primitive cellular phenotype and poor prognosis. The tumor immunology of AFPGC remains largely unexplored. Given its embryonic-like properties, AFPGC is hypothesized to employ distinct immune evasion strategies, with the oncofetal protein human leukocyte antigen (HLA)-G-a key mediator of maternal-fetal immune tolerance-likely playing a pivotal role. To test this, we assessed the expression of HLA-G, along with other key immune evasion markers, including HLA class I (HLA-I) deficiency and PD-L1 expression, in 39 cases of AFPGC, and compared them with those of 44 Epstein-Barr virus (EBV)-positive, 57 microsatellite instability (MSI), 54 intestinal-type, and 45 diffuse-type gastric carcinomas. HLA-G expression was significantly higher in AFPGCs (71%) than in other subtypes (7-28%; P < 0.001). HLA-I deficiency (≥ 1% of tumor cells) was most prevalent in AFPGC (69%), followed by MSI tumors (56%), with lower rates in other subtypes (22-29%). PD-L1 positivity (combined positive score ≥ 5) was observed in 41% of AFPGCs, lower than in EBV-positive (77%) and MSI tumors (44%), but higher than in intestinal-type (13%) and diffuse-type (9%) carcinomas. Furthermore, CD8-positive T-cell infiltration was found to be lowest in AFPGC compared to the other subtypes. These findings suggest that AFPGC employs multiple immune evasion mechanisms, notably through increased HLA-G expression and HLA-I deficiency, likely linked to its primitive cellular phenotype and reactivation of immunogenic oncofetal antigens. Such immune evasion features may underlie the aggressiveness of AFPGC and present promising targets for immunotherapeutic interventions.
Collapse
Affiliation(s)
- Sho Yamazawa
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Akiko Takase
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsushi Kondo
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Joji Matsubara
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Aya Shinozaki-Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Integrative Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
170
|
Yamada A, Kondo T. Hereditary Colorectal Cancer: Clinical Implications of Genomic Medicine and Precision Oncology. J Anus Rectum Colon 2025; 9:167-178. [PMID: 40302859 PMCID: PMC12035340 DOI: 10.23922/jarc.2025-001] [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: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 05/02/2025] Open
Abstract
Approximately 10% of colorectal cancer (CRC) cases occur in the context of hereditary cancer-predisposing conditions caused by germline pathogenic variants (PVs) in cancer predisposition genes, with Lynch syndrome and familial adenomatous polyposis at the top of the list. Although the identification of hereditary CRC has traditionally relied on clinical characteristics, including familial accumulation, multiple and early onset of CRC and other related cancers, and the presence of gastrointestinal polyposis, more comprehensive approaches, such as universal tumor screening and universal germline testing, have recently been employed. From a technical standpoint, next-generation sequencing has enabled genome-wide analysis of genetic alterations in germline and somatic settings. Taking advantage of this technology, germline multigene panel testing has been utilized in genetic testing, which leads to the identification of PVs, not only in well-known hereditary CRC genes but also in rare causal genes, moderate-risk genes, and high-risk genes previously not linked to CRC predisposition. In addition, comprehensive genomic profiling and companion diagnostics for solid tumors occasionally yield unexpected hereditary CRC diagnoses. Thus, more hereditary CRCs have been identified not based on clinical phenotypes but rather by comprehensive approaches or as secondary findings of treatment drug testing. In this review, we discuss the impact of recent advances in genomic medicine on the clinical aspects of hereditary CRC, which has promoted an understanding of the entire landscape of genetic predisposition to CRC.
Collapse
Affiliation(s)
- Atsushi Yamada
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
- Department of Real-World Data Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
171
|
Tang S, Che X, Wang J, Li C, He X, Hou K, Zhang X, Guo J, Yang B, Li D, Cao L, Qu X, Wang Z, Liu Y. T-bet +CD8 + T cells govern anti-PD-1 responses in microsatellite-stable gastric cancers. Nat Commun 2025; 16:3905. [PMID: 40280928 PMCID: PMC12032036 DOI: 10.1038/s41467-025-58958-1] [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: 04/25/2024] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
More than 90% of advanced gastric cancers (GC) are microsatellite-stable (MSS). Compared to the high response rate of immune checkpoint inhibitors (ICI) in microsatellite-instability-high (MSI-H) GCs, only 10% of unstratified MSS GCs respond to ICIs. In this study, we apply semi-supervised learning to stratify potential ICI responders in MSS GCs, achieving high accuracy, quantified by an area under the curve of 0.924. Spatial analysis of the tumor microenvironment of ICI-sensitive GCs reveals a high level of T-bet+ CD8 + T cell infiltration in their tumor compartments. T-bet+ CD8 + T cells exhibit superior anti-tumor activity due to their increased ability to infiltrate tumors and secrete cytotoxic molecules. Adoptive transfer of T-bet+ CD8 + T cells boosts anti-tumor immunity and confers susceptibility to ICIs in immune-ignorant MSS GCs in a humanized mouse model. Spatial RNA sequencing suggests a positive-feedback loop between T-bet+ T cells and PD-L1+ tumor cells, which eventually drives T cell exhaustion and can therefore be leveraged for ICI therapy. In summary, our research provides insights into the underlying mechanism of anti-tumor immunity and deepens our understanding of varied ICI responses in MSS GCs.
Collapse
Affiliation(s)
- Shiying Tang
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Jinyan Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No. 77, Puhe Road, Shenyang, Liaoning, China
| | - Ce Li
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Xin He
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Kezuo Hou
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Xiaojie Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Jia Guo
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Bowen Yang
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Danni Li
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Lili Cao
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China.
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, No.155, Nanjing Street, Shenyang, Liaoning, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, China Medical University, Shenyang, Liaoning, China.
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China.
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China.
| |
Collapse
|
172
|
Inokawa Y, Mizuno H, Yamada M, Kawakatsu S, Watanabe N, Onoe S, Mizuno T, Okayama K, Okumura F, Kajikawa M, Ebata T. Pathological Complete Response after Pembrolizumab Treatment for Unresectable Perihilar Cholangiocarcinoma with High Microsatellite Instability: A Case Report. Surg Case Rep 2025; 11:25-0025. [PMID: 40308703 PMCID: PMC12041437 DOI: 10.70352/scrj.cr.25-0025] [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: 01/20/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
INTRODUCTION Pembrolizumab has been introduced to solid cancers with microsatellite instability (MSI)-high cases; however, its clinical experience for cholangiocarcinoma remains very limited. Here, we present a case who successfully underwent conversion surgery following pembrolizumab treatment for MSI-high perihilar cholangiocarcinoma, which pathologically exhibited complete response. CASE PRESENTATION A 69-year-old male with Bismuth IV perihilar cholangiocarcinoma with bulky lymphadenopathy was referred, who initially required left hepatic trisectionectomy, caudate lobectomy, bile duct resection, and portal vein resection and reconstruction (H123458-B-PV). During the waiting period after preoperative portal vein embolization, the right hepatic artery was involved by rapid tumor progression, needing a modification of the initially scheduled surgical procedure to additional hepatic artery resection and reconstruction (H123458-B-PV-HA). We revised the surgical decision of resectable to locally unresectable disease. He received systemic chemotherapy with gemcitabine and cisplatin as first-line, showing the best effect of stable disease followed by slight tumor progression and re-elevation of tumor marker after 5 courses of treatment. Cancer multi-gene panel analysis using percutaneous biopsy specimen showed the nature of MSI-high. Therefore, he received pembrolizumab treatment as second-line therapy, leading to a drastic downsize >30% in tumor diameter and normalization of the tumor marker as well after only 2 cycles of administration. After confirmation of keeping tumor shrinkage during 22 courses of pembrolizumab treatment without any severe adverse events, we decided to perform conversion surgery and performed left trisectionectomy, caudate lobectomy, and bile duct resection with portal vein resection (H123458-B-PV). Although the right hepatic artery was extensively fibrotic, there was no evidence of malignancy by frozen section histologic diagnosis. The pathological findings showed pathological complete response with no residual tumor cells. The patient is under periodical checkup without adjuvant chemotherapy, and no tumor recurrence was observed at 4 months postoperatively. CONCLUSIONS We experienced clinical partial response but pathological complete response after second-line pembrolizumab treatment for unresectable locally advanced perihilar cholangiocarcinoma with a biologic nature of MSI-high. Conversion surgery may be considered as a promising option for such effective case, whereas there is a possibility to avoid resection in the MSI-high setting.
Collapse
Affiliation(s)
- Yoshikuni Inokawa
- Department of Surgery, Gifu Prefectural Tajimi Hospital, Tajimi, Gifu, Japan
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hironori Mizuno
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Mihoko Yamada
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shoji Kawakatsu
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Nobuyuki Watanabe
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shunsuke Onoe
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Takashi Mizuno
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kohei Okayama
- Department of Gastroenterology, Gifu Prefectural Tajimi Hospital, Tajimi, Gifu, Japan
| | - Fumihiro Okumura
- Department of Gastroenterology, Gifu Prefectural Tajimi Hospital, Tajimi, Gifu, Japan
| | - Masaki Kajikawa
- Department of Surgery, Gifu Prefectural Tajimi Hospital, Tajimi, Gifu, Japan
| | - Tomoki Ebata
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
173
|
Jiang W, Shi J, Zhu Y, Yin L, Song Y, Zhang J, Lin X, Zhong J, Lu Y, Ma Y. A novel prognostic model based on migrasome-related LncRNAs for gastric cancer. Sci Rep 2025; 15:14572. [PMID: 40281132 PMCID: PMC12032148 DOI: 10.1038/s41598-025-99781-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 04/22/2025] [Indexed: 04/29/2025] Open
Abstract
Gastric cancer (GC) represents a substantial public health challenge, characterized by elevated morbidity and mortality rates. Migrasomes, a newly discovered type of extracellular vesicle, have been highlighted as important contributors to cancer progression, though their specific role in GC remains unclear. To address this issue, we developed the first prognostic model utilizing migrasome-related long non-coding RNAs (MRLs). This model aims to deepen the understanding of GC pathogenesis and improve patient outcomes. Clinical and transcriptional data for 407 GC patients from TCGA were classified as training and testing sets. Through Pearson correlation analysis, 537 MRLs were recognized, and LASSO and Cox regression analyses further refined the list to four key lncRNAs (AC012055.1, LINC01150, AC053503.4, AC107021.2) for constructing the prognostic model. Kaplan-Meier survival analysis indicated a significantly poorer prognosis for the high-risk group. PCA confirmed the model's robustness, and univariate and multivariate analyses validated it as an independent predictor of clinical outcomes. The ROC curve and C-index evaluations further affirmed the model's predictive power. We developed a nomogram combining the MRLs signature with clinical parameters to enhance prognostic accuracy. GO, KEGG and GSEA were performed on migrasome-related genes associated with GC. Furthermore, high-risk patients exhibited increased immune cell infiltration and reduced tumor mutation burden, both associated with poorer outcomes. Additionally, twenty-nine potential therapeutic agents were identified. This novel MRLs-based model provides crucial insights into GC biology and represents a valuable tool for improving patient management and therapeutic strategies.
Collapse
Affiliation(s)
- Wenhao Jiang
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Jiaying Shi
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Yingchuan Zhu
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Lan Yin
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Yue Song
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Jingfei Zhang
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Xinyu Lin
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Jiaxiu Zhong
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Yilu Lu
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China
| | - Yongxin Ma
- Department of Medical Genetics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Cheng Du, 610041, China.
| |
Collapse
|
174
|
Gordon A, Cunningham D, Rajan Z, Fong C, Peckitt C, Satchwell L, Cromarty S, Kidd S, Piadel K, Leamon B, Zhitkov O, Davidson M, Thompson J, Maisey N, Darby S, Waddell T, Morgan C, Bradshaw A, Petty R, Fribbens C, Rao S, Starling N, Chau I. Maintenance Capecitabine Plus Ramucirumab After First-Line Chemotherapy in Patients With Advanced Esophagogastric Adenocarcinoma: Results From the Randomized PLATFORM Study. JCO ONCOLOGY ADVANCES 2025; 2:e2400073. [PMID: 40330143 PMCID: PMC12053389 DOI: 10.1200/oa-24-00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/10/2025] [Accepted: 03/27/2025] [Indexed: 05/08/2025]
Abstract
PURPOSE PLATFORM is an adaptive phase II study assessing maintenance therapies in advanced esophagogastric adenocarcinoma (OGA). We evaluated the role of capecitabine plus a vascular endothelial growth factor receptor 2 inhibitor ramucirumab (cape-ram) in these patients. METHODS Human epidermal growth factor receptor 2 (HER2)-negative patients with advanced OGA with stable or responding disease after 18 weeks of induction platinum-based chemotherapy were randomly assigned 1:1 to surveillance or cape-ram. The primary end point was progression-free survival (PFS), and key secondary end points were overall survival (OS) and safety. Recruitment to the cape-ram arm closed prematurely because of industry support withdrawal. A one-sided log-rank test with a 2.5% significance level was considered significant. RESULTS Between April 2019 and November 2022, 25 surveillance and 22 cape-ram patients were contemporaneously randomly assigned. Median follow-up was 24.4 months. Compared with surveillance, cape-ram significantly prolonged PFS (hazard ratio [HR], 0.33 [95% CI, 0.17 to 0.63], P < .001; median PFS: 2.5 months with surveillance versus 5.5 months with cape-ram; 6-month PFS rate: 4% [95% CI, 0.3% to 17.0%] v 42.9% [95% CI, 21.9% to 62.3%], respectively) and OS (HR, 0.51 [95% CI, 0.26 to 1.00], P = .023; median OS: 7.1 months with surveillance v 14.4 months with cape-ram; median OS from start of induction chemotherapy was 12.1 months v 19.5 months, respectively). Of 10 cape-ram patients with measurable disease, 1 had an incremental partial response. Grade ≥3 adverse events (AEs) were seen in 32% surveillance and 57% cape-ram patients. Six cape-ram patients had grade 3 treatment-related AEs, and no new safety signals were identified. CONCLUSION Maintenance cape-ram after induction chemotherapy for patients with HER2-negative OGA significantly improved survival compared with surveillance. To our knowledge, this is the first randomized maintenance study demonstrating survival benefit and provides support for maintenance treatment.
Collapse
Affiliation(s)
- Anderley Gordon
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - David Cunningham
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Zayn Rajan
- Research Data & Statistics Unit, Royal Marsden Clinical Trials Unit, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Caroline Fong
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Clare Peckitt
- Research Data & Statistics Unit, Royal Marsden Clinical Trials Unit, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Laura Satchwell
- Research Data & Statistics Unit, Royal Marsden Clinical Trials Unit, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Susan Cromarty
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Shannon Kidd
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Katarzyna Piadel
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Becky Leamon
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Oleg Zhitkov
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Michael Davidson
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Joyce Thompson
- Oncology Department, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | | | - Suzanne Darby
- Weston Park Cancer Centre, Sheffield, United Kingdom
| | - Tom Waddell
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Carys Morgan
- Department of Clinical Oncology, Velindre Cancer Centre, Cardiff, United Kingdom
| | | | - Russell Petty
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Charlotte Fribbens
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Sheela Rao
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Naureen Starling
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Ian Chau
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| |
Collapse
|
175
|
Sun M, Gu Y, Wang J, Zhang Z, Ling Z, Shao F, Lin C, He H, Li R, Liu H, Xu J. Smad4 loss identifies aggressive subtype with immunotherapy and anti-HER-2 treatment resistance in gastric cancer. Br J Cancer 2025:10.1038/s41416-025-03002-8. [PMID: 40281303 DOI: 10.1038/s41416-025-03002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/15/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND SMAD4 mutation and homozygous deletion represent the most prevalent genomic events driving aggressive biological behavior in gastric cancer (GC). However, clinical outcome and therapeutic response in GC patients with Smad4-loss remains obscure. METHODS This study included 990 GC patients from four independent clinical centers including the Zhongshan Hospital (ZSHS) cohort, the Cancer Genomic Atlas (TCGA) cohort, the Samsung Medical Center (SMC) cohort and the Memorial Sloan Kettering Cancer Center (MSKCC) cohort. RESULTS In ZSHS cohort, 60/454 GC patients harbored Smad4-loss are characterized by lower pN stage, well histology differentiation, lower EBV infection, null p53 staining and lower tumor proliferation. Smad4-loss GC patients exhibit miserable overall survival across ZSHS cohort and TCGA cohort. Moreover, Smad4-loss GC patients yield no impact on adjuvant chemotherapy, poor outcome upon anti-PD-1 immunotherapy or anti-HER-2 therapy. Interestingly, Smad4-loss GC show more well and intermediate differentiation and lower Ki67 staining. Furthermore, Smad4-loss GC exhibit tumor immunosuppressive contexture characterized with enriched CXCL13+CD8+T cells, reduced IFN-γ+ cells and GZMB+ cells infiltration. CONCLUSIONS Smad4 loss yields poor clinical outcome, immunotherapy and anti-HER-2 treatment resistance and tumor immunosuppressive contexture in GC patients. Our findings provide clues for further detailed biological investigation and aggressive clinical management in Smad4-loss GC patients.
Collapse
Affiliation(s)
- Mengyao Sun
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yun Gu
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Gastrointestinal Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieti Wang
- Department of Endoscopy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ziqiu Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhen Ling
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Fei Shao
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Lin
- Department of Emergency Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongyong He
- Department of Emergency Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ruochen Li
- Department of Emergency Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hao Liu
- Department of Gastrointestinal Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jiejie Xu
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
176
|
Yamada D, Kobayashi S, Doki Y, Eguchi H. Genomic landscape of biliary tract cancer and corresponding targeted treatment strategies. Int J Clin Oncol 2025:10.1007/s10147-025-02761-x. [PMID: 40281353 DOI: 10.1007/s10147-025-02761-x] [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: 02/17/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Biliary tract cancers (BTCs) are classified on the basis of their anatomical origin, and the feasibility of surgical resection depends on the tumor location and extent of progression. However, for unresectable BTCs, systemic therapy has been uniformly applied. Gemcitabine and cisplatin (GC) therapy and GC-based therapies were established as the first-line standard BTC treatment. However, no highly effective second-line therapy has been established, and the prognosis remains poor, highlighting the need for further therapeutic advancements. Meanwhile, the era of precision medicine has expanded the use of genetic testing, leading to the identification of actionable molecular targets in BTC. Several targeted therapies, including FGFR inhibitors and IDH1 inhibitors, have been developed, offering new second-line treatment options and the potential for first-line use in appropriate cases. Notably, the frequency of these genetic alterations varies depending on the tumor location, demonstrating the molecular heterogeneity of BTC. Therefore, it has been recognized that a tailored treatment approach for each BTC patient may be more effective than uniform systemic therapy. Consequently, although routine genetic testing before initiating systemic treatment is currently limited by the medical environment (e.g., cost, accessibility, regional differences), it is recommended in ESMO guideline and might be increasingly advocated. However, BTC harbors a wide range of genetic alterations, and numerous targeted therapies are being developed accordingly. This review provides an overview of the reported genetic alterations in BTC, the frequencies of these alterations, and the corresponding targeted therapies, emphasizing the evolving role of precision medicine in BTC treatment.
Collapse
Affiliation(s)
- Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2E2, Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2E2, Yamadaoka, Suita City, Osaka, 565-0871, Japan.
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2E2, Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2E2, Yamadaoka, Suita City, Osaka, 565-0871, Japan
| |
Collapse
|
177
|
Dutta K, Pal D, Li S, Shyam C, Shoghi KI. Corr-A-Net: Interpretable Attention-Based Correlated Feature Learning framework for predicting of HER2 Score in Breast Cancer from H&E Images. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.22.25326227. [PMID: 40313277 PMCID: PMC12045401 DOI: 10.1101/2025.04.22.25326227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) expression is a critical biomarker for assessing breast cancer (BC) severity and guiding targeted anti-HER2 therapies. The standard method for measuring HER2 expression is manual assessment of IHC slides by pathologists, which is both time intensive and prone to inter- and intra-observer variability. To address these challenges, we developed an interpretable deep-learning pipeline with Correlational Attention Neural Network (Corr-A-Net) to predict HER2 score from H&E images. Each prediction was accompanied with a confidence score generated by the surrogate confidence score estimation network trained using incentivized mechanism. The shared correlated representations generated using the attention mechanism of Corr-A-Net achieved the best predictive accuracy of 0.93 and AUC-ROC of 0.98. Additionally, correlated representations demonstrated the highest mean effective confidence (MEC) score of 0.85 indicating robust confidence level estimation for prediction. The Corr-A-Net can have profound implications in facilitating prediction of HER2 status from H&E images.
Collapse
Affiliation(s)
- Kaushik Dutta
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Debojyoti Pal
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Suya Li
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Chandresh Shyam
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Kooresh I. Shoghi
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
- Department of Biomedical Engineering, Washington University in St Louis, St Louis, MO USA
- Lead contact
| |
Collapse
|
178
|
Kang D, Li J, Li Y, Xu J, Yang J, Zhang Z. Prognostic significance of KRAS, NRAS, BRAF, and PIK3CA mutations in stage II/III colorectal cancer: A retrospective study and meta-analysis. PLoS One 2025; 20:e0320783. [PMID: 40279317 PMCID: PMC12027030 DOI: 10.1371/journal.pone.0320783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/25/2025] [Indexed: 04/27/2025] Open
Abstract
The prognostic significance of KRAS and BRAF mutations is well-established in metastatic colorectal cancer (CRC) but remains uncertain in early-stage tumors. This study retrospectively analyzed 47 stage II/III CRC patients undergoing curative surgery to assess the association of mutations in KRAS, NRAS, BRAF, and PIK3CA with overall survival (OS) and disease-free survival (DFS). Additionally, a meta-analysis was conducted to validate the prognostic relevance of these gene mutations. We included post hoc analyses of phase III randomized controlled trials (RCTs) in stage II/III patients receiving adjuvant therapy after curative resection in the meta-analysis. Pooled hazard ratio (HR) and 95% confidence interval (CI) was calculated using a random-effect model in the overall population, stratified subgroups adjusted for microsatellite instability (MSI) status, and within MSI-high (MSI-H) and microsatellite-stable (MSS) populations. In the retrospective cohort, mutations in KRAS, NRAS, BRAF, and PIK3CA were identified in 29.8%, 4.3%, 8.5%, and 14.9% of patients, respectively. No significant association between individual genes and survival was observed. However, in MSS patients, concurrent mutations were significantly associated with shorter OS and DFS (log-rank test, P < 0.05). The meta-analysis incorporated 13 eligible studies, including 15,034 patients. Pooled analyses revealed that KRAS and BRAF mutations were significantly linked to poor OS (KRAS: HR = 1.25, 95%CI: 1.06-1.47, P = 0.008; BRAF: HR = 1.43, 95%CI: 1.26-1.63, P < 0.001) and DFS (KRAS: HR = 1.36, 95%CI: 1.21-1.53, P < 0.001; BRAF: HR = 1.21, 95%CI: 1.02-1.44, P = 0.032). The prognostic impact of BRAF mutation increased with MSI adjustment compared those without MSI adjustment. In MSS tumors, KRAS-mutant patients demonstrated significantly shorter DFS (HR = 1.63, 95%CI: 1.25-2.13, P < 0.001), while BRAF-mutant patients exhibited reduced OS (HR = 1.53, 95%CI: 1.24-1.89, P < 0.001) and DFS (HR = 1.72, 95%CI: 1.20-2.46, P = 0.003) compared to wildtype patients. Conversely, no significant survival differences were found between mutant and wildtype patients in the MSI-H population. Although PIK3CA mutation was nominally associated with OS (HR = 0.86, 95%CI: 0.75-1.00, P = 0.046), the pooled result lacked robustness. In conclusion, KRAS and BRAF mutations had a negative prognostic impact on MSS stage II/III CRC patients receiving adjuvant therapy following curative resection. These patients may benefit from more effective adjuvant treatment strategies.
Collapse
Affiliation(s)
- Di Kang
- Department of General Surgery, Tianjin Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Jing Li
- Department of General Surgery, Tianjin Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Yangyang Li
- Department of General Surgery, Tianjin Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Jingquan Xu
- Department of General Surgery, Tianjin Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Jianlei Yang
- Department of General Surgery, Tianjin Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Zili Zhang
- Department of General Surgery, Tianjin Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| |
Collapse
|
179
|
Zhu X, Zhang X, Qin Y, Chen Y, Feng X, Deng S, Hu F, Yuan Y, Luo X, Du K, Chang S, Fan X, Ashktorab H, Smoot D, Jin Z, Peng Y. Circular RNA circATM binds PARP1 to suppress Wnt/β-catenin signaling and induce cell cycle arrest in gastric cancer cells. J Adv Res 2025:S2090-1232(25)00277-2. [PMID: 40288674 DOI: 10.1016/j.jare.2025.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/08/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Gastric cancer (GC) is a common malignancy, which is associated with high rates of morbidity and mortality. Despite therapeutic advancements, there is an overall lack of effective treatment options for patients with GC, particularly those with advanced and metastatic disease. The roles of circular (circ)RNAs in tumorigenesis are being increasingly recognized, among which circRNAs are defined as miRNA/protein sponges, scaffolds, or protein coding templates. OBJECTIVES The aim of the present study is to investigate the functions of circATM in GC and elucidate the underlying molecular mechanism. METHODS By circRNA sequencing in GC tissues, we identified a novel 526 nt circRNA, circATM, generating from exons 3-6 of the ATM gene. Through circRNA pull-down and RNA immunoprecipitation assays, we identified PARP1 as one of circATM binding proteins. The EdU, colony formation, wound healing, dual-luciferase reporter, cell cycle assays were employed to evaluated circATM functions in vitro. The GC xenograft model was used to determine the role of circATM in vivo. RESULTS Knocking down circATM promoted GC cells growth in vivo and in vitro. Meanwhile, the overexpression of circATM increased the levels of p16, p21, and p27, and decreased those of β-catenin and c-Myc. Furthermore, we identified PARP1 as a circATM-interacting partner. Mechanistically, circATM bound to the zinc finger motif of Ⅱ-Ⅲ domains of PARP1 to block its recruitment to sites of DNA damage, triggering cell cycle arrest and sequestering β-catenin from the PARP1/β-catenin/TCF4 complex, leading to the suppression of Wnt/β-catenin signaling. Additionally, circATM facilitated the ubiquitin-proteasome degradation of PARP1, further jeopardizing its ability to mediate DNA damage repair. CONCLUSION Taken together, we defined circATM as a novel gastric tumor suppressor via interacting with PARP1, which indicate that circATM may be a promising biomarker for the diagnosis and therapy of GC.
Collapse
Affiliation(s)
- Xiaohui Zhu
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Xiaojing Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Ying Qin
- Department of Gastrointestinal Surgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518000, China
| | - Yang Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Xianling Feng
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Shiqi Deng
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Fan Hu
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Yuan Yuan
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Xiaonuan Luo
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Kaining Du
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Shanshan Chang
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Xinmin Fan
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Hassan Ashktorab
- Department of Medicine and Cancer Center, Howard University, College of Medicine, Washington, DC 20060, USA
| | - Duane Smoot
- Department of Medicine, Meharry Medical Center, Nashville, TN 37208, USA
| | - Zhe Jin
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China.
| | - Yin Peng
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Department of Pathology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
180
|
Jehanno L, Logeart J. [Zolbetuximab in combination with chemotherapy in HER2-negative and CLDN 18.2-positive locally advanced unresectable or metastatic gastric or esogastric junction adenocarcinomas]. Bull Cancer 2025:S0007-4551(25)00126-2. [PMID: 40280797 DOI: 10.1016/j.bulcan.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/04/2025] [Indexed: 04/29/2025]
Affiliation(s)
- Léa Jehanno
- Département d'oncologie médicale, institut Curie, 35, rue Dailly, 92210 Saint-Cloud, France.
| | - Juliette Logeart
- Département d'oncologie médicale, institut Curie, 35, rue Dailly, 92210 Saint-Cloud, France
| |
Collapse
|
181
|
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
|
182
|
Wang LJ, Lei CL, Wang TA, Lin ZF, Feng SJ, Wei T, Li YQ, Shen MR, Li Y, Liao LF. Prognostic value of the preoperative systemic immune-inflammation nutritional index in patients with gastric cancer. World J Clin Oncol 2025; 16:102294. [PMID: 40290682 PMCID: PMC12019271 DOI: 10.5306/wjco.v16.i4.102294] [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: 10/14/2024] [Revised: 12/04/2024] [Accepted: 01/21/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is the fifth most common cancer and the third leading cause of cancer-related deaths in China. Many patients with GC frequently experience symptoms related to the disease, including anorexia, nausea, vomiting, and other discomforts, and often suffer from malnutrition, which in turn negatively affects perioperative safety, prognosis, and the effectiveness of adjuvant therapeutic measures. Consequently, some nutritional indicators such as nutritional risk index (NRI), prognostic nutritional index (PNI), and systemic immune-inflammatory-nutritional index (SIINI) can be used as predictors of the prognosis of GC patients. AIM To examine the prognostic significance of PNI, NRI, and SIINI in postoperative patients with GC. METHODS A retrospective analysis was conducted on the clinical data of patients with GC who underwent surgical treatment at the Guangxi Medical University Cancer Hospital between January 2010 and December 2018. The area under the receiver operating characteristic (ROC) curve was assessed using ROC curve analysis, and the optimal cutoff values for NRI, PNI, and SIINI were identified using the You-Review-HTMLden index. Survival analysis was performed using the Kaplan-Meier method. In addition, univariate and multivariate analyses were conducted using the Cox proportional hazards regression model. RESULTS This study included a total of 803 patients. ROC curves were used to evaluate the prognostic ability of NRI, PNI, and SIINI. The results revealed that SIINI had superior predictive accuracy. Survival analysis indicated that patients with GC in the low SIINI group had a significantly better survival rate than those in the high SIINI group (P < 0.05). Univariate analysis identified NRI [hazard ratio (HR) = 0.68, 95% confidence interval (CI): 0.52-0.89, P = 0.05], PNI (HR = 0.60, 95%CI: 0.46-0.79, P < 0.001), and SIINI (HR = 2.10, 95%CI: 1.64-2.69, P < 0.001) as prognostic risk factors for patients with GC. However, multifactorial analysis indicated that SIINI was an independent risk factor for the prognosis of patients with GC (HR = 1.65, 95%CI: 1.26-2.16, P < 0.001). CONCLUSION Analysis of clinical retrospective data revealed that SIINI is a valuable indicator for predicting the prognosis of patients with GC. Compared with NRI and PNI, SIINI may offer greater application for prognostic assessment.
Collapse
Affiliation(s)
- Li-Jing Wang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Cai-Lu Lei
- School of Pharmaceutical Science, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Ting-An Wang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Feng Lin
- School of Pharmaceutical Science, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Shi-Jie Feng
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Tao Wei
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yan-Qin Li
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Meng-Ru Shen
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yan Li
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Liu-Feng Liao
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
183
|
Luo SQ, Dai L, Zhou YJ, He T, Wang FJ, Jin XR, Wang Q. Multiple primary tumors patient developed microsatellite stable gastric cancer after cadonilimab treatment for liver cancer: A case report. World J Clin Oncol 2025; 16:102418. [PMID: 40290697 PMCID: PMC12019263 DOI: 10.5306/wjco.v16.i4.102418] [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: 10/16/2024] [Revised: 01/22/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Multiple primary malignant tumors refer to the occurrence of two or more primary malignant tumors in the same organ or multiple organs or tissues at the same time or successively in the same patient, and can occur anywhere in the body. The treatment guidelines for patients with multiple primary malignant tumors are currently controversial. CASE SUMMARY A 51-year-old male patient with liver cancer and portal hypertension received 42 months of co-treatment with atezolizumab and bevacizumab. After that, the disease was rated stable disease. The patient was then diagnosed with gastric cancer. Since the patient was not sensitive to anti-programmed death ligand 1 immunosuppressive agents, a co-treatment with oxaliplatin, tegafur, apatinib, and cadonilimab was selected after multidisciplinary consultation and the patient's agreement. After four cycles of treatment, partial response and stable disease were observed in gastric and liver cancers, respectively. Surgical treatment was performed considering the high-risk factors of gastrointestinal bleeding in patients with gastroesophageal varices. Postoperative pathology showed that the Tumor Regression Grade was 1. Moreover, the genetic testing of postoperative tumor specimens indicated negative programmed death ligand 1 and microsatellite stability. In addition, the latest follow-up indicated an 8 and 40-month progression-free survival in gastric and liver cancer patients, respectively. Currently, the patient is receiving postoperative immunotherapy with cadonilimab. CONCLUSION Cadonilimab not only treats microsatellite stability gastric cancer patients but can also be used for liver cancer treatment.
Collapse
Affiliation(s)
- Si-Qi Luo
- Department of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Li Dai
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yong-Jin Zhou
- Department of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Tong He
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Fang-Jie Wang
- Department of Emergency, The People’s Hospital of Xishui, Zunyi 564600, Guizhou Province, China
| | - Xiang-Ren Jin
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Qian Wang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| |
Collapse
|
184
|
Chen X, Jiang Z, Pan J, Xu W, Li Y, Chen X, Pan Y, Weng Y, Hu D, Qiu S. Integrated multi-omics reveal lactate metabolism-related gene signatures and PYGL in predicting HNSCC prognosis and immunotherapy efficacy. BMC Cancer 2025; 25:773. [PMID: 40275154 PMCID: PMC12023518 DOI: 10.1186/s12885-025-13982-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) treatment faces significant clinical challenges. Lactate metabolism plays a crucial role in the initiation of many cancers and the tumor microenvironment (TME). However, the prognostic significance of lactate metabolism-related genes (LMRGs) and the role of TME in HNSCC require further elucidation. METHODS We built a prognostic multigene signature with LMRGs and systematically correlated the risk signature with immunological characteristics and immunotherapy efficacy. Next, a series of single-cell sequencing analyses were used to characterize lactate metabolism in TME. Finally, single-cell sequencing analysis, immunofluorescence analyses, and a series of in vitro experiments were used to explore the role of PYGL in HNSCC. Potential drugs targeting PYGL were screened using AutoDock 4.2. RESULTS A prognostic multigene signature based on LMRGs was developed, which effectively stratified patients into high- and low-risk groups, with significant differences in overall survival (OS) and progression-free survival (PFS). Patients in the low-risk group exhibited reduced lactate metabolism, higher CD8 + T cell infiltration, and improved response to immunotherapy. Single-cell sequencing revealed that tumor cells had the most active lactate metabolism compared to other cells in the TME. PYGL, identified as the most critical prognostic gene, was highly expressed in tumor-associated macrophages and played a role in inhibiting M1 macrophage polarization. Knockdown of PYGL led to reduced lactate levels, and its expression was inversely correlated with CD8 + T cell infiltration. Furthermore, PYGL was involved in copper-dependent cell death, highlighting its potential as a therapeutic target. Drug screening identified elesclomol, which showed promising results in PYGL-knockdown cells. CONCLUSIONS The study established a robust LMRGs-based prognostic model that not only predicts patient survival but also correlates with the immune microenvironment in HNSCC. PYGL emerged as a key biomarker with significant implications for both prognosis and therapeutic intervention. Its role in regulating lactate metabolism and immune suppression suggests that targeting PYGL could enhance the efficacy of immunotherapies. This research provides a foundation for future clinical strategies aimed at improving outcomes in HNSCC by modulating the tumor's metabolic and immune landscapes.
Collapse
Affiliation(s)
- Xiaochuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Zhangying Jiang
- Department of Pathology, Fuzhou Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Junping Pan
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Wenqian Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Ying Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xin Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yuhui Pan
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Youliang Weng
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Dan Hu
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| | - Sufang Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| |
Collapse
|
185
|
Li R, Yang Y, Gao Y, Lv J, Dai C, Zhai Y, Mao C, Jiang J, Fan J, Yu Y, Wu L, Lin Z. Knowledge map of programmed cell death in esophageal cancer: a bibliometric analysis. Discov Oncol 2025; 16:609. [PMID: 40274628 PMCID: PMC12022209 DOI: 10.1007/s12672-025-02376-8] [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: 01/11/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
OBJECTIVES This study aimed to delineate the evolving knowledge structure of programmed cell death in esophageal cancer and identify key thematic trends, influential collaborations, and emerging areas for future research. METHODS A bibliometric approach was applied to 2677 publications retrieved from the Web of Science Core Collection (2000-2024). Three complementary tools-CiteSpace, VOSviewer, and bibliometrix-were employed to visualize co-citation networks, detect citation bursts, and map collaborative patterns among authors, institutions, and countries. Inclusion criteria focused on articles and reviews that addressed esophageal cancer in conjunction with apoptosis, necroptosis, pyroptosis, ferroptosis, autophagy, or related pathways. RESULTS Publication outputs grew markedly, reflecting a shift from early investigations of basic apoptotic mechanisms to broader explorations of necroptosis, pyroptosis, and ferroptosis. China led in publication volume and citations, driven by substantial governmental funding and large clinical cohorts. The United States and Japan also contributed significantly, forming international research networks that spanned Asia and Europe. Leading institutions, particularly Zhengzhou University, demonstrated extensive collaborations. Journals such as Oncology Letters and Oncology Reports were prominent outlets for new findings, while highly cited references highlighted hypoxia, immune checkpoint blockade, and emerging gene-editing strategies. Keyword analyses revealed the ascendance of immuno-oncology, network pharmacology, and translational applications targeting multiple regulated cell death pathways. CONCLUSION Bibliometric evidence underscores a rapid expansion of multidisciplinary research that integrates diverse cell death pathways in esophageal cancer. Continued international collaborations, leveraging advanced genomics and immunologic strategies, are poised to accelerate translational breakthroughs and enable more personalized, effective therapies.
Collapse
Affiliation(s)
- Rulin Li
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yanchun Yang
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yang Gao
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Jing Lv
- Department of Orthopedics, Ziyang Central Hospital, Ziyang, 641300, China
| | - Chuanqiang Dai
- Department of Orthopedics, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yuanwei Zhai
- Department of Medical Imaging, Ziyang Central Hospital, Ziyang, 641300, China
| | - Chirong Mao
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Jiudong Jiang
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Jiangang Fan
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yang Yu
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Liang Wu
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Zhiwu Lin
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China.
| |
Collapse
|
186
|
Gao LL, Gao DN, Yuan HT, Chen WQ, Yang J, Peng JQ. Combining anti-PD-1 antibodies with surufatinib for gastrointestinal neuroendocrine carcinoma: Two cases report and review of literature. World J Clin Oncol 2025; 16:102297. [PMID: 40290678 PMCID: PMC12019270 DOI: 10.5306/wjco.v16.i4.102297] [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: 10/14/2024] [Revised: 12/20/2024] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Gastrointestinal neuroendocrine carcinoma (GI NEC) has a low incidence rate and poor prognosis. Most patients already have metastatic disease when they are diagnosed. Platinum chemotherapy is the main means of treating metastatic GI NECs. There is a lack of effective treatment methods after chemotherapy failure. Therefore, Therefore, selecting appropriate posterior-line treatment programs to improve the prognosis of patients is urgently needed. CASE SUMMARY A 64-year-old female was diagnosed with stage IV NEC of the rectum due to abdominal pain and rectal bleeding. After multiline chemotherapy, the condition progressed, and the patient was treated with a combination of camrelizumab and surufatinib. The efficacy evaluation revealed partial remission (PR) and stable conditions, with the expression of the tumor marker neuron-specific enolase (NSE) returning to normal. The adverse reactions were controllable, and the overall condition was good, with weight gain achieved in the past four years. Another 51-year-old female experienced recurrence and metastasis of a duodenal NEC after surgery. After multiline chemotherapy, she received sintilimab combined with surufatinib. The curative effect fluctuated between PR and stability. During treatment, she recovered from immune-related diabetes and later died due to deterioration of her condition. During the treatment, the patient's NSE level returned to normal. CONCLUSION The combination of antiangiogenic targeted drugs and immunotherapy provides a new therapeutic approach for the treatment of metastatic GI-NECs.
Collapse
Affiliation(s)
- Lou-Lu Gao
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Dong-Ni Gao
- Department of Oncology, Shandong Public Health Clinical Center, Jinan 250100, Shandong Province, China
| | - Hong-Tu Yuan
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, China
| | - Wen-Qiang Chen
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Jing Yang
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, China
| | - Jie-Qiong Peng
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, China
| |
Collapse
|
187
|
Martins F, Machado AL, Carvalho J, Almeida CR, Beck HC, Carvalho AS, Backman V, Matthiesen R, Velho S. Differential unfolded protein response regulation in KRAS silencing sensitive and innately resistant colorectal cancer cells. Sci Rep 2025; 15:14329. [PMID: 40274922 PMCID: PMC12022182 DOI: 10.1038/s41598-025-94549-2] [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: 12/19/2024] [Accepted: 03/14/2025] [Indexed: 04/26/2025] Open
Abstract
Despite the development of mutant-selective KRAS inhibitors, colorectal cancer (CRC) responses remain limited, with stable disease and rapid recurrence being common outcomes. The molecular mechanisms enabling CRC cells to tolerate KRAS inhibition and ultimately develop resistance remain poorly understood. Here, we investigated early transcriptional and proteomic responses to KRAS silencing in 3D CRC cell line spheroid models, aiming to identify pathways associated with sensitivity or resistance to KRAS blockade. Cell lines were stratified into KRAS silencing-sensitive (HCT116 and SW480) and -resistant (LS174T and SW837) groups based on spheroid growth, cell cycle progression, and apoptosis induction. Transcriptional profiling revealed the unfolded protein response (UPR) and WNT/β-catenin signaling as pathways specifically upregulated in KRAS silencing-sensitive cells and downregulated in resistant cells. Proteomic analysis of membrane-enriched fractions further supported UPR deregulation, showing a pronounced downregulation of translation-related proteins in sensitive cells. Functional assays validated that the sensitive cell line HCT116 exhibits reduced protein aggregation and lower translational capacity upon KRAS knockdown, consistent with UPR activation. Pharmacological inhibition of IRE1α-mediated UPR signaling did not revert KRAS silencing-induced cell cycle arrest or apoptosis in this cell line. Collectively, our results highlight the UPR activation as an early adaptive response of KRAS-dependent CRC cells to KRAS silencing.
Collapse
Affiliation(s)
- Flávia Martins
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ana L Machado
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Joana Carvalho
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Catarina R Almeida
- Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
- Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Hans C Beck
- Centre for Clinical Proteomics, Department of Clinical Biochemistry, Odense University Hospital, Odense C, 5000, Denmark
| | - Ana S Carvalho
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Vadim Backman
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Rune Matthiesen
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Sérgia Velho
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.
- i3S-Institute for Research and Innovation in Health, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
| |
Collapse
|
188
|
Tougeron D, Louvet C, Desramé J, Evesque L, Angelergues A, Carnot A, Breysacher G, Zaanan A, Etchepare N, Mabro M, Kaluzinski L, Petorin C, Chibaudel B, Aparicio T, Bodere A, Rinaldi Y, Le Malicot K, Emile JF, Lepage C, Baures A, Djamai H, Taly V, Laurent-Puig P. Circulating tumor DNA strongly predicts efficacy of chemotherapy plus immune checkpoint inhibitors in patients with advanced gastro-esophageal adenocarcinoma. COMMUNICATIONS MEDICINE 2025; 5:136. [PMID: 40275077 PMCID: PMC12022060 DOI: 10.1038/s43856-025-00867-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: 07/22/2024] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Efficacy of 2nd line treatment in advanced gastric or gastro-esophageal junction (GEJ) adenocarcinoma remains limited with no identified strong predictor of treatment efficacy. We evaluated the prognostic value of circulating tumor DNA (ctDNA) in predicting the efficacy of immune checkpoint inhibitors (ICI) plus chemotherapy in the randomized PRODIGE 59-FFCD 1707-DURIGAST trial. METHODS ctDNA was evaluated before treatment (baseline) and at 4 weeks (before the third cycle of treatment, C3) using droplet-digital PCR assays based on the detection of CpG methylation. RESULTS Progression-free survival (PFS) and overall survival (OS) were shorter in patients with a high (>1.1 ng/mL) versus low (<1.1 ng/mL) ctDNA concentration at baseline (2.3 vs. 5.8 months; HR = 2.19; 95% CI, 1.09-4.41; p = 0.03 and 4.5 vs. 12.9 months; HR = 2.73; 95% CI, 1.29-5.75; p < 0.01), respectively, after adjustment for identified prognostic variables. Patients with a ctDNA decrease ≤75% between baseline and C3 versus a ctDNA decrease >75% had a worse objective response rate (p = 0.007), shorter PFS (2.2 vs. 7.4 months, HR = 1.90; 95% CI, 1.03-3.51; p = 0.04) and OS (6.6 vs 16.0 months; HR = 2.18; 95% CI, 1.09-4.37; p = 0.03). CONCLUSIONS An early decrease in ctDNA concentration is a strong predictor of the therapeutic efficacy of ICI plus chemotherapy in advanced gastric/GEJ adenocarcinoma. Clinical Trial Information NCT03959293 (DURIGAST).
Collapse
Affiliation(s)
- David Tougeron
- Department of Gastroenterology and Hepatology, Poitiers University Hospital, Poitiers, France.
| | - Christophe Louvet
- Department of Medical Oncology, Institute Mutualiste Montsouris, Paris, France
| | - Jérôme Desramé
- Department of Gastroenterology, Mermoz Hospital, Lyon, France
| | - Ludovic Evesque
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | | | - Aurélien Carnot
- Department of Gastroenterology and Digestive Oncology, Oscar Lambret Centre, Lille, France
| | - Gilles Breysacher
- Department of Gastroenterology and Hepatology, Colmar Hospital, Colmar, France
| | - Aziz Zaanan
- Department of Digestive Oncology, Georges Pompidou European Hospital, AP-HP, Université Paris Cité, Paris Cancer Institute CARPEM, Paris, France
| | | | - May Mabro
- Department of Oncology, Foch Hospital, Suresnes, France
| | - Laure Kaluzinski
- Department of Oncology, Cherbourg-en-Cotentin Hospital, Cherbourg-en-Cotentin, France
| | - Caroline Petorin
- Department of Oncology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Benoist Chibaudel
- Department of Oncology, Franco-Britannique Hospital - Fondation Cognacq-Jay, Levallois, France
| | - Thomas Aparicio
- Department of Gastroenterology and Digestive Oncology, Saint Louis Hospital, Paris, France
| | | | - Yves Rinaldi
- Department of Gastroenterology, Marseille European Hospital, Marseille, France
| | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, Bourgogne Franche-Comté University, Dijon, France
| | - Jean-François Emile
- Pathology Department, Paris-Saclay University, Versailles SQY University, EA4340-BECCOH, Assistance Publique-Hôpitaux de Paris (APHP), Ambroise-Paré Hospital, Boulogne, France
| | - Côme Lepage
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, Bourgogne Franche-Comté University, Dijon, France
| | - Aurélia Baures
- Centre de recherche des cordeliers, Université Paris Cité, Sorbonne Université, UMR-S1138, CNRS SNC5096, Équipe Labélisée Ligue Nationale Contre le Cancer, Paris, France
| | - Hanane Djamai
- Centre de recherche des cordeliers, Université Paris Cité, Sorbonne Université, UMR-S1138, CNRS SNC5096, Équipe Labélisée Ligue Nationale Contre le Cancer, Paris, France
| | - Valérie Taly
- Centre de recherche des cordeliers, Université Paris Cité, Sorbonne Université, UMR-S1138, CNRS SNC5096, Équipe Labélisée Ligue Nationale Contre le Cancer, Paris, France
- METHYS Dx, Paris, France
| | - Pierre Laurent-Puig
- Centre de recherche des cordeliers, Université Paris Cité, Sorbonne Université, UMR-S1138, CNRS SNC5096, Équipe Labélisée Ligue Nationale Contre le Cancer, Paris, France
- Department of Genomic Medicine of Tumors and Cancers APHP, Institut Cancer Paris Carpem, APHP, Paris, France
| |
Collapse
|
189
|
Perez-Silva L, Herraez E, Marijuan RP, Reviejo M, Lozano E, Bujanda L, Abad M, Macias RIR, Briz O, Marin JJG. Role of tumor suppressor genes P53 and PTEN in CD44-mediated gastric adenocarcinoma multidrug resistance. Biomed Pharmacother 2025; 187:118057. [PMID: 40280031 DOI: 10.1016/j.biopha.2025.118057] [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: 01/13/2025] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Gastric adenocarcinoma (GAC) is often diagnosed at advanced stages, when curative options are limited and marked chemoresistance is already present. Although tumor suppressor genes (TSGs) are frequently altered in GAC, their impact on chemoresistance is not well understood. Gene expression data from The Cancer Genome Atlas cohort TCGA-STAD were validated by RT-qPCR in a Spanish cohort of GAC. In the human GAC cell line AGS, gene knocking-out was performed using CRISPR/Cas9. Cell viability (MTT-formazan test) and proliferation rate (digital holographic microscopy) were determined. Among the most frequently inactivated TSGs, TP53, PTEN, and ARID1A were selected for further studies. In GAC samples, TP53 was upregulated, whereas PTEN and ARID1A were downregulated. Mutations in these TSGs led to a consistent alteration in the expression of their target genes. AGS cells exhibited TSG expression profiles like those observed in GAC, which supports their suitability as an in vitro model. Knocking-out ARID1A (ARID1AKO) enhanced cell chemosensitivity. In contrast, silencing TP53 (p53KO) or PTEN (PTENKO) led to increased resistance to platinum-based drugs, doxorubicin, epirubicin, and docetaxel. Characterization of the resistome was performed using TaqMan Low-Density Arrays. In p53KO and PTENKO cells, the expression of UGT1A and CD44 was altered. Additional silencing of CD44 in these cells partially reversed their chemoresistance. Moreover, pharmacological inhibition of CD44 with verbascoside sensitized p53KO and PTENKO cells to anticancer drugs. In conclusion, dysfunctional TP53 and PTEN contribute to altered drug responses of GAC. Moreover, we identified pharmacological vulnerabilities that could be useful to chemosensitize these tumors.
Collapse
Affiliation(s)
- Laura Perez-Silva
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Elisa Herraez
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Rebeca P Marijuan
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Maria Reviejo
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Elisa Lozano
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Luis Bujanda
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), San Sebastian, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Mar Abad
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Department of Pathology, University Hospital of Salamanca, University of Salamanca, Salamanca, Spain
| | - Rocio I R Macias
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Oscar Briz
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Jose J G Marin
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health, Madrid, Spain.
| |
Collapse
|
190
|
Zaanan A, Bouché O, de la Fouchardière C, Le Malicot K, Pernot S, Louvet C, Artru P, Le Brun Ly V, Aldabbagh K, Khemissa-Akouz F, Lecomte T, Castanie H, Laly M, Botsen D, Roth G, Samalin E, Muller M, Breysacher G, Manfredi S, Phelip JM, Taieb J. TFOX versus FOLFOX in first-line treatment of patients with advanced HER2-negative gastric or gastro-oesophageal junction adenocarcinoma (PRODIGE 51- FFCD-GASTFOX): an open-label, multicentre, randomised, phase 3 trial. Lancet Oncol 2025:S1470-2045(25)00130-5. [PMID: 40286809 DOI: 10.1016/s1470-2045(25)00130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Perioperative FLOT (fluorouracil, oxaliplatin, and docetaxel) triplet chemotherapy is the standard of care for localised and resectable gastric and gastro-oesophageal junction adenocarcinoma. We aimed to compare a modified FLOT regimen (also known as TFOX) with FOLFOX as first-line treatment for patients with HER2-negative advanced gastric and gastro-oesophageal junction adenocarcinoma. METHODS PRODIGE 51-FFCD-GASTFOX is an open-label, multicentre, randomised, phase 3 trial conducted at 96 medical centres in France. Eligible individuals were aged 18 years or older, had histologically confirmed, HER2-negative adenocarcinoma of the stomach or gastro-oesophageal junction that was locally advanced unresectable or metastatic and previously untreated, measurable disease per Response Evaluation Criteria in Solid Tumours, an Eastern Cooperative Oncology Group performance status of 0 or 1, and adequate organ function. Patients were randomly assigned (1:1), using the minimisation method, to receive FOLFOX (folinic acid 400 mg/m2, oxaliplatin 85 mg/m2, and 5-fluorouracil bolus 400 mg/m2 then 5-fluorouracil 2400 mg/m2 as a continuous 46 h infusion every 2 weeks) or TFOX (docetaxel 50 mg/m2, folinic acid 400 mg/m2, and oxaliplatin 85 mg/m2 then 5-fluorouracil 2400 mg/m2 as a continuous 46 h infusion every 2 weeks). Randomisation was stratified by centre, ECOG performance status, (neo)adjuvant chemotherapy or chemoradiotherapy, tumour stage, tumour location, and pathological histological subtype. The primary endpoint was progression-free survival (assessed in the intention-to-treat population), defined as time from randomisation to the first radiological or clinical progression (or both), or death due to any cause, whichever occurred first. Secondary endpoints included overall survival (defined as time from randomisation to death due to any cause) and objective response rate (defined as the proportion of patients with a best overall complete or partial response). Hazard ratio and 95% CIs were estimated using an unstratified Cox proportional hazards model. When the proportional hazards assumption was violated, the restricted mean survival time was used to estimate the treatment effect size. This study is registered with ClinicalTrials.gov, NCT03006432, and EudraCT, 2016-002331-16. FINDINGS Between Dec 19, 2016, and Dec 26, 2022, 507 patients were randomly assigned (254 to the TFOX group and 253 to the FOLFOX group [intention-to-treat population]). The median age was 64·2 years (IQR 56·7-70·8), and 399 (79%) participants were male and 108 (21%) were female. At median follow-up of 42·8 months (25·8-49·9), the median progression-free survival was 7·59 months (95% CI 7·06-7·95) in the TFOX group versus 5·98 months (5·65-6·97) in the FOLFOX group. The assumption of proportional hazards was violated (p=0·013); therefore, the 12-month restricted mean progression-free survival was calculated: 7·52 months (7·06-7·97) in the TFOX group versus 6·62 months (6·16-7·09) in the FOLFOX group (p=0·0072). The median overall survival was 15·08 months (13·70-16·72) in the TFOX group versus 12·65 months (10·94-14·00) in the FOLFOX group (proportional hazards assumption was confirmed; HR 0·82 [0·68-0·99]; p=0·048) and the objective response rate was 62·3% (56·0-68·3) versus 53·4% (47·0-59·8; p=0·045). The most common grade 3 and 4 treatment-emergent adverse events were diarrhoea (37 [15%] in the TFOX group vs 18 [7%] in the FOLFOX group), peripheral neuropathy (80 [32%] vs 49 [20%]), neutropenia (67 [27%] vs 44 [18%]), and fatigue (40 [16%] vs 20 [8%]). Serious treatment-related adverse events occurred in 66 (27%) participants in the TFOX group and 33 (13%) in the FOLFOX group. There were two (<1%) treatment-related deaths in the TFOX group (one due to septic shock and one due to gastrointestinal perforation) and one (<1%) in the FOLFOX group (due to septic shock). INTERPRETATION The modified FLOT/TFOX regimen significantly improved progression-free survival, overall survival, and objective response rate compared with FOLFOX in previously untreated patients with advanced HER2-negative gastric and gastro-oesophageal junction adenocarcinoma. The modified FLOT/TFOX regimen might represent a new first-line treatment option for patients eligible for this docetaxel triplet chemotherapy. FUNDING Fédération Francophone de Cancérologie Digestive.
Collapse
Affiliation(s)
- Aziz Zaanan
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, University of Paris Cité, Paris, France.
| | - Olivier Bouché
- Department of Digestive Oncology, CHU Reims, Université Reims Champagne Ardenne, Reims, France
| | | | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, Faculté de Médecine, University of Burgundy and Franche Comté, Dijon, France
| | - Simon Pernot
- Medical Oncology Department, Institut Bergonié, Bordeaux, France
| | - Christophe Louvet
- Medical Oncology Department, Institut Mutualiste Montsouris, Paris, France
| | - Pascal Artru
- Medical Oncology Department, Hôpital privé Jean Mermoz, Lyon, France
| | | | - Kais Aldabbagh
- Medical Oncology Department, Polyclinique Saint Côme, Compiègne, France
| | - Faiza Khemissa-Akouz
- Department of Gastroenterology and Digestive Oncology, Centre Hospitalier de Perpignan, Perpignan, France
| | - Thierry Lecomte
- Department of Hepatogastroenterology and Digestive Oncology, Tours University Hospital, Tours, France
| | - Hélène Castanie
- Medical Oncology Department, Hôpital Privé du Confluent, Nantes, France
| | - Margot Laly
- Department of Gastroenterology and Digestive Oncology, Centre Hospitalier Départemental Vendée, La Roche-sur-Yon, France
| | - Damien Botsen
- Medical Oncology Department, Institut Godinot, Reims, France
| | - Gael Roth
- University Grenoble Alpes-Department of Hepato-Gastroenterology and Digestive Oncology, CHU Grenoble Alpes-Institute for Advanced Biosciences, CNRS UMR 5309-INSERM U1209, Grenoble, France
| | - Emmanuelle Samalin
- Department of Medical Oncology, Institut du cancer de Montpellier, Université de Montpellier, Montpellier, France
| | - Marie Muller
- Department of Hepatogastroenterology and Digestive Oncology, Nancy University Hospital, University of Lorraine, Vandoeuvre-lès-Nancy, France
| | - Gilles Breysacher
- Department of Gastroenterology and Digestive Oncology, Hôpitaux Civils de Colmar, Colmar, France
| | - Sylvain Manfredi
- Department of Hepatogastroenterology and Digestive Oncology, Centre Hospitalier Universitaire de Dijon, Dijon, France
| | - Jean-Marc Phelip
- Department of Hepatogastroenterology and Digestive Oncology, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, University of Paris Cité, Paris, France
| |
Collapse
|
191
|
Xu P, Hong C, Liu L, Xiao L. PD-1/PD-L1 blockade therapy in hepatocellular carcinoma: Current status and potential biomarkers. Biochim Biophys Acta Rev Cancer 2025; 1880:189334. [PMID: 40280499 DOI: 10.1016/j.bbcan.2025.189334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related death and the sixth most prevalent cancer worldwide. However, most patients with HCC are at an advanced stage at the time of clinical diagnosis, making surgery impossible. In the past, targeted therapeutic drugs such as sorafenib and lenvatinib were the main treatments. With recent breakthroughs in medicine, immunotherapy, particularly immune checkpoint inhibitors (ICIs), has garnered interest and has been extensively studied for clinical treatment. In addition to single-agent therapies, combination regimens involving ICIs have also been developed. Despite this progress, not all patients with HCC benefit from immunotherapy. Therefore, to improve the treatment response rates, it is crucial to identify patients with HCC who are suitable for immunotherapy. The exploration and validation of markers to predict the outcomes of immunotherapeutic treatments in patients with HCC are of clinical importance. In this article, we provide a comprehensive review of research progress in immunotherapy, particularly ICIs and combination therapies, for HCC. Furthermore, we summarize the clinical indicators and tumor markers discovered in recent years to forecast immunotherapy outcomes in patients with HCC. We also outline predictive markers for the occurrence of immune-related adverse events in patients with HCC receiving immunotherapy and discuss future research directions in the immunotherapeutic treatment landscape.
Collapse
Affiliation(s)
- Peishuang Xu
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chang Hong
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Li Liu
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Lushan Xiao
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
192
|
Guan J, Gong X, Zeng H, Zhang W, Qin Q, Gou H, Liu X, Song B. Gastrointestinal tumor personalized immunotherapy: an integrated analysis from molecular genetics to imaging biomarkers. Therap Adv Gastroenterol 2025; 18:17562848251333527. [PMID: 40297204 PMCID: PMC12035075 DOI: 10.1177/17562848251333527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The immunotherapy landscape for gastrointestinal (GI) tumors is rapidly evolving. There is an urgent need for reliable biomarkers capable of predicting treatment outcomes to optimize therapeutic strategies and enhance patient prognosis. This review presents a comprehensive overview of biomarkers associated with the immunotherapy response of GI tumors, covering advances in molecular genetics, histopathological markers, and imaging. Key molecular biomarkers, such as microsatellite instability, tumor mutational burden, and programmed death-ligand 1 expression, remain critical for identifying patients likely to benefit from immune checkpoint inhibitors. The significance of tumor-infiltrating lymphocytes, notably the CD8+ T cell to regulatory T cell ratio, as a predictor of immunotherapy response is explored. In addition, advanced imaging techniques, including computed tomography (CT), magnetic resonance imaging, and positron emission tomography-CT, facilitate the noninvasive evaluation of tumor biology and therapeutic response. By bridging molecular and imaging data, this integrated strategy enhances precision in patient selection, treatment monitoring, and adaptive therapy design. Future studies should aim to validate these biomarkers in larger, multicenter cohorts and focus on clinical translation to advance precision medicine in GI oncology.
Collapse
Affiliation(s)
- Jian Guan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Xiaoling Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hanjiang Zeng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Qing Qin
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijiao Liu
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| |
Collapse
|
193
|
Fukunaga H, Fukunaga M. Mitochondrial DNA copy numbers in gastric cancer tissues: a possible biomarker for estimating cancer progression. Jpn J Clin Oncol 2025:hyaf066. [PMID: 40263745 DOI: 10.1093/jjco/hyaf066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Mitochondria have their own genome (mtDNA), which in humans is a circular multi-copy genome consisting of 16 569 base pairs. Abnormalities in the mtDNA have been reported to correlate with various age-related pathophysiologies. METHODS Based on a total of 182 DNA samples extracted from gastric cancer tissues, we measured mtDNA copy numbers (mtDNA-CN) using real-time polymerase chain reaction (PCR) and then examined alongside sex, age, tumor stage, Laurén classification, and the overexpression of Human Epidermal Growth Factor Receptor 2 (HER2). RESULTS We found no sex differences in mtDNA-CN and no correlation with age, but significant differences according to tumor stage. The mtDNAcn of intestinal type by Laurén classification was significantly larger than that of diffuse type. There was no significant difference in mtDNA-CN between HER2-positive and -negative tissues. Multiple regression analyses showed that only the tumor stage was a significant variable, while Laurén classification was not. CONCLUSION These results indicate that mitochondrial genomic abnormalities contribute the progression of gastric cancer independently of HER2 overexpression, and may shed light on the emerging role of mtDNA-CN in situ as a possible biomarker for estimating cancer progression.
Collapse
Affiliation(s)
- Hisanori Fukunaga
- Department of Biomedical Science and Engineering, Faculty of Health Sciences, Hokkaido University, N12 W5 Kita-ku, Sapporo 060-0812, Japan
| | - Mayuko Fukunaga
- Department of Internal Medicine, Sapporo Daiichi Hospital, Niju-Yonken 4-jo 3-chome 4-26, Nishi-ku, Sapporo 063-0804, Japan
- Department of Gastroenterology and Hepatology, School of Medicine, Sapporo Medical University, S1 W16 Chuo-ku, Sapporo 060-8543, Japan
| |
Collapse
|
194
|
Tian L, Guo Q, Fu D, Ma X, Wang L. Adjuvant chemotherapy compared with observation in patients with resected biliary tract cancer: A systematic review and meta-analysis of randomized controlled trials. PLoS One 2025; 20:e0295583. [PMID: 40267153 PMCID: PMC12017477 DOI: 10.1371/journal.pone.0295583] [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: 09/20/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVES Several randomized controlled trials compared adjuvant systemic chemotherapy with observation in patients with resected biliary tract cancer (BTC) have yielded inconsistent outcomes. In order to assess the efficacy of adjuvant therapy in these patients, we conducted this systematic review and meta-analysis. METHODS We conducted a thorough search in various databases, which included MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, ASCO Abstracts, ESMO Abstracts and ClinicalTrials.gov. All relevant randomized controlled trials investigating the adjuvant chemotherapy compared with observation in resected biliary tract cancer were identified. The primary outcome of interest was overall survival (OS), while secondary outcome was relapse-free survival (RFS). Statistical analyses were conducted using Review Manager 5.3. Additionally, publication bias was evaluated using Egger's test in Stata 12.0. RESULTS A total of 5 randomized controlled trials, involving 1406 patients, were included in this analysis. Compared with observation, adjuvant chemotherapy improved RFS [HR 0.84 (0.73-0.96), p=0.01] (I2=0%, p=0.89) but not OS [HR 0.89 (0.77-1.03), p=0.12] (I2=51%, p=0.09) in the entire population after BTC resection. Subgroup analyses revealed that adjuvant chemotherapy did improve both OS [HR 0.76 (0.62-0.93), p=0.009] (I2=7%, p=0.37) and RFS [HR 0.74 (0.58-0.95), p=0.02] (I2=0%, p=0.39) in patients with lymph node positivity. Furthermore, patients receiving oral fluoropyrimidine monotherapy showed benefit from the adjuvant therapy, with longer OS [HR 0.78 (0.65-0.94), p=0.009] (I2=2%, p=0.31) and RFS [HR 0.81 (0.68-0.95), p=0.01] (I2=0%, p=0.95). CONCLUSIONS To conclude, adjuvant chemotherapy have the potential to offer advantages in patients with resected BTC. Specifically, patients demonstrating positive lymph node status have a higher likelihood of benefiting from adjuvant therapy. Our analysis supports the current standard of care of adjuvant fluoropyrimidine. However, the recommendation of oral fluoropyrimidine monotherapy as the preferred option is not definitive, as it is based on limited studies. Further validation of these outcomes is necessary by conducting extensive randomized controlled trials.
Collapse
Affiliation(s)
- Liying Tian
- Department of Medical, Jinan High-tech East District Hospital, Shandong Healthcare Industry Development Group Co., Ltd, Jinan, Shandong, Peoples’ Republic of China
| | - Qian Guo
- Day Care Unit, Zibo Central Hospital, Shandong University, Zibo, Shandong, Peoples’ Republic of China
| | - Daidi Fu
- Department of Oncology, Zibo Central Hospital, Shandong University, Zibo, Shandong, Peoples’ Republic of China
| | - Xiao Ma
- Department of Internal Medicine, Zhangqiu People’s Hospital, Zhangqiu, Shandong, People’ Republic of China
| | - Linjun Wang
- Department of Medical, Jinan High-tech East District Hospital, Shandong Healthcare Industry Development Group Co., Ltd, Jinan, Shandong, Peoples’ Republic of China
| |
Collapse
|
195
|
Li C, Wang Y, Fang B, Liu M, Sun S, Qu J, Zhang S, Du C. Options for postoperative radiation therapy in patients with de novo metastatic breast cancer. Breast 2025; 82:104483. [PMID: 40286762 DOI: 10.1016/j.breast.2025.104483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Although meta-analyses have demonstrated survival benefits associated with primary tumor resection in MBC, guidelines lack consensus on the survival benefit of postoperative radiation therapy (RT). METHODS In this study, we included 1392 patients with de novo metastatic breast cancer (dnMBC) by integrating data from the SEER database (2010-2019) to systematically assess the efficacy of postoperative RT and develop a machine learning-driven prognostic tool. The primary endpoint was overall survival (OS). RESULTS Propensity score matching (PSM) results showed that postoperative RT significantly improved OS (HR = 0.573, 95 % CI = 0.475-0.693), but this survival gain showed great heterogeneity among different subgroups. It is found that patients with HR-/HER2-or HR+/HER2-subtypes gained significant OS benefit from (p < 0.001) postoperative RT, whereas patients with HER2+ subtype did not gain any survival benefit since the effect of targeted therapy overshadowed the postoperative RT. Further risk stratification by the random survival forest (RSF) model revealed that high-risk patients with T4/N3 stage, high tumor grade and poor response to chemotherapy had significantly prolonged OS after receiving RT (p < 0.001), while low-risk patients showed no additional benefit. The model had excellent predictive efficacy (training set C-index = 0.741, validation set C-index = 0.720) with key predictors including HER2 status, chemotherapy response and tumor grade. The research team developed an interactive web application (https://lee2287171854.shinyapps.io/RSFshiny/) based on this model, which can generate individualized survival risk scores in real-time to guide clinical decision-making. CONCLUSION This study is the first to propose a risk stratification strategy for postoperative RT in dnMBC, and innovatively integrates machine learning and clinical tools to provide a new paradigm for optimizing precision therapy.
Collapse
Affiliation(s)
- Chaofan Li
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an, Shaanxi, PR China
| | - Yusheng Wang
- Department of Otolaryngology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an, Shaanxi, PR China
| | - Biyun Fang
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an, Shaanxi, PR China
| | - Mengjie Liu
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an, Shaanxi, PR China
| | - Shiyu Sun
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an, Shaanxi, PR China
| | - Jingkun Qu
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an, Shaanxi, PR China
| | - Shuqun Zhang
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an, Shaanxi, PR China.
| | - Chong Du
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an, Shaanxi, PR China.
| |
Collapse
|
196
|
El Zaitouni S, Laraqui A, Boustany Y, Benmokhtar S, El Annaz H, Abi R, Tagajdid MR, El Kochri S, Bouaiti EA, Amine IL, Ameziane El Hassani R, Ennibi K. Potency and Safety of KRAS G12C Inhibitors in Solid Tumors: A Systematic Review. Clin Med Insights Oncol 2025; 19:11795549251331759. [PMID: 40297021 PMCID: PMC12035108 DOI: 10.1177/11795549251331759] [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: 04/19/2024] [Accepted: 03/13/2025] [Indexed: 04/30/2025] Open
Abstract
Background The Kirsten rat sarcoma viral oncogene homolog (KRAS) gene, specifically the cysteine residue mutation KRAS (G12C), has garnered significant attention as a therapeutic target for solid cancer patients with KRAS mutations. Despite this interest, the efficacy and safety profiles of KRAS G12C inhibitors remain incompletely understood. In this study, we comprehensively evaluate the effectiveness and toxicity of relevant KRAS G12C inhibitors (Sotorasib, Adagrasib, Garsorasib, and Divarasib) in patients with colorectal cancer (CRC), non-small-cell lung cancer (NSCLC), and pancreatic ductal adenocarcinomas (PDAC). Methods Our systematic review is guided by Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We review the available clinical trials data on KRAS G12C inhibitors in KRAS G12C-mutated solid tumors. We searched PubMed, EMBASE, Cochrane Library, and major international conferences for clinical trials from January 2020 until August 2023. Results A total of 17 eligible studies were included. KRAS G12C inhibitions with Sotorasib (41.2%) and Adagrasib (41.2%) each of them were reported in 7 studies. Divarasib was reported in 2 studies (11.8%) and Garsorasib was reported in 1 study (6.7%). Sotorasib showed a significant clinical benefit in terms of objective response rate (ORR) (7.1%-47%), progression-free survival (PFS) (4-6.8 months), and overall survival (OS) (4-24 months); it is more efficient in NSCLC patients with an OS of 2 years, PFS of 6.3 months, and an ORR of 41%. Adagrasib also showed significant clinical activity with an ORR (19%-53%), PFS (3.3-11.1 months), and OS (10.5-23.4 months), with more effectiveness in NSCLC patients with an OS of 23.4 months, PFS of 11.1 months, and an ORR of 53.3%. Adagrasib is more efficient with an ORR of 35.1%, PFS of 7.4 months, and an OS of 14 months in patients with PDAC, than Sotorasib which showed an ORR of 21%, PFS of 4 months, and an OS of 6.9 months. However, Adagrasib and Sotorasib are moderately efficient in CRC clinical trials. Conclusion This study confirms that patients treated with these KRAS G12C inhibitors, exclusively or combined with conventional therapies, achieve better treatment responses and modulate the progressions of these solid tumors.
Collapse
Affiliation(s)
- Sara El Zaitouni
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Abdelilah Laraqui
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - Youssra Boustany
- Microbiology and Molecular Biology Team, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Soukaina Benmokhtar
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Hicham El Annaz
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - Rachid Abi
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - Mohamed Rida Tagajdid
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - Safae El Kochri
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - El Arbi Bouaiti
- Laboratory of Biostatistics, Clinical Research and Epidemiology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Idriss Lahlou Amine
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - Rabii Ameziane El Hassani
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Khalid Ennibi
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| |
Collapse
|
197
|
Akkus E, Öksüz NE, Erul E. KRAS G12C inhibitors as monotherapy or in combination for metastatic colorectal cancer: A proportion and comparative meta-analysis of efficacy and toxicity from phase I-II-III trials. Crit Rev Oncol Hematol 2025; 211:104741. [PMID: 40274247 DOI: 10.1016/j.critrevonc.2025.104741] [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: 02/19/2025] [Revised: 03/26/2025] [Accepted: 04/19/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND 1-2 % of metastatic colorectal cancers (mCRC) harbor an activating KRAS-G12C mutation. This study aims to pool the results of available clinical trials of KRAS-G12C inhibitors, comparing monotherapy and combinations. METHODS A systematic literature search was conducted in the MEDLINE database and ESMO/ASCO meeting abstracts. Phase I-II-III trials that investigated a KRAS-G12C inhibitor in patients with mCRC were included. The primary endpoints were objective response rate (ORR) and progression-free survival (PFS). Pooled proportions and comparative subgroup analyses for monotherapy and combinations were presented with the random effects model. RESULTS 596 patients with previously treated mCRC in 14 study cohorts treated with one of sotorasib, adagrasib, divarasib, or olomorasib as monotherapy or in combination with cetuximab/panitumumab were included. Combination treatment revealed an ORR of 33.9 % (95 %CI: 20.7-48.4) (I2: 87.1), which is significantly higher than monotherapy [16.7 %, (95 %CI: 8.3-27.3) (I2: 73.2)] (p = 0.045). Median PFS was significantly longer with the combination [5.7 months (95 %CI: 4.4-7.1) (I2: 80.8) vs. 4.2 months (95 %CI: 3.6-4.7) (I2:0.0), p = 0.027]. Grade 3-4 treatment-related adverse events (TRAEs) were significantly more frequent with the combination [32.8 % (95 %CI: 26.4-39.6) (I2:42.5) vs.16.5 % (95 %CI: 4.9-33.1) (I2: 84.2), p = 0.047]. Common adverse events specific to the combinations were skin toxicities, paronychia, and hypomagnesemia. CONCLUSION This analysis suggests that KRAS-G12C inhibitors in combination with anti-EGFR agents may provide a doubled ORR and 1.5-month PFS benefit compared to monotherapy in previously treated mCRC patients, but with a doubled grade 3-4 TRAEs, including skin toxicities, paronychia, and hypomagnesemia. Treatment preferences should be individualized in these highly pretreated patients.
Collapse
Affiliation(s)
- Erman Akkus
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University Cancer Research Institute, Ankara, Türkiye.
| | - Nejat Emre Öksüz
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University Cancer Research Institute, Ankara, Türkiye
| | - Enes Erul
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University Cancer Research Institute, Ankara, Türkiye
| |
Collapse
|
198
|
Hou Y, Xue X, Zhang Z, Mai D, Luo W, Zhou M, Liu Z, Huang Y. Genomic and clinical characterization of HER2 exon 20 mutations in non-small cell lung cancer: insights from a multicenter study in South China. BMC Cancer 2025; 25:752. [PMID: 40264034 PMCID: PMC12012961 DOI: 10.1186/s12885-025-14125-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND The objective of this study was to investigate the clinical and genetic characteristics and clinical relevance of HER2 exon 20 oncogenic variants in non-small cell lung cancer (NSCLC) patients. METHODS This prospective study analyzed 51 NSCLC patients with HER2 mutations, identified via next-generation sequencing (NGS) of tissue, blood, cerebrospinal fluid, or pleural effusion samples. Patients were grouped based on the presence of exon 20 mutations (exon 20 vs. non-exon 20) and further divided based on whether they had received prior anti-tumor treatments (baseline vs. non-baseline). Clinical and genetic data, treatment responses were analyzed. Progression-free survival (PFS) and overall survival (OS) were evaluated using Kaplan-Meier methods and compared with log-rank tests. Gene ontology (GO) analysis was performed to uncover the biological significance of the mutated genes. RESULTS In a cohort of 651 NSCLC patients, 51 (7.83%) harbored HER2 alterations, including 20 (3.08%) with exon 20 mutations. The median age of the HER2-altered subgroup was 58.5 years. Adenocarcinoma was the most prevalent subtype (96.1%), and most patients presented at stage IV (72.5%). The most common metastatic sites were the lungs (68.6%), lymph nodes (52.9%), and brain (43.1%). Among the HER2 mutated patients, 20 (39.3%) had exon 20 mutations. Exon 20 mutations were more prevalent in the non-baseline group (55.0% vs. 29.0%, P = 0.049) and males (75.0%, P = 0.025). These mutations were associated with a higher rate of metastasis to the lungs, lymph nodes (P < 0.001). Patients with exon 20 mutations demonstrated poorer overall survival (OS) outcomes (P = 0.048). No significant differences were observed in age, smoking history, histological subtype, or TNM stage at diagnosis between groups. The majority of exon 20 mutations were in-frame indel mutations (92.0%), with the most common specific mutation being p.Y772_A775dup (70%). Gene Ontology (GO) analysis linked exon 20 mutations to unregulated protein kinase activity and anoikis. CONCLUSIONS Our study found that NSCLC patients with HER2 exon 20 oncogenic variants have a higher risk of metastasis and drug resistance, leading to worse outcomes than non-exon 20 mutations. This highlights the urgent need for targeted therapies aimed at exon 20 insertions to improve survival and treatment outcomes in this subgroup.
Collapse
Affiliation(s)
- Yating Hou
- Department of Oncology, Maoming People's Hospital, 101 Weimin Road, Maoming, 525000, Guangdong, China
| | - Xingyang Xue
- Department of Thoracic Surgery and Oncology, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Centre for Respiratory Disease, Guangzhou, China
| | - Zhuoyun Zhang
- Department of Oncology, Maoming People's Hospital, 101 Weimin Road, Maoming, 525000, Guangdong, China
| | - Dahai Mai
- Department of Oncology, Maoming People's Hospital, 101 Weimin Road, Maoming, 525000, Guangdong, China
| | - Wei Luo
- Department of Oncology, Maoming People's Hospital, 101 Weimin Road, Maoming, 525000, Guangdong, China
| | - Mingyu Zhou
- Department of Oncology, Maoming People's Hospital, 101 Weimin Road, Maoming, 525000, Guangdong, China
| | - Zichuan Liu
- Internal Medicine Section 2, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| | - Yisheng Huang
- Department of Oncology, Maoming People's Hospital, 101 Weimin Road, Maoming, 525000, Guangdong, China.
| |
Collapse
|
199
|
Abrahão R, Cooley JJP, Kahn JM, Brunson A, Alvarez EM, Mahajan A, Wun T, Verma R, Ruddy KJ, Keegan THM. Peripheral Neuropathy Incidence in Children, and Adolescents and Young Adults With Cancer and Medicaid Insurance in California. JCO Oncol Pract 2025:OP2400748. [PMID: 40262078 DOI: 10.1200/op-24-00748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/11/2025] [Accepted: 03/06/2025] [Indexed: 04/24/2025] Open
Abstract
PURPOSE Chemotherapy-induced peripheral neuropathy is a potentially debilitating adverse effect of cancer therapy that can lead to delay, reduction, or discontinuation of cancer treatment. Population-based data on peripheral neuropathy incidence among young cancer survivors are lacking. METHODS Using a linkage between Medicaid, the California Cancer Registry, and hospitalization and emergency department data, we identified 6,028 adolescents and young adults (15-39 years) with Hodgkin lymphoma (HL), non-Hodgkin lymphoma (NHL), breast cancer, colorectal cancer, or testicular cancer and 418 children (<15 years) with HL or NHL during 2005-2017. We determined the cumulative incidence of peripheral neuropathy and its association with neurotoxic chemotherapy identified from Medicaid claims, using multivariable Cox proportional hazards regression models. RESULTS Of 6,446 patients, 1,007 were diagnosed with peripheral neuropathy. Across each cancer type, incidence was higher among patients receiving neurotoxic drugs. For example, compared with non-neurotoxic agents, 5-year cumulative incidence was higher with oxaliplatin for colorectal cancer (24.0% v 6.2%) and paclitaxel for breast cancer (22.6% v 5.1%). In multivariable analysis, the agents most strongly associated with peripheral neuropathy were brentuximab (±other neurotoxic drugs) for HL (hazard ratio [HR], 9.53 [95% CI, 5.95 to 15.26]); brentuximab (±vinca alkaloids; HR, 7.00 [95% CI, 4.13 to 11.87]) for NHL, paclitaxel for breast cancer (HR, 4.03 [95% CI, 3.05 to 5.31]); oxaliplatin for colorectal cancer (HR, 3.46 [95% CI, 2.23 to 5.36]); and cisplatin and etoposide for testicular cancer (HR, 2.06 [95% CI, 1.37 to 3.11]). CONCLUSION The high incidence of peripheral neuropathy highlights the need for frequent monitoring, new supportive care approaches, and development of novel therapeutic agents to minimize toxicity while maintaining treatment efficacy.
Collapse
Affiliation(s)
- Renata Abrahão
- Division of Hematology and Oncology, Center for Oncology Hematology Outcomes Research and Training (COHORT), University of California Davis Comprehensive Cancer Center, Sacramento, CA
| | - Julianne J P Cooley
- California Cancer Reporting and Epidemiologic Surveillance Program, University of California Davis Comprehensive Cancer Center, Sacramento, CA
| | - Justine M Kahn
- Department of Pediatrics, Columbia University Medical Center, New York, NY
| | - Ann Brunson
- Division of Hematology and Oncology, Center for Oncology Hematology Outcomes Research and Training (COHORT), University of California Davis Comprehensive Cancer Center, Sacramento, CA
| | - Elysia M Alvarez
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of California Davis School of Medicine, Sacramento, CA
| | - Anjlee Mahajan
- Division of Hematology and Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA
| | - Ted Wun
- Division of Hematology and Oncology, Center for Oncology Hematology Outcomes Research and Training (COHORT), University of California Davis Comprehensive Cancer Center, Sacramento, CA
- California Cancer Reporting and Epidemiologic Surveillance Program, University of California Davis Comprehensive Cancer Center, Sacramento, CA
| | - Rashmi Verma
- Division of Hematology and Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA
| | | | - Theresa H M Keegan
- Division of Hematology and Oncology, Center for Oncology Hematology Outcomes Research and Training (COHORT), University of California Davis Comprehensive Cancer Center, Sacramento, CA
- California Cancer Reporting and Epidemiologic Surveillance Program, University of California Davis Comprehensive Cancer Center, Sacramento, CA
| |
Collapse
|
200
|
Wang Y, Sun XY, Ma FQ, Ren MM, Zhao RH, Qin MM, Zhu XH, Xu Y, Cao ND, Chen YY, Dong TG, Pan YF, Zhao AG. Morin inhibits ubiquitination degradation of BCL-2 associated agonist of cell death and synergizes with BCL-2 inhibitor in gastric cancer cells. JOURNAL OF INTEGRATIVE MEDICINE 2025:S2095-4964(25)00050-0. [PMID: 40319008 DOI: 10.1016/j.joim.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/26/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVE Gastric cancer (GC) is one of the most common malignancies seen in clinic and requires novel treatment options. Morin is a natural flavonoid extracted from the flower stalk of a highly valuable medicinal plant Prunella vulgaris L., which exhibits an anti-cancer effect in multiple types of tumors. However, the therapeutic effect and underlying mechanism of morin in treating GC remains elusive. The study aims to explore the therapeutic effect and underlying molecular mechanisms of morin in GC. METHODS For in vitro experiments, the proliferation inhibition of morin was measured by cell counting kit-8 assay and colony formation assay in human GC cell line MKN45, human gastric adenocarcinoma cell line AGS, and human gastric epithelial cell line GES-1; for apoptosis analysis, microscopic photography, Western blotting, ubiquitination analysis, quantitative polymerase chain reaction analysis, flow cytometry, and RNA interference technology were employed. For in vivo studies, immunohistochemistry, biomedical analysis, and Western blotting were used to assess the efficacy and safety of morin in a xenograft mouse model of GC. RESULTS Morin significantly inhibited the proliferation of GC cells MKN45 and AGS in a dose- and time-dependent manner, but did not inhibit human gastric epithelial cells GES-1. Only the caspase inhibitor Z-VAD-FMK was able to significantly reverse the inhibition of proliferation by morin in both GC cells, suggesting that apoptosis was the main type of cell death during the treatment. Morin induced intrinsic apoptosis in a dose-dependent manner in GC cells, which mainly relied on B cell leukemia/lymphoma 2 (BCL-2) associated agonist of cell death (BAD) but not phorbol-12-myristate-13-acetate-induced protein 1. The upregulation of BAD by morin was due to blocking the ubiquitination degradation of BAD, rather than the transcription regulation and the phosphorylation of BAD. Furthermore, the combination of morin and BCL-2 inhibitor navitoclax (also known as ABT-737) produced a synergistic inhibitory effect in GC cells through amplifying apoptotic signals. In addition, morin treatment significantly suppressed the growth of GC in vivo by upregulating BAD and the subsequent activation of its downstream apoptosis pathway. CONCLUSION Morin suppressed GC by inducing apoptosis, which was mainly due to blocking the ubiquitination-based degradation of the pro-apoptotic protein BAD. The combination of morin and the BCL-2 inhibitor ABT-737 synergistically amplified apoptotic signals in GC cells, which may overcome the drug resistance of the BCL-2 inhibitor. These findings indicated that morin was a potent and promising agent for GC treatment. Please cite this article as: Wang Y, Sun XY, Ma FQ, Ren MM, Zhao RH, Qin MM, Zhu XH, Xu Y, Cao ND, Chen YY, Dong TG, Pan YF, Zhao AG. Morin inhibits ubiquitination degradation of BCL-2 associated agonist of cell death and synergizes with BCL-2 inhibitor in gastric cancer cells. J Integr Med. 2025; Epub ahead of print.
Collapse
Affiliation(s)
- Yi Wang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xiao-Yu Sun
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Fang-Qi Ma
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ming-Ming Ren
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ruo-Han Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Meng-Meng Qin
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xiao-Hong Zhu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan Xu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ni-da Cao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yuan-Yuan Chen
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Tian-Geng Dong
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yong-Fu Pan
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Ai-Guang Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|