1
|
Liu S, Wang G, Niu X, Wang W. Association of Log Odds of Positive Lymph Nodes with Overall Survival in Patients with Primary Malignant Anorectal Melanoma: A Population-Based Study. J INVEST SURG 2025; 38:2493886. [PMID: 40313209 DOI: 10.1080/08941939.2025.2493886] [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: 03/01/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Malignant anorectal melanoma (AM) is a rare tumor characterized by its aggressive behavior. To date, there has been no comprehensive investigation into the association between the log odds of positive lymph nodes (LODDS) and the prognosis of patients with malignant AM who have undergone surgical intervention. METHODS Data were extracted from the Surveillance, Epidemiology and End Results (SEER) database for patients diagnosed with primary malignant AM who have undergone surgical intervention between 2000 and 2021. The Kaplan-Meier (KM) method with the log-rank test was used to compare survival curves between different LODDS groups. Multivariate Cox models were employed to evaluate the independent association between LODDS and overall survival (OS). An analysis to explore the potential influence of unmeasured confounders on the relationship between LODDS and OS was conducted using the E value. RESULTS The study included a total of 126 eligible patients. KM analysis revealed that patients with low LODDS exhibited significantly improved OS compared to those with high LODDS (p < 0.001). The multivariate Cox analysis demonstrated that LODDS was independently associated with OS. The calculated E value suggested that the potential impact of unmeasured confounders on the association between LODDS and OS was likely minimal. CONCLUSION The findings indicate that LODDS is independently associated with the prognosis of patients with AM undergoing surgical intervention. These results may enhance clinicians' understanding of the prognosis of this rare malignancy and provide a basis for guiding therapeutic decisions.
Collapse
Affiliation(s)
- Shaofeng Liu
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Guixiang Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Xiaoping Niu
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Wei Wang
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
2
|
Zhou MW, Ma JX, Yan ZQ, Qiao ZD, Li ZY, Gu XD, Yan S, Zang YW, Chen ZY, Zhou YM, Ding JH, Xiang JB. A Porcine Model of Laparoscopic Intersphincteric Resection for Ultra-Low Rectal Cancer. J INVEST SURG 2025; 38:2497327. [PMID: 40329519 DOI: 10.1080/08941939.2025.2497327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVE To develop a porcine model for laparoscopic intersphincteric resection (ISR) targeting ultra-low rectal cancer. METHODS Six Bama mini-pigs underwent a standard laparoscopic ISR procedure: (1) Ligation and transection of the inferior mesenteric vessels; (2) Exposure and sparing of the pelvic autonomic nerves; (3) Dissection of the rectum; (4) Dissection and transection of hiatal ligament and rectourethralis muscle; (5) Transanal dissection of the anal canal; (6) Dissection of the intersphincteric space (ISS); (7) End-to-end handsewn coloanal anastomosis (CAA). Pathological evaluations were conducted to ascertain the precision of the ISR. Furthermore, postoperative fecal incontinence scores were systematically assessed on a weekly basis throughout a 12-week post-surgery follow-up period. RESULTS All six pigs survived surgery without conversion to an open approach. There were no occurrences of incisional infection or any surgical complications, including anastomotic leakage, anastomotic bleeding, or ileus. Postoperatively, one pig manifested constipation and three exhibited anal redness, both conditions ameliorating within a week. No erosive ulcers were detected. Notably, all animals exhibited fecal staining on their tails within the first two weeks post-surgery, transitioning to formed feces by the third week. Improvement in defecation frequency was observed after 6 weeks, with a noticeable reduction in defecation areas after one month. Incontinence scores, evaluated three months post ISR surgery, revealed no significant abnormalities in anal function compared to preoperative assessments. CONCLUSIONS This study established a replicable porcine model for laparoscopic ISR, demonstrating its potential utility in clinical and fundamental research related to ultra-low rectal cancer treatment.
Collapse
Affiliation(s)
- Min-Wei Zhou
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ji-Xin Ma
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zheng-Qing Yan
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zheng-Dong Qiao
- Experimental Animal Center, Pudong Hospital, Fudan University, Shanghai, China
| | - Zhen-Yang Li
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiao-Dong Gu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Sen Yan
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Wen Zang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zong-You Chen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Ming Zhou
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian-Hua Ding
- Department of Colorectal Surgery, Characteristic Medical Centre of PLA Rocket Force, Beijing, China
| | - Jian-Bin Xiang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Ying H, Chen Y, Hong Y, Ying K, Li S, Zhang Y, Mei T, Song X, He Y, Yao C, Yu F. L3-SMI as a predictor of overall survival in oesophageal cancer patients receiving PD-1 inhibitors combined with chemotherapy. Ann Med 2025; 57:2440114. [PMID: 39665392 PMCID: PMC11639058 DOI: 10.1080/07853890.2024.2440114] [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: 07/22/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Programmed death ligand-1 (PD-1), as an immunotherapy target, has been increasingly used in tumour therapies. But as reactions and outcomes to PD-1 inhibitors combined with chemotherapy vary individually, it is primarily important to identify an ideal indicator for predicting the therapeutic effectiveness in individual patients. Oesophageal cancer (EC) patients often have difficulty eating due to tumour blockage of the oesophagus, leading to malnutrition and muscle loss. Sarcopenia is one of the influencing factors for poor prognosis in tumour patients, but its role in PD-1 inhibitors combined with chemotherapy of EC patients is not fully clarified. In this study, we aimed to explore the prognostic significance of Sarcopenia measured by CT in EC patients treated with PD-1 antibody combined with chemotherapy. METHODS The third lumbar skeletal muscle mass index (L3-SMI) was obtained from 83 EC patients before and 3 months after administration of PD-1 inhibitors combined with chemotherapy using conventional CT scans. RESULTS Baseline L3-SMI and 3-month L3-SMI values were found not suitable for predicting the overall survival (OS) of EC patients (p = 0.32 & p = 0.055). Longitudinal change in L3-SMI (ΔL3-SMI) during PD-1 inhibitors combined with chemotherapy was identified as a relevant marker of OS in univariable analysis (HR: 0.98, 95% CI: 0.96-1.00, p = 0.042) and multivariable analysis (HR: 0.96, 95% CI: 0.93-0.99, p = 0.02). L3-SMI-positive patients generally had better OS (p = 0.041). CONCLUSION Excessive muscle loss rather than muscle loss before and after administration of PD-1 inhibitors combined with chemotherapy is an important prognostic factor for therapeutic outcomes and OS in EC patients.
Collapse
Affiliation(s)
- Huiya Ying
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuhao Chen
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiwen Hong
- WenZhou Medical University, Wenzhou, Zhejiang, China
| | - Kanglei Ying
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shiyu Li
- WenZhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxuan Zhang
- WenZhou Medical University, Wenzhou, Zhejiang, China
| | - Tianhao Mei
- WenZhou Medical University, Wenzhou, Zhejiang, China
| | - Xian Song
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuanhang He
- WenZhou Medical University, Wenzhou, Zhejiang, China
| | - Chenrui Yao
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fujun Yu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
4
|
Valle J. Biofilm-associated proteins: from the gut biofilms to neurodegeneration. Gut Microbes 2025; 17:2461721. [PMID: 39898557 PMCID: PMC11792866 DOI: 10.1080/19490976.2025.2461721] [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: 10/25/2024] [Revised: 12/07/2024] [Accepted: 01/28/2025] [Indexed: 02/04/2025] Open
Abstract
Human microbiota form a biofilm with substantial consequences for health and disease. Numerous studies have indicated that microbial communities produce functional amyloids as part of their biofilm extracellular scaffolds. The overlooked interplay between bacterial amyloids and the host may have detrimental consequences for the host, including neurodegeneration. This work gives an overview of the biofilm-associated amyloids expressed by the gut microbiota and their potential role in neurodegeneration. It discusses the biofilm-associated proteins (BAPs) of the gut microbiota, maps the amyloidogenic domains of these proteins, and analyzes the presence of bap genes within accessory genomes linked with transposable elements. Furthermore, the evidence supporting the existence of amyloids in the gut are presented. Finally, it explores the potential interactions between BAPs and α-synuclein, extending the literature on amyloid cross-kingdom interactions. Based on these findings, this study propose that BAP amyloids act as transmissible catalysts, facilitating the misfolding, accumulation, and spread of α-synuclein aggregates. This review contributes to the understanding of complex interactions among the microbiota, transmissible elements, and host, which is crucial for developing novel therapeutic approaches to combat microbiota-related diseases and improve overall health outcomes.
Collapse
Affiliation(s)
- Jaione Valle
- Microbial Biotechnology Department, Instituto de Agrobiotecnología, CSIC-Gobierno de Navarra, Mutilva, Navarra, Spain
| |
Collapse
|
5
|
Masaadeh AH, Eletrebi M, Parajuli B, De Jager N, Bosch DE. Human colitis-associated colorectal carcinoma progression is accompanied by dysbiosis with enriched pathobionts. Gut Microbes 2025; 17:2479774. [PMID: 40094201 PMCID: PMC11917176 DOI: 10.1080/19490976.2025.2479774] [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: 01/06/2025] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Dysbiosis and pathobionts contribute to inflammation and the risk of colitis-associated carcinoma (CAC) in animal models, but their roles in humans with this uncommon disease are unknown. We identified microbiome differences in human CAC compared with longstanding inflammatory bowel disease (IBD) and sporadic colorectal carcinoma (CRC). Twenty-four CAC resections were matched with CRC and IBD controls. Methods included histopathology, 16S rDNA metagenomics, and pathobiont-specific qPCR. Beta diversity differed by diagnosis (PERMANOVA p = 0.007). The distinguishing taxa included Akkermansia enriched in CRC, and Bacteroides spp. enriched in IBD. The non-neoplastic mucosae presented distinct beta diversity (p = 0.005), but the CAC/CRC tumor microbiomes were similar (p = 0.7). Within metastases and margins, Enterobacteriaceae were enriched in CAC, and Bacteroidales in CRC. Pathobiont-specific qPCR confirmed a greater frequency of pks+ E. coli and enterotoxigenic Bacteroides fragilis in CAC than IBD. High alpha diversity was associated with active inflammation, advanced cancer stage, and shorter overall survival (log-rank p = 0.008). Mucosal microbiomes distinguish CAC from longstanding IBD, implicating pathobionts as markers for disease progression. Integrating our findings with prior animal model research, pathobionts promote carcinogenesis in IBD patients through genotoxicity and host cell signaling.
Collapse
Affiliation(s)
- Amr H Masaadeh
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mohamed Eletrebi
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Bishal Parajuli
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nicola De Jager
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Dustin E Bosch
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, Iowa City, IA, USA
| |
Collapse
|
6
|
Davola ME, Cormier O, Lepard M, McNicol J, Collins S, Hammill JA, Silvestri C, Bramson JL, Gillgrass A, Mossman KL. Humanized mouse models of KRAS-mutated colorectal and pancreatic cancers with HLA-class-I match for pre-clinical evaluation of cancer immunotherapies. Oncoimmunology 2025; 14:2473163. [PMID: 40017442 PMCID: PMC11875485 DOI: 10.1080/2162402x.2025.2473163] [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: 06/07/2024] [Revised: 12/17/2024] [Accepted: 02/24/2025] [Indexed: 03/01/2025] Open
Abstract
Cancer immunotherapy promises to treat challenging cancers including KRAS-mutated colorectal cancer (CRC) and pancreatic ductal adenocarcinoma (PDAC). However, pre-clinical animal models that better mimic patient tumor and immune system interactions are required. While humanized mice are promising vehicles for pre-clinical immunotherapy testing, currently used cancer models retain limitations, such as lack of a human thymus for human leukocyte antigen (HLA)-based education of human T cells. As cytotoxic T lymphocyte (CTL) activity underlies many immunotherapies, we developed more clinically relevant KRAS-mutated CRC and PDAC humanized cancer models using transgenic NRG-A2 mice expressing HLA-A2.1 to enable HLA-class-I match between mouse tissues (including the thymus), the humanized immune system and human tumors. Using these novel humanized cancer models and a CTL-mediated combination (immuno)therapy with clinical potential, we were able to recapitulate the complexity and therapy-induced changes reported in patient biopsies, demonstrating the use of these HLA-matched models for pre-clinical validation of novel immunotherapies.
Collapse
Affiliation(s)
- Maria E. Davola
- Department of Medicine, Centre for Discovery in Cancer Research and McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Olga Cormier
- Department of Medicine, Centre for Discovery in Cancer Research and McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Madeleine Lepard
- Department of Medicine, Centre for Discovery in Cancer Research and McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Jamie McNicol
- Department of Medicine, Centre for Discovery in Cancer Research and McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Susan Collins
- Department of Medicine, Centre for Discovery in Cancer Research and McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Joanne A. Hammill
- Department of Medicine, Centre for Discovery in Cancer Research and McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Christopher Silvestri
- Department of Medicine, Centre for Discovery in Cancer Research and McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Jonathan L. Bramson
- Department of Medicine, Centre for Discovery in Cancer Research and McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Amy Gillgrass
- Department of Medicine, Centre for Discovery in Cancer Research and McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Karen L. Mossman
- Department of Medicine, Centre for Discovery in Cancer Research and McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
7
|
Xue N, Wen X, Wang Q, Shen Y, Qu Y, Xu Q, Chen S, Chen J. Establishing and validating models integrated with hematological biomarkers and clinical characteristics for the prognosis of non-esophageal squamous cell carcinoma patients. Ann Med 2025; 57:2483985. [PMID: 40152751 PMCID: PMC11956093 DOI: 10.1080/07853890.2025.2483985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/11/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND This study aimed to construct a novel model and validate its predictive power in non-esophageal squamous cell carcinoma (NESCC) patients. METHODS This retrospective study included 151 patients between October 2006 and September 2016. The LASSO Cox and Random Survival Forest (RSF) models were developed with the help of hematological biomarkers and clinical characteristics. The concordance index (C-index) was used to assess the prognostic power of the LASSO Cox model, RSF model, and TNM staging. Based on the risk scores of the LASSO Cox and RSF models, we divided patients into low-risk and high-risk subgroups. RESULTS We constructed two models in NESCC patients according to LASSO Cox regression and RSF models. The RSF model reached a C-index of 0.841 (95% CI: 0.792-0.889) in the primary cohort and 0.880 (95% CI: 0.830-0.930) in the validation cohort, which was higher than the C-index of the LASSO Cox model 0.656 (95% CI: 0.580-0.732) and 0.632 (95% CI: 0.542-0.720) in the two cohorts. The integrated C/D area under the ROC curve (AUC) values for the LASSO Cox and RSF models were 0.701 and 0.861, respectively. In both two models, Kaplan-Meier survival analysis and the estimated restricted mean survival time (RMST) values indicated that the low-risk subgroup had a better prognostic outcome than the high-risk subgroup (p < 0.05). CONCLUSIONS The RSF model has better prediction power than the LASSO Cox and the TNM staging models. It has a guiding value for the choice of individualized treatment in patients with NESCC.
Collapse
Affiliation(s)
- Ning Xue
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Xiaoyan Wen
- Central Sterilization Supply Department, The Guanghua Stomatological College of Sun Yat-sen University, Hospital of Stomatology, SunYat-sen University, Guangzhou, P. R. China
| | - Qian Wang
- Department of radiation oncology, China–Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Yong Shen
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Yuanye Qu
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Qingxia Xu
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Shulin Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Research Center for Translational Medicine, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P. R. China
| | - Jing Chen
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| |
Collapse
|
8
|
Kong Y, Huang X, Peng G, Cao X, Zhou X. Efficacy of first-line radiofrequency ablation combined with systemic chemotherapy plus targeted therapy for initially unresectable colorectal liver metastases. Int J Hyperthermia 2025; 42:2432988. [PMID: 39894452 DOI: 10.1080/02656736.2024.2432988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/16/2024] [Accepted: 11/18/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND/OBJECTIVE The optimal strategy for patients with colorectal liver metastases is still controversially discussed. This study aimed to evaluate the efficacy of radiofrequency ablation (RFA) combined with systemic chemotherapy plus targeted therapy as first-line treatment in patients with initially unresectable colorectal liver metastases (CRLM), to identify prognostic factors and construct nomograms predicting survival. METHODS This retrospective study included patients with initially unresectable CRLM treated with (study group n = 74) or without (control group n = 83) RFA at the National Cancer Center from January 2018 to January 2021. Survival curves were assessed using the Kaplan-Meier method and log-rank test. Univariate and multivariate Cox regression analyses were used to determine prognostic factors and include these factors in the nomograms to predict progression-free survival (PFS) and overall survival (OS). RESULTS The study group had significantly better median PFS (17.16 months vs. 8.35 months, p < 0.01) and OS (34.9 months vs. 21.1 months, p < 0.01) than the control group after propensity score matching. Cox regression analyses identified RFA treatment and clinical risk score (CRS) as independent prognostic factors for PFS. The largest diameter of liver metastases, RFA treatment, and CRS were independent prognostic factors for OS. Based on this finding, nomograms with good discrimination and calibration were constructed. CONCLUSION RFA combined with systemic chemotherapy plus targeted therapy as first-line treatment could significantly prolong PFS and OS in patients with initially unresectable CRLM compared with systemic chemotherapy plus targeted therapy. The nomograms predicting PFS and OS might help clinicians select personalized treatment.
Collapse
Affiliation(s)
- Yaqing Kong
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyu Huang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Peng
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaojing Cao
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiang Zhou
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Sheng J, Yang Y, Qiu Y, Lu C, Fu X. Solid and cystic intrapancreatic accessory spleen: report of 10 cases in a single institution. Ann Med 2025; 57:2463564. [PMID: 39927469 PMCID: PMC11812107 DOI: 10.1080/07853890.2025.2463564] [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: 09/05/2024] [Revised: 10/21/2024] [Accepted: 01/07/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Precise diagnosis of intrapancreatic accessory spleen (IPAS) remains challenging due to its rarity and diverse presentations. Despite comprehensive examinations, including radiography and other diagnostic methods, the potential for malignancy cannot be excluded, often leading to unnecessary pancreatic surgeries. We review our institutional experience to provide insights for accurately distinguishing IPAS. METHODS We retrospectively reviewed 10 patients who underwent distal pancreatectomy for the lesion in the pancreas tail which was determined to be IPAS on final pathology at our institution between January 2020 and April 2024. The presenting symptoms, medical history, preoperative imaging, operative therapy, final pathology and postoperative course were evaluated. RESULTS Patient ages ranged from 30 to 72 (median 55.5), including six women and four men. Most patients were asymptomatic. One patient had the medical history of splenectomy. Lesions ranged from 1.4 to 7.3 cm (mean 2.9 cm). All lesions were located in the pancreatic tail. On radiologic evaluation, these lesions had both solid and cystic presentations. The most common operative approach was laparoscopic distal pancreatectomy and splenectomy. Four patients were diagnosed with epidermoid cysts arising in intrapancreatic accessory spleen (ECIPAS) on final pathologic evaluation. CONCLUSIONS IPAS are predominantly benign lesions which have solid and cystic presentations commonly mistaken for pancreatic neoplasms. Combining CT, MRI, EUS-FNA and nuclear medicine may enhance IPAS detection, though no definitive diagnostic method exists. Increased awareness of IPAS in the differential diagnosis of pancreatic tail tumors, coupled with advancements in imaging techniques could improve diagnostic accuracy and exclude malignancy, preventing unnecessary surgeries.
Collapse
Affiliation(s)
- Jianjie Sheng
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yifei Yang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yudong Qiu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chenglin Lu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xu Fu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
10
|
Lin J, Wang J, Zhao K, Li Y, Zhang X, Sheng J. Molecular targets and mechanisms of traditional Chinese medicine combined with chemotherapy for gastric cancer: a meta-analysis and multi-omics approach. Ann Med 2025; 57:2494671. [PMID: 40317214 PMCID: PMC12051567 DOI: 10.1080/07853890.2025.2494671] [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: 07/21/2024] [Revised: 01/23/2025] [Accepted: 04/05/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND The combination of traditional Chinese medicine (TCM) with chemotherapy has been widely applied in the treatment of gastric cancer (GC). However, previous clinical studies have been constrained by small sample sizes and a lack of investigation into the molecular mechanisms of TCM. This study aims to assess the efficacy of TCM in treating GC by leveraging the strengths of meta-analysis and multi-omics approaches while also summarizing the underlying pharmacological mechanisms. METHODS A systematic literature review and meta-analysis were conducted using online databases to collect data before May 2024. This was to investigate the association between TCM combined with chemotherapy and the prognosis in GC. The molecular targets between the high-frequency TCMs and GC were identified through network pharmacology. The underlying mechanisms were investigated using multi-omics. RESULTS 9 studies with 2,158 patients were included. The meta-analysis results demonstrated that the combination of TCM and chemotherapy significantly improved the overall survival (OS) of GC patients (OR = 2.91; 95% CI: 2.70-3.12, p < 0.00001) and enhanced their quality of life (OR = 4.00; 95% CI: 1.99-8.03, p < 0.0001). Network pharmacology analysis identified 13 potential molecular targets of TCM in GC; additionally, multi-omics analysis highlighted the significant roles of MK, MIF, GALECTIN, and CypA signaling pathways in GC. CONCLUSION The combination of TCM with chemotherapy significantly improves the prognosis of GC; future research can focus on these key molecular targets and signaling pathways. This supports the application of precision medicine in cancer treatment and suggests the rational use of TCM in managing GC.
Collapse
Affiliation(s)
- Jie Lin
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jincheng Wang
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Kai Zhao
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yongzhi Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xuewen Zhang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jiyao Sheng
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Huang FJ, Fang YY, Wen JY, Li JJ, Lin Q, Su QY, Chen YY, Wang L, Zeng JJ, Chi BT, He RQ, Qin DY, Yang LH, Chen G. From PD-1/PD-L1 to tertiary lymphoid structures: Paving the way for precision immunotherapy in cholangiocarcinoma treatment. Hum Vaccin Immunother 2025; 21:2444697. [PMID: 39757910 DOI: 10.1080/21645515.2024.2444697] [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: 09/17/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a highly malignant hepatobiliary tumor characterized by limited treatment options and poor prognosis. The recent rise of immunotherapy has significantly influenced research in this field. This study presents a bibliometric analysis of 416 articles retrieved from the WOSCC, Wan fang Data, CNKI and VIP databases, spanning contributions from 32 countries, 589 institutions and 3,200 authors. The analysis identified "PD-L1," "PD-1" and "pembrolizumab" as central research foci, while "immune checkpoint inhibitors," "tumor immune microenvironment," "tertiary lymphoid structures" and "durvalumab" emerged as key areas of interest. These findings emphasize the pivotal role of immunotherapy in improving survival outcomes for CCA, and they highlight the significance of tertiary lymphoid structures within the tumor microenvironment as a promising target for future research. This study offers a strategic overview of the evolving landscape of CCA immunotherapy, providing valuable insights to guide future scientific endeavors in this domain.
Collapse
Affiliation(s)
- Fang-Ju Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Ye-Ying Fang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jia-Ying Wen
- Department of Radiotherapy, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jian-Jun Li
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Qian Lin
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Qin-Yan Su
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Yi-Yang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Lei Wang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jian-Jia Zeng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Bang-Teng Chi
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Di-Yuan Qin
- Department of Computer Science and Technology, School of Computer and Electronic Information, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Li-Hua Yang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| |
Collapse
|
12
|
Christou CD, Sitsiani O, Boutos P, Katsanos G, Papadakis G, Tefas A, Papalois V, Tsoulfas G. Comparison of ChatGPT-3.5 and GPT-4 as potential tools in artificial intelligence-assisted clinical practice in renal and liver transplantation. World J Transplant 2025; 15:103536. [DOI: 10.5500/wjt.v15.i3.103536] [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: 11/21/2024] [Revised: 01/26/2025] [Accepted: 03/05/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Kidney and liver transplantation are two sub-specialized medical disciplines, with transplant professionals spending decades in training. While artificial intelligence-based (AI-based) tools could potentially assist in everyday clinical practice, comparative assessment of their effectiveness in clinical decision-making remains limited.
AIM To compare the use of ChatGPT and GPT-4 as potential tools in AI-assisted clinical practice in these challenging disciplines.
METHODS In total, 400 different questions tested ChatGPT’s/GPT-4 knowledge and decision-making capacity in various renal and liver transplantation concepts. Specifically, 294 multiple-choice questions were derived from open-access sources, 63 questions were derived from published open-access case reports, and 43 from unpublished cases of patients treated at our department. The evaluation covered a plethora of topics, including clinical predictors, treatment options, and diagnostic criteria, among others.
RESULTS ChatGPT correctly answered 50.3% of the 294 multiple-choice questions, while GPT-4 demonstrated a higher performance, answering 70.7% of questions (P < 0.001). Regarding the 63 questions from published cases, ChatGPT achieved an agreement rate of 50.79% and partial agreement of 17.46%, while GPT-4 demonstrated an agreement rate of 80.95% and partial agreement of 9.52% (P = 0.01). Regarding the 43 questions from unpublished cases, ChatGPT demonstrated an agreement rate of 53.49% and partial agreement of 23.26%, while GPT-4 demonstrated an agreement rate of 72.09% and partial agreement of 6.98% (P = 0.004). When factoring by the nature of the task for all cases, notably, GPT-4 demonstrated outstanding performance, providing a differential diagnosis that included the final diagnosis in 90% of the cases (P = 0.008), and successfully predicting the prognosis of the patient in 100% of related questions (P < 0.001).
CONCLUSION GPT-4 consistently provided more accurate and reliable clinical recommendations with higher percentages of full agreements both in renal and liver transplantation compared with ChatGPT. Our findings support the potential utility of AI models like ChatGPT and GPT-4 in AI-assisted clinical practice as sources of accurate, individualized medical information and facilitating decision-making. The progression and refinement of such AI-based tools could reshape the future of clinical practice, making their early adoption and adaptation by physicians a necessity.
Collapse
Affiliation(s)
- Chrysanthos D Christou
- Center for Research and Innovation in Solid Organ Transplantation, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54622, Greece
| | - Olga Sitsiani
- School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54622, Greece
| | - Panagiotis Boutos
- School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54622, Greece
| | - Georgios Katsanos
- Center for Research and Innovation in Solid Organ Transplantation, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54622, Greece
| | - Georgios Papadakis
- Department of Nephrology and Transplantation, Guy’s Hospital, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 1UL, United Kingdom
| | - Anastasios Tefas
- Computational Intelligence and Deep Learning Group, Department of Informatics, Aristotle University of Thessaloniki, Thessaloniki 54636, Greece
| | - Vassilios Papalois
- Renal and Transplant Unit, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London W120HS, United Kingdom
| | - Georgios Tsoulfas
- Center for Research and Innovation in Solid Organ Transplantation, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54622, Greece
| |
Collapse
|
13
|
Peng H, Sun L, Zhao J, Cui G. Electrochemical detection of circulating-free DNA methylation: A new indicator for early cancer screening. Talanta 2025; 292:127925. [PMID: 40081249 DOI: 10.1016/j.talanta.2025.127925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/23/2025] [Accepted: 03/10/2025] [Indexed: 03/15/2025]
Abstract
This study introduces a novel electrochemical approach for detecting circulating-free DNA (cfDNA) by leveraging distinct methylation patterns characteristic of cfDNA from healthy controls and cancer patients. Our findings reveal that cfDNA from cancer patients exhibits a unique affinity for gold surfaces due to abnormal methylation, which enables its electrochemical differentiation. By using square wave voltammetry (SWV) with optimized parameters, we achieved high sensitivity (0.89) and specificity (0.73) in distinguishing cfDNA from healthy controls and cancer patients. Clinical trials demonstrated the effectiveness of this method, surpassing conventional tumor markers in early cancer screening. SEPTIN9 (SEPT9) methylation is used as an example to demonstrate the generalizability of our method for analyzing DNA methylation changes, which can subsequently be applied to identify the presence of cancer.This approach offers notable advantages, including simplicity, rapid response, and cost-effectiveness. While the current methodology is primarily suited for detecting the presence of cancer, its potential for prognostic and diagnostic applications warrants further investigation.
Collapse
Affiliation(s)
- Han Peng
- School of Mechanical and Automotive Engineering, South China University of Technology, Guangzhou, 510640, PR China.
| | - Liyue Sun
- Department of Health Management Centre, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China.
| | - Jie Zhao
- School of Mechanical and Automotive Engineering, South China University of Technology, Guangzhou, 510640, PR China.
| | - Guofeng Cui
- School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, PR China.
| |
Collapse
|
14
|
Gao F, Ma Y, Yu C, Duan Q. miR-125b-5p regulates FFA-induced hepatic steatosis in L02 cells by targeting estrogen-related receptor alpha. Gene 2025; 959:149419. [PMID: 40113187 DOI: 10.1016/j.gene.2025.149419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/02/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND & AIMS NAFLD is a global and complex liver disease caused by multiple factors. Intrahepatocellular steatosis is the primary prerequisite for the occurrence and development of NAFLD. It has been shown that miR-125b-5p is highly correlated with NAFLD, and ESRRA is a factor that regulates lipid metabolism. The purpose of our study is to investigate whether miR-125b-5p regulates FFA-induced steatosis in L02 cells by targeting ESRRA. APPROACHES AND RESULTS Estrogen-related receptor alpha (ESRRA) was identified as a direct target of miR-125b-5p through database prediction and a dual-luciferase reporter gene assay. L02 cells were induced with free fatty acids (OA:PA, 2:1) at concentrations of 0.3 mM, 0.6 mM, 0.9 mM, 1.2 mM and 1.5 mM for 24 h, 48 h and 72 h, respectively. The degree of hepatocyte steatosis and triglyceride content were separately manifested by oil red O staining and colorimetric method. Cell viability per group was detected by CCK-8 assay. Eventually, 0.9 mM and 24 h were screened out as the optimal concentration and time for establishing the in-vitro model of hepatic steatosis. Followingly, miR-125b-5p and ESRRA were knocked down by transient transfection. We monitored the expressions of lipid metabolism factors SREBP-1c, ACC1 and FAS and determine triglyceride content within the cells per group. The data showed that knockdown of ESRRA led to down-regulation of the expressions of SREBP-1, ACC1, FAS and triglyceride content. Meanwhile, knockdown of ESRRA and miR-125b-5p resulted that the expressions of ESRRA, SREBP-1, ACC1, FAS and triglyceride content rebounded. CONCLUSIONS MiR-125b-5p down-regulates the expressions of lipid metabolism-related factors by negatively regulating ESRRA, thereby improving hepatic steatosis.
Collapse
Affiliation(s)
- Fen Gao
- Gansu University of Chinese Medicine, Gansu 730000, China.
| | - Yanhua Ma
- Gansu University of Chinese Medicine, Gansu 730000, China.
| | - Chun Yu
- Gansu University of Chinese Medicine, Gansu 730000, China
| | | |
Collapse
|
15
|
Saadh MJ, Allela OQB, Kareem RA, Baldaniya L, Ballal S, Vashishth R, Parmar M, Sameer HN, Hamad AK, Athab ZH, Adil M. Prognostic gene expression profile of colorectal cancer. Gene 2025; 955:149433. [PMID: 40122415 DOI: 10.1016/j.gene.2025.149433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer is a major global health burden, with significant heterogeneity in clinical outcomes among patients. Identifying robust prognostic gene expression signatures can help stratify patients, guide treatment decisions, and improve clinical management. This review provides an overview of current prognostic gene expression profiles in colorectal cancer research. We have synthesized evidence from numerous published studies investigating the association between tumor gene expression patterns and patient survival outcomes. The reviewed literature reveals several promising gene signatures that have demonstrated the ability to predict disease-free survival and overall survival in CRC patients, independent of standard clinicopathological risk factors. These genes are crucial in fundamental biological processes, including cell cycle control, epithelial-mesenchymal transition, and immune regulation. The implementation of prognostic gene expression tests in clinical practice holds great potential for enabling more personalized management strategies for colorectal cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Manisha Parmar
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India.
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq.
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq.
| | | |
Collapse
|
16
|
Dapper H, Fleischmann M, Tselis N, Diefenhardt M, Hofheinz RD, Weiß C, Grabenbauer GG, Merten R, Grosu AL, Kirste S, Rieken S, Claßen J, Langer C, Kuhnt T, Schmidberger H, Ghadimi M, Giordano F, Nestle U, Koerber SA, Bock F, Geiger M, Polat B, Bruns CJ, Dieplinger G, Popp F, Zander T, Brunner T, Tribius S, Arnold D, Wurschi G, Piso P, Friede T, Hörner-Rieber J, Gkika E, Rödel C, Fokas E. ACO/ARO/AIO-22 - External beam radiotherapy combined with endorectal high-dose-rate brachytherapy in elderly and frail patients with rectal cancer: A prospective multicentre trial of the German Rectal Cancer Study Group. Clin Transl Radiat Oncol 2025; 53:100958. [PMID: 40276115 PMCID: PMC12020863 DOI: 10.1016/j.ctro.2025.100958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/01/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
Purpose Rectal cancer disproportionately affects the elderly population, with more than half of cases diagnosed in individuals aged 70 years or older. Frail patients in this group often face significant challenges tolerating the standard treatment of total mesorectal excision (TME), due to elevated risks of surgical complications and perioperative morbidity. Advances in radiotherapy techniques offer a viable alternative, providing effective tumor control while minimizing treatment-related toxicity. Recent clinical trials, including OPERA and MORPEUS, have demonstrated that dose-escalated radiotherapy, which integrates external beam (chemo)radiotherapy (EBRT) with high-dose-rate endorectal brachytherapy (HDR-BT) or contact X-ray brachytherapy (CXB), can achieve high clinical response rates and facilitate organ preservation in younger, healthier patient cohorts. Building on these findings, the ACO/ARO/AIO-22 study seeks to adapt these innovative approaches to meet the needs of elderly and frail patients with rectal cancer. Methods/Design The ACO/ARO/AIO-22 trial is a prospective multicentre controlled trial. Elderly (age ≥70 years) and/or frail patients with non-metastatic rectal adenocarcinoma (cT1-3d N0/+ M0, mrCRM - / +) localized 0-16 cm from the ano-cutaneous line, unable to undergo radical surgery can be included. The initial treatment comprises an external beam radiation (EBRT) regime with 13 × 3 Gy (total: 39 Gy) over a period of two and a half weeks. Following initial restaging 6.5 weeks after completion of EBRT, endorectal HDR-BT will be delivered with 3 weekly fractions of 8 Gy to a total dose of 24 Gy (prescribed at the radial margin of the tumor; with a maximum prescription depth of 10 mm); alternatively, with CXB with 90 Gy in 3 weekly fractions. The primary objective is complete or near complete clinical response (cCR or ncCR) and the second primary endpoint is quality of life (QoL) measured with the EORTC QLQ-ELD14, both at 12 months after treatment start. Discussion The ACO/ARO/AIO-22 prospective multicentre trial will evaluate organ preservation rates and QoL after combining EBRT with endorectal HDR-BT in elderly and/or frail patients with rectal cancer.Trial Registration:Clinicialtrials.gov number: NCT06729645.
Collapse
Affiliation(s)
- Hendrik Dapper
- Department of Radiation Oncology, Cyberknife and Radiation Therapy, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Maximilian Fleischmann
- Department of Radiation Oncology, University Hospital Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Nikolaos Tselis
- Department of Radiation Oncology, University Hospital Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Markus Diefenhardt
- Department of Radiation Oncology, University Hospital Johann Wolfgang Goethe University, Frankfurt, Germany
- Frankfurt Cancer Institute, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Christian Weiß
- Department of Radiotherapy and Oncology, Community Hospital, Darmstadt, Germany
| | - Gerhard G. Grabenbauer
- Department of Radiation Oncology and Radiotherapy, DiaCura & Klinikum Coburg, Coburg, Germany
| | - Ricarda Merten
- Department of Radiation Therapy, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, Freiburg, Germany
- German Cancer Research Center, German Cancer Consortium, Freiburg, Germany
| | - Simon Kirste
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, Comprehensive Cancer Center Lower Saxony (CCC-N), University Hospital Göttingen (UMG), Göttingen, Germany
| | - Johannes Claßen
- Department of Radiation Oncology, St. Vincentius-Kliniken gAG, Karlsruhe, Germany
| | | | - Thomas Kuhnt
- Department of Radiation Therapy, University of Leipzig, Leipzig, Germany
| | - Heinz Schmidberger
- Department of Radiation Oncology and Radiation Therapy, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center, Göttingen, Germany
| | - Frank Giordano
- Department of Radiation Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Kliniken Maria Hilf, Moenchengladbach, Germany
| | - Stefan A. Koerber
- Department of Radiation Oncology, Barmherzige Brüder Hospital Regensburg, Regensburg, Germany
| | - Felix Bock
- Department of Radiotherapy and Oncology, University of Rostock, Rostock, Germany
| | - Matthias Geiger
- Department of Radiotherapy and Oncology, Ostfildern Medius Kliniken, Ostfildern, Germany
| | - Bülent Polat
- Department of Radiation Oncology, University of Würzburg, Würzburg, Germany
| | | | - Georg Dieplinger
- Department of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düesseldorf, University Hospital of Cologne, Cologne, Germany
| | - Felix Popp
- Department of Visceral Surgery, University of Cologne, Cologne, Germany
| | - Thomas Zander
- Department of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düesseldorf, University Hospital of Cologne, Cologne, Germany
| | - Thomas Brunner
- Department of Radiation Oncology, Medical University of Graz, Graz, Austria
| | - Silke Tribius
- Asklepios Tumorzentrum Hamburg, AK St. Georg, Hermann-Holthusen Institute for Radiotherapy, Hamburg, Germany
| | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Department of Oncology and Hematology, Hamburg, Germany
| | - Georg Wurschi
- Department of Radiotherapy and Radiation Oncology, Jena University Hospital, Jena, Germany
| | - Pompiliu Piso
- Department of General Surgery, Krankenhaus Barmherzige Brüder Regensburg, Regensburg, Germany
| | - Tim Friede
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | | | - Eleni Gkika
- Department of Radiation Oncology, University Hospital Bonn, Bonn, Germany
| | - Claus Rödel
- Department of Radiation Oncology, University Hospital Johann Wolfgang Goethe University, Frankfurt, Germany
- Frankfurt Cancer Institute, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Emmanouil Fokas
- Department of Radiation Oncology, Cyberknife and Radiation Therapy, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Frankfurt Cancer Institute, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - on behalf of the German Rectal Cancer Study Group
- Department of Radiation Oncology, Cyberknife and Radiation Therapy, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Department of Radiation Oncology, University Hospital Johann Wolfgang Goethe University, Frankfurt, Germany
- Frankfurt Cancer Institute, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Mannheim Cancer Center, University Hospital Mannheim, Mannheim, Germany
- Department of Radiotherapy and Oncology, Community Hospital, Darmstadt, Germany
- Department of Radiation Oncology and Radiotherapy, DiaCura & Klinikum Coburg, Coburg, Germany
- Department of Radiation Therapy, University of Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, Freiburg, Germany
- German Cancer Research Center, German Cancer Consortium, Freiburg, Germany
- Department of Radiation Oncology, Comprehensive Cancer Center Lower Saxony (CCC-N), University Hospital Göttingen (UMG), Göttingen, Germany
- Department of Radiation Oncology, St. Vincentius-Kliniken gAG, Karlsruhe, Germany
- Kempten Clinic, Kempten, Germany
- Department of Radiation Therapy, University of Leipzig, Leipzig, Germany
- Department of Radiation Oncology and Radiation Therapy, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of General, Visceral and Pediatric Surgery, University Medical Center, Göttingen, Germany
- Department of Radiation Oncology, University Hospital Mannheim, Mannheim, Germany
- Department of Radiation Oncology, Kliniken Maria Hilf, Moenchengladbach, Germany
- Department of Radiation Oncology, Barmherzige Brüder Hospital Regensburg, Regensburg, Germany
- Department of Radiotherapy and Oncology, University of Rostock, Rostock, Germany
- Department of Radiotherapy and Oncology, Ostfildern Medius Kliniken, Ostfildern, Germany
- Department of Radiation Oncology, University of Würzburg, Würzburg, Germany
- Department of Visceral Surgery, University of Cologne, Cologne, Germany
- Department of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düesseldorf, University Hospital of Cologne, Cologne, Germany
- Department of Radiation Oncology, Medical University of Graz, Graz, Austria
- Asklepios Tumorzentrum Hamburg, AK St. Georg, Hermann-Holthusen Institute for Radiotherapy, Hamburg, Germany
- Asklepios Tumorzentrum Hamburg, AK Altona, Department of Oncology and Hematology, Hamburg, Germany
- Department of Radiotherapy and Radiation Oncology, Jena University Hospital, Jena, Germany
- Department of General Surgery, Krankenhaus Barmherzige Brüder Regensburg, Regensburg, Germany
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
- Department of Radiation Oncology, University Hospital Düsseldorf, Germany
- Department of Radiation Oncology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
17
|
Huang Y, Chen L, Chen Y, Zhou S, Xie X, Xie J, Yu M, Chen J. High-density lipoprotein-based nanoplatform reprograms tumor microenvironment and enhances chemotherapy against pancreatic ductal adenocarcinoma. Biomaterials 2025; 318:123147. [PMID: 39908877 DOI: 10.1016/j.biomaterials.2025.123147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/25/2024] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly aggressive, with limited success in traditional therapies due to the fibrotic, immunosuppressive, pro-metastatic tumor microenvironment (TME), which collectively impede the drug accumulation and accelerate the tumor progression. In this work, we developed a PDAC-customized nutrient-mimicking reconstituted high-density lipoprotein (rHDL) capable of efficiently co-encapsulate versatile TME regulating cannabidiol and cytotoxic gemcitabine to simultaneously reprogram TME while suppressing PDAC progression. Specifically, a small-sized, nutrient-like rHDL was constructed to realize deep PDAC parenchyma penetration and efficient intra-tumoral uptake. Next, natural herbal compound cannabidiol was screened and incorporated into rHDL to regulate TME via attenuating fibrosis, reliving immunosuppression and mitigating metastatic tendency. At last, gemcitabine, the PDAC gold standard first-line therapy was co-delivered by the PDAC-customized rHDL to overcome drug resistance and amplify its PDAC suppression. Our findings demonstrate the feasibility of an integrated multi-stage TME regulation strategy for improved PDAC therapy, and might represent a modality in promoting chemotherapy against PDAC.
Collapse
Affiliation(s)
- Yukun Huang
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China; Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liang Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Yu Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Songlei Zhou
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Xiaoying Xie
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Jing Xie
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Minghua Yu
- Fudan University Clinical Research Center for Cell-based Immunotherapy & Department of Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Shanghai, 201399, China
| | - Jun Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
18
|
Wang Q, Fan C, Tsujio G, Sakuma T, Maruo K, Yamamoto Y, Imanishi D, Kawabata K, Nishikubo H, Kanei S, Aoyama R, Kushiyama S, Ohira M, Yashiro M. Establishment and characterization of a new human gallbladder cancer cell line, OCUG-2. World J Exp Med 2025; 15:100443. [DOI: 10.5493/wjem.v15.i2.100443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/19/2024] [Accepted: 01/07/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Gallbladder cancer (GBC) is a highly aggressive malignant tumor originating from the biliary tract. As one of the most common malignancies in the biliary system, GBC is particularly challenging due to its tendency to remain asymptomatic, which often results in delayed diagnoses even at advanced stages. Combined with its invasive potential and poor response to conventional therapies, GBC has a high mortality rate, highlighting the critical need for innovative therapeutic approaches. Identifying molecular biomarkers for early detection and discovering novel therapeutic targets might be essential to improving outcomes of patients with GBC.
AIM To establish a novel GBC cell line to investigate the molecular mechanisms underlying GBC progression and evaluate potential therapeutic targets.
METHODS We developed a unique GBC cell line, named OCUG-2, derived from a metastatic peritoneal implant, and verified its authenticity using short tandem repeat (STR) profiling. RT-PCR and RNA sequencing (RNA-seq) were performed to assess gene expression profiles, with functional enrichment analyzed through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The MTT cell proliferation assay and an invasion assay were performed to evaluate response to nine inhibitors. Immunohistochemistry (IHC) was conducted on 34 GBC samples to analyze insulin-like growth factor 1 receptor (IGF1R) expression.
RESULTS OCUG-2 cells displayed adhesive growth with dendritic morphology and a 30-hour doubling time. Subcutaneous inoculation of OCUG-2 cells into mice confirmed their tumorigenic potential. STR analysis authenticated the cell line, and there was high mRNA and protein expression of IGF1R in OCUG-2 cells. The IGF1R inhibitor picropodophyllotoxin significantly inhibited OCUG-2 cell proliferation, yielding an IC50 of 0.49 μM. RNA-seq analysis identified gene fusions, and GO/KEGG functional enrichment analyses revealed pathways implicated in cancer progression. IHC analysis showed IGF1R positivity in 18 of 34 GBC cases, with significant association between IGF1R expression and poor prognosis. In invasion assays, an IGF1R inhibitor effectively reduced OCUG-2 cell invasiveness.
CONCLUSION IGF1R might be a promising target for GBC. The newly established OCUG-2 cell line serves as a valuable model for investigating the molecular mechanisms of GBC and evaluating therapeutic strategies.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Canfeng Fan
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Gen Tsujio
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Takashi Sakuma
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Koji Maruo
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Yurie Yamamoto
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Daiki Imanishi
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Kyoka Kawabata
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Hinano Nishikubo
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Saki Kanei
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Rika Aoyama
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Syuhei Kushiyama
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Masaichi Ohira
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-0051, Japan
| |
Collapse
|
19
|
Pascale MM, Frongillo F, Vasta P, Massimiani G, Nure E, Agnes S. Significance of the neutrophil-to-lymphocyte ratio and the platelet-to-lymphocyte ratio as prognostic predictors after liver transplantation. World J Transplant 2025; 15:100157. [DOI: 10.5500/wjt.v15.i2.100157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 12/20/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND The use of biomarkers, such as the neutrophil-to-lymphocyte ratio (NLR) and the neutrophil-to-platelet ratio (NPR), has shown promise in evaluating early outcomes after medical, interventional, and surgical treatments. NLR has emerged as an indicator of systemic inflammation and physiological stress. NPR has emerged as a potential indicator of inflammation and thrombotic risk in the context of surgical and radiological procedures.
AIM To analyze the correlation of NLR and NPR with the development of post-liver transplantation (LT) early complications after stratification for hepatocellular carcinoma diagnosis.
METHODS Consecutive patients undergone LT between January 2019 and December 2023 were enrolled. Data regarding the concentration of hemoglobin and the differential leukocyte count on postoperative days (POD) 0, 1, 3, and 5 were collected.
RESULTS The dataset included 161 consecutive patients undergone LT. Clavien-Dindo IV-V complications had a good correlation with NLR POD 1 (P = 0.05), NLR POD 3 (P < 0.001), NLR POD 7 (P < 0.001), NPR POD 3 (P < 0.001). In addition, the NPR ratio on POD 3 correlated with the onset of 30-day hemorrhage (P = 0.009). Finally, 30-day mortality had a significant association with the NLR POD 1 (P = 0.03) and with NLR POD 7 (P = 0.004), while NPR had a significant correlation with 30-day mortality in NPR POD 7 (P = 0.004).
CONCLUSION The analysis of NLR and NPR are strictly correlated with Clavien-Dindo IV-V complications and 30-day post-LT death.
Collapse
Affiliation(s)
- Marco Maria Pascale
- General Surgery and Liver Transplantation Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| | - Francesco Frongillo
- General Surgery and Liver Transplantation Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| | - Pierangelo Vasta
- Department of Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| | - Giuseppe Massimiani
- Department of Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| | - Erida Nure
- General Surgery and Liver Transplantation Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| | - Salvatore Agnes
- General Surgery and Liver Transplantation Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| |
Collapse
|
20
|
Wang Z, Su X, Zhan Z, Wang H, Zhou S, Mao J, Xu H, Duan S. miR-660: A novel regulator in human cancer pathogenesis and therapeutic implications. Gene 2025; 953:149434. [PMID: 40120868 DOI: 10.1016/j.gene.2025.149434] [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: 09/29/2024] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
MicroRNAs (miRNAs) are non-coding RNAs that regulate gene expression. Among these, miR-660, located on chromosome Xp11.23, is increasingly studied for its role in cancer due to its abnormal expression in various biological contexts. It is regulated by 8 competing endogenous RNAs (ceRNAs), which adds complexity to its function. miR- 660 targets 19 genes involved in 6 pathways such as PI3K/AKT/mTOR, STAT3, Wnt/β-catenin, p53, NF‑κB, and RAS, influencing cell cycle, proliferation, apoptosis, and invasion/migration. It also plays a role in resistance to chemotherapies like cisplatin, gemcitabine, and sorafenib in lung adenocarcinoma (LUAD), pancreatic ductal adenocarcinoma (PDAC), and hepatocellular carcinoma (HCC), thus highlighting its clinical importance. Additionally, leveraging liposomes as nanocarriers presents a promising avenue for enhancing cancer drug delivery. Our comprehensive study not only elucidates the aberrant expression patterns, biological functions, and regulatory networks of miR-660 and its ceRNAs but also delves into the intricate signaling pathways implicated. We envisage that our findings will furnish a robust framework and serve as a seminal reference for future investigations of miR-660, fostering advancements in cancer research and potentially catalyzing breakthroughs in cancer diagnosis and treatment paradigms.
Collapse
Affiliation(s)
- Zehua Wang
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Xinming Su
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhiqing Zhan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hangxuan Wang
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shuhan Zhou
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Jiasheng Mao
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hening Xu
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shiwei Duan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
21
|
Saadh MJ, Allela OQB, Kareem RA, Sanghvi G, Ballal S, Naidu KS, Bareja L, Chahar M, Gupta S, Sameer HN, Yaseen A, Athab ZH, Adil M. Exploring preventive and treatment strategies for oral cancer: Modulation of signaling pathways and microbiota by probiotics. Gene 2025; 952:149380. [PMID: 40089085 DOI: 10.1016/j.gene.2025.149380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/11/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
The evidence suggests that the microbiome plays a crucial role in cancer development. The oral cavity has many microorganisms that can influence oral cancer progression. Understanding the mechanisms and signaling pathways of the oral, gum, and teeth microbiome in tumor progression can lead to new treatment strategies. Probiotics, which are friendly microorganisms, have shown potential as anti-cancer agents. These positive characteristics of probiotic strains make them suitable for cancer prevention or treatment. The oral-gut microbiome axis supports health and homeostasis, and imbalances in the oral microbiome can disrupt immune signaling pathways, epithelial barriers, cell cycles, apoptosis, genomic stability, angiogenesis, and metabolic processes. Changes in the oral microbiome in oral cancer may suggest using probiotics-based treatments for their direct or indirect positive roles in cancer development, progression, and metastasis, specifically oral squamous cell carcinoma (OSCC). Here, reported relationships between probiotics, oral microbiota, and oral cancer are summarized.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003 Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401 Punjab, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Sofia Gupta
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307 Punjab, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy college, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
22
|
Jeong SH, Min JS, Kim YW, Yoon HM, An JY, Eom BW, Hur H, Lee YJ, Cho GS, Park YK, Jung MR, Park JH, Hyung WJ, Kook MC, Han M, Nam BH, Ryu KW. Clinicopathological Factors Affecting Stomach Preservation Following Laparoscopic Sentinel Node Navigation Surgery in Patients with Early Gastric Cancer: A Secondary Analysis of the Multicenter Randomized Phase III SENORITA Trial. Ann Surg Oncol 2025; 32:4280-4291. [PMID: 40117015 DOI: 10.1245/s10434-025-17114-1] [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: 12/18/2024] [Accepted: 02/18/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND The SENORITA phase III trial demonstrated the effectiveness of laparoscopic sentinel node navigation surgery (LSNNS) in preserving stomach function for patients with early gastric cancer (EGC), although some patients experienced surgical failure or recurrence. The purpose of this study was to analyze patients' clinicopathologic features from the SENORITA trial who were allocated to LSNNS with stomach-preserving surgery but ultimately did not preserve stomach or experienced recurrence. PATIENTS AND METHODS Patients were categorized into two groups: the failure group (stomach preservation failure or cancer recurrence after LSNNS) and the success group (stomach preservation without recurrence following LSNNS). This study analyzed the detailed clinicopathologic characteristics of patients in the failure group from the SENORITA trial. RESULTS Among 258 patients who underwent LSNNS, 193 patients (74.8%) achieved stomach preservation, while 65 patients (25.2%) failed to preserve. Intraoperative failure was the most common cause of unsuccessful stomach preservation, occurring in 35 of 65 cases (53.8%). Advanced pathological TNM stage was the only independent risk factor by multivariate analysis, with stage IB and IIA patients showing 5.9- and 45.0-fold higher failure risks. The main causes of failure included sentinel basin detection failure, metastatic lymph nodes, positive tumors at resection margins, and complications. The failure group also included five cases of gastric cancer recurrence following LSNNS. CONCLUSION Accurate preoperative staging and patient selection are crucial for optimizing LSNNS outcomes. Ensuring precise resection with an adequate number of harvested sentinel basin nodes is essential to succeed the stomach-preserving surgery.
Collapse
Affiliation(s)
- Sang-Ho Jeong
- Department of Surgery, Gyeongsang National University College of Medicine and Gyeongsang National University Changwon Hospital, Changwon, Korea
| | - Jae-Seok Min
- Division of Foregut Surgery, Korea University Anam Hospital, Seoul, and Department of Surgery, Korea University College of Medicine, Seoul, Korea.
- Department of Surgery, Dongnam Institute of Radiological and Medical Sciences, Cancer Center, Busan, Korea.
| | - Young-Woo Kim
- Center for Gastric Cancer, Research Institute & Hospital, National Cancer Center, Goyang, Korea
- Department of Cancer Policy and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Korea
| | - Hong Man Yoon
- Center for Gastric Cancer, Research Institute & Hospital, National Cancer Center, Goyang, Korea
| | - Ji Yeong An
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Bang Wool Eom
- Center for Gastric Cancer, Research Institute & Hospital, National Cancer Center, Goyang, Korea
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, Korea
| | - Young Joon Lee
- Department of Surgery, Gyeongsang National University College of Medicine and Gyeongsang National University Hospital, Jinju, Korea
| | - Gyu Seok Cho
- Department of Surgery, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Young-Kyu Park
- Department of Surgery, Chonnam National University Medical School, Gwangju, Korea
| | - Mi Ran Jung
- Department of Surgery, Chonnam National University Medical School, Gwangju, Korea
| | - Ji-Ho Park
- Department of Surgery, Gyeongsang National University College of Medicine and Gyeongsang National University Hospital, Jinju, Korea
| | - Woo Jin Hyung
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Myeong-Cherl Kook
- Center for Gastric Cancer, Research Institute & Hospital, National Cancer Center, Goyang, Korea
| | - Mira Han
- Biostatistics Collaboration Team, National Cancer Center, Goyang, Korea
- Medical Research Collaborating Center, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Byung-Ho Nam
- Department of Cancer Control and Policy, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
- Clinical Design Research Center, HERINGS The Institution of Advanced Clinical and Biomedical Research, Seoul, Korea
| | - Keun Won Ryu
- Center for Gastric Cancer, Research Institute & Hospital, National Cancer Center, Goyang, Korea
| |
Collapse
|
23
|
Li YF, Yao LQ, Li C, Ren H, Gong JB, Wu H, Gu LH, Liang YJ, Yang YZ, Lin KY, Li ZQ, Zheng QX, Chen TH, Zhou YH, Wang H, Guo HW, Xu JH, Chen Z, Shen F, Wang MD, Yang T. Statistical Cure After Hepatectomy for Hepatitis B Virus-Associated Hepatocellular Carcinoma: A Risk-Stratification Model. Ann Surg Oncol 2025; 32:4396-4407. [PMID: 40188279 DOI: 10.1245/s10434-025-17176-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/27/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Statistical cure, defined as achieving life expectancy comparable with that of disease-free individuals, has not been specifically investigated in hepatitis B virus-associated hepatocellular carcinoma (HBV-HCC), which accounts for more than 50% of the global HCC burden. This study aimed to develop a cure model for HBV-HCC after hepatectomy using matched HBV carriers and the general population as reference groups. METHODS From a Chinese multicenter database, HBV-HCC patients who underwent curative-intent hepatectomy were retrospectively reviewed. Independent prognostic factors were identified through Cox regression. A spline-based cure model was applied using two reference populations: matched Chinese HBV carriers (from Shanghai Center for Disease Control and Prevention) and the general population (from the National Bureau of Statistics). RESULTS The study analyzed 740 HBV-HCC patients. The following eight independent risk factors were identified: preoperative high viral load (hazard ratio [HR] 1.27), Child-Pugh grade (HR 1.21 and 1.43), multiple tumors (HR 1.70), tumor size greater than 5.0 cm (HR 1.47), macrovascular invasion (HR 3.33), microvascular invasion (HR 1.25), intraoperative blood transfusion (HR 1.21), and postoperative HBV reactivation (HR 1.89). The overall cure probability was 21.2% versus that for HBV carriers and 11.1% versus that for the general population. Risk stratification identified distinct groups relative to HBV carriers. Low risk (64.2%) showed an initial cure rate of 30.3% and achieved a 95% cure probability by 8.6 years, whereas high risk (10.5%) showed negligible cure probability. CONCLUSIONS This first HBV-HCC-specific cure model demonstrated that statistical cure is achievable for a subset of patients after hepatectomy. Risk stratification identifies patients with varying cure probabilities, providing valuable guidance for personalized treatment strategies and surveillance protocols.
Collapse
Affiliation(s)
- Yi-Fan Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Lan-Qing Yao
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Chao Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Hong Ren
- Department of Viral Hepatitis Control and Prevention, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Jin-Bo Gong
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Han Wu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Li-Hui Gu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Ying-Jian Liang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu-Ze Yang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, China
| | - Kong-Ying Lin
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fujian, China
| | - Zi-Qiang Li
- Department of Liver Transplantation and Hepatic Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Qi-Xuan Zheng
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ting-Hao Chen
- Department of General Surgery, Ziyang First People's Hospital, Ziyang, China
| | - Ya-Hao Zhou
- Department of Hepatobiliary Surgery, Pu'er People's Hospital, Pu'er, China
| | - Hong Wang
- Department of General Surgery, Liuyang People's Hospital, Liuyang, China
| | - Hong-Wei Guo
- The 2nd Department of General Surgery, The Second People's Hospital of Changzhi, Changzhi, China
| | - Jia-Hao Xu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Zhong Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Ming-Da Wang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
| | - Tian Yang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
24
|
Murray K, Oldfield L, Stefanova I, Gentiluomo M, Aretini P, O'Sullivan R, Greenhalf W, Paiella S, Aoki MN, Pastore A, Birch-Ford J, Rao BH, Uysal-Onganer P, Walsh CM, Hanna GB, Narang J, Sharma P, Campa D, Rizzato C, Turtoi A, Sever EA, Felici A, Sucularli C, Peduzzi G, Öz E, Sezerman OU, Van der Meer R, Thompson N, Costello E. Biomarkers, omics and artificial intelligence for early detection of pancreatic cancer. Semin Cancer Biol 2025; 111:76-88. [PMID: 39986585 DOI: 10.1016/j.semcancer.2025.02.009] [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: 11/29/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is frequently diagnosed in its late stages when treatment options are limited. Unlike other common cancers, there are no population-wide screening programmes for PDAC. Thus, early disease detection, although urgently needed, remains elusive. Individuals in certain high-risk groups are, however, offered screening or surveillance. Here we explore advances in understanding high-risk groups for PDAC and efforts to implement biomarker-driven detection of PDAC in these groups. We review current approaches to early detection biomarker development and the use of artificial intelligence as applied to electronic health records (EHRs) and social media. Finally, we address the cost-effectiveness of applying biomarker strategies for early detection of PDAC.
Collapse
Affiliation(s)
- Kate Murray
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Lucy Oldfield
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Irena Stefanova
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Rachel O'Sullivan
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - William Greenhalf
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Salvatore Paiella
- Pancreatic Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Italy
| | - Mateus N Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Brazil
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, Scuola Normale Superiore di Pisa, Italy
| | - James Birch-Ford
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Bhavana Hemantha Rao
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| | - Pinar Uysal-Onganer
- School of Life Sciences, Cancer Mechanisms and Biomarkers Group, The University of Westminster, United Kingdom
| | - Caoimhe M Walsh
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | | | | | | | | | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Treatment Lab, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, France
| | - Elif Arik Sever
- Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | | | - Elif Öz
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | - Osman Uğur Sezerman
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
25
|
Zhang J, Ma L, He L, Xu Q, Ding Y, Wang L. MicroRNA-541-3p/Rac2 signaling bridges radiation-induced lung injury and repair. Noncoding RNA Res 2025; 12:10-19. [PMID: 40026446 PMCID: PMC11869541 DOI: 10.1016/j.ncrna.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 03/05/2025] Open
Abstract
Background While radiation-induced lung injury decreases quality of life and suppresses efficacy of radiotherapy, to date, the relationship between radiation-induced lung injury and repair remains unclear. Our previous studies revealed that TNFRSF10B-RIPK1/RIPK3-MLKL signaling induces necroptosis of alveolar epithelial cells and potentiates radiation-induced lung injury. We also found that microRNA-541-3p is differentially expressed in radiation-damaged lungs. The connection between microRNA-541-3p, TNFRSF10B signaling, and TGFβ1 signaling is also unclear. Objective This study was performed to explore the regulatory effects of microRNA-541-3p on TNFRSF10B and TGFβ1 signaling. Methods Mouse alveolar epithelial cells were transfected with a vector expressing microRNA-541-3p to regulate expression of target genes. Flow cytometry, polymerase chain reaction, and western blotting were used to analyze cell necroptosis, target gene expression, and target protein expression, respectively. Results Overexpression of microRNA-541-3p positively regulated TNFRSF10B-RIPK1/RIPK3-MLKL signaling through Rac2 to induce cell necroptosis. MicroRNA-541-3p negatively regulates Rac2. MicroRNA-541-3p and Rac2 regulate the expression of Tgf-beta1 and its encoded proteins. Conclusions The Rac2 gene synchronously regulates TNFRSF10B-RIPK1/RIPK3-MLKL and TGFβ1 signaling. MicroRNA-541-3P/Rac2 act as mediators of radiation damage and repair signaling.
Collapse
Affiliation(s)
- Jiandong Zhang
- Clinical School of Medicine, Henan University of Science and Technology, Luoyang City, Henan Province, 471023, China
- The First Affiliated Hospital of Henan University of Science and Technology, Luoyang City, Henan Province, 471023, China
| | - Lei Ma
- Department of Radiation Oncology, Nanyang First People's Hospital Affiliated to Henan University, Nanyang City, Henan Province, 473000, China
| | - Limin He
- Department of Radiation Oncology, Nanyang First People's Hospital Affiliated to Henan University, Nanyang City, Henan Province, 473000, China
| | - Quanxiao Xu
- Department of Oncology, the Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, 215000, China
| | - Yan Ding
- Department of Radiation Oncology, Nanyang First People's Hospital Affiliated to Henan University, Nanyang City, Henan Province, 473000, China
| | - Lidong Wang
- Clinical School of Medicine, Henan University of Science and Technology, Luoyang City, Henan Province, 471023, China
- The First Affiliated Hospital of Henan University of Science and Technology, Luoyang City, Henan Province, 471023, China
| |
Collapse
|
26
|
Lin A, Song L, Wang Y, Yan K, Tang H. Future prospects of deep learning in esophageal cancer diagnosis and clinical decision support (Review). Oncol Lett 2025; 29:293. [PMID: 40271007 PMCID: PMC12016012 DOI: 10.3892/ol.2025.15039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Esophageal cancer (EC) is one of the leading causes of cancer-related mortality worldwide, still faces significant challenges in early diagnosis and prognosis. Early EC lesions often present subtle symptoms and current diagnostic methods are limited in accuracy due to tumor heterogeneity, lesion morphology and variable image quality. These limitations are particularly prominent in the early detection of precancerous lesions such as Barrett's esophagus. Traditional diagnostic approaches, such as endoscopic examination, pathological analysis and computed tomography, require improvements in diagnostic precision and staging accuracy. Deep learning (DL), a key branch of artificial intelligence, shows great promise in improving the detection of early EC lesions, distinguishing benign from malignant lesions and aiding cancer staging and prognosis. However, challenges remain, including image quality variability, insufficient data annotation and limited generalization. The present review summarized recent advances in the application of DL to medical images obtained through various imaging techniques for the diagnosis of EC at different stages. It assesses the role of DL in tumor pathology, prognosis prediction and clinical decision support, highlighting its advantages in EC diagnosis and prognosis evaluation. Finally, it provided an objective analysis of the challenges currently facing the field and prospects for future applications.
Collapse
Affiliation(s)
- Aiting Lin
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R. China
- Department of Thoracic Surgery, The Second Affiliated Hospital of Naval Medical University, Shanghai 200003, P.R. China
| | - Lirong Song
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R. China
| | - Ying Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R. China
| | - Kai Yan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, P.R. China
| | - Hua Tang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Naval Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
27
|
Bhattacharya S, Beaty W, Faye AS, Axelrad JE. Right-Sided Dysplasia in Inflammatory Bowel Disease Is Not Associated with Conventional Risk Factors for Neoplasia. GASTROENTEROLOGY INSIGHTS 2025; 16:14. [PMID: 40242653 PMCID: PMC11999668 DOI: 10.3390/gastroent16020014] [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] [Indexed: 04/18/2025] Open
Abstract
Introduction In the general population, right I-sided dysplasia presents a higher risk for colorectal cancer (CRC) and metachronous dysplasia compared to left (L)-sided dysplasia. Given that patients with inflammatory bowel disease (IBD) are at higher risk for dysplasia than the general population, we sought to assess the risk factors as well as the differences in outcomes between patients with R-sided, L-sided, and both R- and L-sided dysplasia. Methods A retrospective chart review was performed on patients at NYU Langone Health who had evidence of dysplasia on a colonoscopy between 2011 and 2021. Demographics and pertinent medical history were compiled. Cohorts were based on the dysplasia location (R-sided, L-sided, or R- and L-sided) and the IBD-related outcomes were analyzed. Results A total of 71 patients had colonic dysplasia. The mean age was 54 years old (SD ± 17). The majority were male (72%), white (69%), and non-Hispanic (94%). A total of 76% had ulcerative colitis (UC) and 24% had Crohn's disease (CD). Of all dysplastic lesions, 57 (80%) patients had unifocal disease and the remainder had multifocal disease. A total of 39 (55%) patients had R-sided dysplasia, 24 (34%) had L-sided dysplasia, and 8 (11%) had both R- and L-sided dysplasia. Patients with UC were more likely to have L-sided dysplasia (92% vs. 8% in CD; p = 0.04). Pseudopolyps were more likely associated with R- and L-sided dysplasia (38% in R- and L-sided dysplasia, 10% in R-sided dysplasia, and 4% in L-sided dysplasia; p = 0.03). Conclusions Patients with UC had a higher risk for L-sided colonic dysplasia compared to patients with CD; however, there were no differences in the progression of dysplasia between those who had R-sided and those who had L-sided dysplasia. Larger studies are needed to assess the risk factors and outcomes related to the laterality of dysplasia and further validate these findings among patients with IBD.
Collapse
Affiliation(s)
- Sumona Bhattacharya
- Division of Gastroenterology, Department of Medicine, Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - William Beaty
- Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Adam S. Faye
- Division of Gastroenterology, Department of Medicine, Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Jordan E. Axelrad
- Division of Gastroenterology, Department of Medicine, Grossman School of Medicine, New York University, New York, NY 10016, USA
| |
Collapse
|
28
|
Abudalo R, Alqudah A, Alnajjar R, Abudalo R, Abuqamar A, Oqal M, Qnais E. KRAS/NRAS/BRAF mutational profile and association with clinicopathological characteristics in patients with metastatic colorectal cancer. Oncol Lett 2025; 29:312. [PMID: 40342724 PMCID: PMC12059616 DOI: 10.3892/ol.2025.15058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/26/2025] [Indexed: 05/11/2025] Open
Abstract
Colorectal cancer (CRC) is increasingly prevalent in Jordan and poses a significant public health challenge. The presence of Kirsten rat sarcoma viral oncogene homolog (KRAS), neuroblastoma RAS viral oncogene homolog (NRAS) and v-Raf Murine Sarcoma Viral Oncogene Homolog B (BRAF) mutations is key in CRC diagnostics, as these mutations are associated with resistance to monoclonal antibodies targeting the epidermal growth factor receptor. The present study aimed to identify these mutations in patients with CRC and assess their associations with clinicopathological characteristics. A retrospective analysis was conducted using data from 262 patients with metastatic CRC (mCRC) at the Jordanian Military Cancer Center-Royal Medical Services (Amman, Jordan). Variables such as age, sex, tumor differentiation and the mutational status of KRAS, NRAS and BRAF, along with tumor location, were analyzed statistically to explore associations between mutations and tumor characteristics. Among the included patients, 48.5% had KRAS mutations, 3.8% had NRAS mutations and 0.8% had BRAF mutations. The majority of KRAS mutations were in exon 2 at codons 12 and 13, with the highest mutational rate at 45.8%. In the univariate model, NRAS mutations were significantly associated with moderately differentiated tumors and the multivariate hierarchical regression analysis established that KRAS mutations were significantly associated with histological subtypes [mucinous adenocarcinoma, tubular adenocarcinoma, signet adenocarcinoma and adenocarcinoma (not specified)]. These results highlighted the molecular profiles and clinicopathological characteristics of patients with mCRC, which demonstrated the associations between mutational status and the varying clinicopathological aspects based on the type of RAS mutation. Thus, these specific traits (patient's age, sex, CRC site, histological subtypes and tumor grade) may be taken into account when evaluating the predictive significance of RAS and BRAF status in CRC and tailored treatment strategies.
Collapse
Affiliation(s)
- Rawan Abudalo
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Roaa Alnajjar
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Jordan University, Amman 11942, Jordan
| | - Razan Abudalo
- Department of Radiology, Jordanian Royal Medical Services, Amman 855122, Jordan
| | - Ayman Abuqamar
- Department of Oncology and Hematology, Jordanian Royal Medical Services, Amman 855122, Jordan
| | - Muna Oqal
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Esam Qnais
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa 13133, Jordan
| |
Collapse
|
29
|
Han MQ, Zhao YJ, Pang S, Zhu HJ, Luo DQ, Liu YF, Yang K, Cao F. Modulating culture method promotes the production of disulfide-linked resorcylic acid lactone dimers with anti-proliferative activity. Bioorg Chem 2025; 159:108418. [PMID: 40168886 DOI: 10.1016/j.bioorg.2025.108418] [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: 02/13/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
Sulfur-containing natural products are distinguished by their unique chemical structures and notable biological activities, rendering them highly valuable in drug discovery and development. Recent advancements in chemical epigenetic modifications, sulfur source regulation, and fungal co-cultivation have significantly facilitated the discovery of novel sulfur-containing compounds. In this study, the modulating culture method, incorporating DMSO and sea salt into the culture medium, was utilized to induce the marine-derived fungus Penicillium sp. to produce novel disulfide-linked resorcylic acid lactone dimers, dipenirestone A and B (1 and 2), along with their monomeric precursors (3-13). The absolute configurations of the new compounds 1-6 were elucidated through calculated NMR and ECD methods, as well as X-ray crystallography. Notably, the dimeric compounds (1 and 2) exhibited significantly enhanced anti-proliferative activity against HGC-27 cells compared to the monomers 3-13. It was revealed that compounds 1 and 2 exerted an antiproliferative effect through the modulation of the PI3K/AKT/mTOR signaling pathway. This was manifested as cell cycle arrest in the G1 phase, reduction in mitochondrial membrane potential, and induction of apoptosis.
Collapse
Affiliation(s)
- Ming-Qian Han
- College of Pharmaceutical Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnostics of Education Ministry of China, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China
| | - Ying-Jie Zhao
- College of Pharmaceutical Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnostics of Education Ministry of China, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China
| | - Sen Pang
- Huanghe Science & Technology College, Zhengzhou 450005, China
| | - Hua-Jie Zhu
- School of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Du-Qiang Luo
- College of Life Sciences, Hebei University, Baoding 071002, China
| | - Yun-Feng Liu
- College of Life Sciences, Hebei University, Baoding 071002, China.
| | - Kan Yang
- College of Pharmaceutical Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnostics of Education Ministry of China, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China.
| | - Fei Cao
- College of Pharmaceutical Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnostics of Education Ministry of China, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China.
| |
Collapse
|
30
|
Hu Y, Tuo B. The function of chloride channels in digestive system disease (Review). Int J Mol Med 2025; 55:99. [PMID: 40314091 PMCID: PMC12045473 DOI: 10.3892/ijmm.2025.5540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/11/2025] [Indexed: 05/03/2025] Open
Abstract
Cation channels have been extensively studied in the context of digestive disorders, but comparatively little attention has been given to anions and their associated channels. Chloride ions, the most abundant anions in the human body, act as signaling molecules, modulating cellular behavior and playing a key role in regulating multiorgan physiological and pathophysiological mechanisms. The intra‑ and extracellular distributions of chloride ions are primarily controlled by various chloride channels and transporters. Currently, these chloride channels are classified into several groups: The chloride channels family, cystic fibrosis transmembrane conductance regulator, calcium‑activated chloride channels, volume‑regulated anion channels, proton‑activated chloride channels and ligand‑gated anion channels. This review aims to summarize the roles of chloride ion channels and transporter proteins in digestive system diseases, providing a theoretical basis for future research and offering potential new strategies for disease treatment.
Collapse
Affiliation(s)
- Yanxia Hu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
31
|
Ahn S, Kaipparettu BA. G-protein coupled receptors in metabolic reprogramming and cancer. Pharmacol Ther 2025; 270:108849. [PMID: 40204142 DOI: 10.1016/j.pharmthera.2025.108849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/09/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
G-protein coupled receptors (GPCR) are one of the frequently investigated drug targets. GPCRs are involved in many human pathophysiologies that lead to various disease conditions, such as cancer, diabetes, and obesity. GPCR receptor activates multiple signaling pathways depending on the ligand and tissue type. However, this review will be limited to the GPCR-mediated metabolic modulations and the activation of relevant signaling pathways in cancer therapy. Cancer cells often have reprogrammed cell metabolism to support tumor growth and metastatic plasticity. Many aggressive cancer cells maintain a hybrid metabolic status, using both glycolysis and mitochondrial metabolism for better metabolic plasticity. In addition to glucose and glutamine pathways, fatty acid is a key mitochondrial energy source in some cancer subtypes. Recently, targeting alternative energy pathways like fatty acid beta-oxidation (FAO) has attracted great interest in cancer therapy. Several in vitro and in vivo experiments in different cancer models reported encouraging responses to FAO inhibitors. However, due to the potential liver toxicity of FAO inhibitors in clinical trials, new approaches to indirectly target metabolic reprogramming are necessary for in vivo targeting of cancer cells. This review specifically focused on free fatty acid receptors (FFAR) and β-adrenergic receptors (β-AR) because of their reported significance in mitochondrial metabolism and cancer. Further understanding the pharmacology of GPCRs and their role in cancer metabolism will help repurpose GPCR-targeting drugs for cancer therapy and develop novel drug discovery strategies to combine them with standard cancer therapy to increase anticancer potential and overcome drug resistance.
Collapse
Affiliation(s)
- Songyeon Ahn
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Benny Abraham Kaipparettu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
32
|
Jain A, Mishra AK, Hurkat P, Shilpi S, Mody N, Jain SK. Navigating liver cancer: Precision targeting for enhanced treatment outcomes. Drug Deliv Transl Res 2025; 15:1935-1961. [PMID: 39847205 DOI: 10.1007/s13346-024-01780-x] [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: 12/18/2024] [Indexed: 01/24/2025]
Abstract
Cancer treatments such as surgery and chemotherapy have several limitations, including ineffectiveness against large or persistent tumors, high relapse rates, drug toxicity, and non-specificity of therapy. Researchers are exploring advanced strategies for treating this life-threatening disease to address these challenges. One promising approach is targeted drug delivery using prodrugs or surface modification with receptor-specific moieties for active or passive targeting. While various drug delivery systems have shown potential for reaching hepatic cells, nano-carriers offer significant size, distribution, and targetability advantages. Engineered nanocarriers can be customized to achieve effective and safe targeting of tumors by manipulating physical characteristics such as particle size or attaching receptor-specific ligands. This method is particularly advantageous in treating liver cancer by targeting specific hepatocyte receptors and enzymatic pathways for both passive and active therapeutic strategies. It highlights the epidemiology of liver cancer and provides an in-depth analysis of the various targeting approaches, including prodrugs, liposomes, magneto-liposomes, micelles, glycol-dendrimers, magnetic nanoparticles, chylomicron-based emulsion, and quantum dots surface modification with receptor-specific moieties. The insights from this review can be immensely significant for preclinical and clinical researchers working towards developing effective treatments for liver cancer. By utilizing these novel strategies, we can overcome the limitations of conventional therapies and offer better outcomes for liver cancer patients.
Collapse
Affiliation(s)
- Ankit Jain
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, 333031, India.
| | - Ashwini Kumar Mishra
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
- Central Ayurveda Research Institute, Jhansi, Uttar Pradesh, 284003, India
| | - Pooja Hurkat
- Dr. Hari Singh Gour Central University, Sagar, 470003, MP, India
| | - Satish Shilpi
- School of Pharmaceuticals and Population Health Informatics, FOP, DIT University, Dehradun, Uttarakahnad, India
| | - Nishi Mody
- Dr. Hari Singh Gour Central University, Sagar, 470003, MP, India
| | | |
Collapse
|
33
|
Kim S, Kim S, Ko C, Lee W, Kim HD. A microfluidic electrochemical immunosensor for detection of CEA and Ki67 in 3D tumor spheroids. Mater Today Bio 2025; 32:101768. [PMID: 40290895 PMCID: PMC12022681 DOI: 10.1016/j.mtbio.2025.101768] [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: 02/04/2025] [Revised: 03/27/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
Microfluidic chip-based electrochemical sensors have been developed to detect cancer biomarkers and monitor changes in the tumor microenvironment. However, the limitation of detecting only a single biomarker restricts their utility as accurate diagnostic tools. Simultaneous detection of multiple tumor biomarkers is important for early diagnosis of cancer. In this work, we report the development of a microfluidic-based electrochemical immunosensor platform capable of simultaneously observing multiple biomarkers expressed by three dimensions (3D) cell spheroids. The sensor platform employs a nanocomposite electrode material consisting of gold nanoparticles and carbon nanotubes, which enables sensitive and selective detection. The sensor was fabricated using 3D and printed circuit boards (PCB) printing techniques, demonstrating the feasibility of cost-effective manufacturing. The developed platform was able to quantitatively detect two key cancer biomarkers, carcinoembryonic antigen (CEA) and Ki67, with limits of detection of 0.97 ng/mL for each. Furthermore, the sensor was successfully utilized to observe the knockdown of these biomarkers, showcasing its potential for both diagnostic and therapeutic monitoring applications. These results suggest that the presented electrochemical sensor platform provides a promising lab-on-a-chip technology for comprehensive 3D cell spheroid-based cancer biomarker analysis, which could have significant implications for future clinical diagnostics and personalized medicine.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Seonyeop Kim
- Department of IT-Energy Convergence (BK21 Four), Chemical Industry Institute, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Chanjin Ko
- Department of IT-Energy Convergence (BK21 Four), Chemical Industry Institute, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Wonseok Lee
- Department of IT-Energy Convergence (BK21 Four), Chemical Industry Institute, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
- Department of Electrical Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Hwan Drew Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| |
Collapse
|
34
|
Glushko T, Costello J, Chima R, McGettigan M, Kim R, Jeong D, Qayyum A. Molecular signatures of intrahepatic cholangiocarcinoma: role in targeted therapy selection. Eur J Radiol 2025; 187:112056. [PMID: 40222184 DOI: 10.1016/j.ejrad.2025.112056] [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: 09/21/2024] [Revised: 02/08/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025]
Abstract
Cholangiocarcinoma is a highly lethal disease with a 5-year overall survival rate of 7-20%. A minority of patients present with resectable disease, and relapse rates remain high. Emerging data from next generation sequencing analysis have identified various actionable mutations which drive the different disease courses opening door to precision medicine and targeted therapies. This review focuses on the clinical significance of genetic alterations as well as the role of systemic therapies, immunotherapy and targeted therapies for intrahepatic cholangiocarcinoma.
Collapse
Affiliation(s)
- Tetiana Glushko
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States.
| | - James Costello
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| | - Ranjit Chima
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| | - Melissa McGettigan
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| | - Richard Kim
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| | - Daniel Jeong
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| | - Aliya Qayyum
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| |
Collapse
|
35
|
Crecca E, Di Giuseppe G, Camplone C, Vigiano Benedetti V, Melaiu O, Mezza T, Cencioni C, Spallotta F. The multifaceted role of agents counteracting metabolic syndrome: A new hope for gastrointestinal cancer therapy. Pharmacol Ther 2025; 270:108847. [PMID: 40216262 DOI: 10.1016/j.pharmthera.2025.108847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/27/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
Metabolic syndrome (MetS) is defined by the presence of at least three of five clinical parameters including abdominal obesity, insulin resistance, elevated triglycerides, reduced high-density lipoprotein (HDL) and hypertension. Major features describing MetS have been recognized risk factors for cancer onset, with an alarming impact on gastrointestinal (GI) tumors. Intriguingly, therapeutic administration of drugs to improve glycemic control and dyslipidemia (including metformin, statins) has been shown to have a preventive role in the development and in prognosis improvement of several cancer types. Overall, these observations highlight the key role of altered metabolism prevalently in cancer risk development and unveil anti-MetS agent repurposing potential beyond their conventional pharmacological action. The objective of this review is to summarize the current knowledge about the antitumor activity of anti-diabetic and anti-lipemic agents in GI cancer onset and progression. Here, pre-clinical evidence of their therapeutic potential and of their integration in novel compelling therapeutic strategies will be discussed. Possible clinical outcomes of these novel therapeutic combined protocols specifically dedicated to GI cancer patients will be put under the spotlight. In the future, these novel therapeutic options should be considered to improve conventional chemotherapy response and prognosis of this group of patients.
Collapse
Affiliation(s)
- Elena Crecca
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185 Rome, Italy
| | - Gianfranco Di Giuseppe
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy; Department of Translational Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Claudia Camplone
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | | | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Teresa Mezza
- Department of Translational Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy; Pancreas Unit, CEMAD Digestive Diseases Center, Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Chiara Cencioni
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185 Rome, Italy.
| | - Francesco Spallotta
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy.
| |
Collapse
|
36
|
Zhang L, Yuan J, Yao S, Wen G, An J, Jin H, Tuo B. Role of m5C methylation in digestive system tumors (Review). Mol Med Rep 2025; 31:142. [PMID: 40183387 PMCID: PMC11979572 DOI: 10.3892/mmr.2025.13507] [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: 11/26/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Currently, the incidence of digestive system tumors has been increasing annually, thus becoming a prevalent cause of cancer‑related mortalities. Although significant strides have been made in targeting the molecular mechanisms that underpin the development of these tumors, their treatment and prognosis still pose substantial challenges. This is primarily due to the ambiguity of early diagnostic indicators and the fact that most digestive system tumors are detected at an advanced stage. However, epigenetic modifications are capable of altering the expression of oncogenes and regulating biological processes in cancer. In recent years, the study of methylation in relation to tumor pathogenesis has become a focus of prominent research. Among the various types of methylation, 5‑methylcytosine (m5C) methylation plays a crucial role in the development of digestive system tumors and is anticipated to serve as a novel therapeutic target. However, to date, a comprehensive and systematic review concerning the role of m5C methylation in digestive system tumors is lacking. Consequently, the present study reviewed the role of m5C methylation in digestive system tumors such as esophageal cancer, gastric cancer and hepatocellular carcinoma, with the aim of providing a valuable reference for future research endeavors.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianbo Yuan
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
37
|
Giovarelli M, Mocciaro E, Carnovale C, Cervia D, Perrotta C, Clementi E. Immunosenescence in skeletal muscle: The role-play in cancer cachexia chessboard. Semin Cancer Biol 2025; 111:48-59. [PMID: 40020976 DOI: 10.1016/j.semcancer.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
With the increase in life expectancy, age-related conditions and diseases have become a widespread and relevant social burden. Among these, immunosenescence and cancer cachexia play a significant often intertwined role. Immunosenescence is the progressive aging decline of both the innate and adaptive immune systems leading to increased infection susceptibility, poor vaccination efficacy, autoimmune disease, and malignancies. Cancer cachexia affects elderly patients with cancer causing severe weight loss, muscle wasting, inflammation, and reduced response to therapies. Whereas the connections between immunosenescence and cancer cachexia have been raising attention, the molecular mechanisms still need to be completely elucidated. This review aims at providing the current knowledge about the interplay between immunosenescence, skeletal muscle, and cancer cachexia, analyzing the molecular pathways known so far to be involved. Finally, we highlight potential therapeutic strategies suited for elderly population aimed to block immunosenescence and to preserve muscle mass in cachexia, also presenting the analysis of the current state-of-the-art of related clinical trials.
Collapse
Affiliation(s)
- Matteo Giovarelli
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy.
| | - Emanuele Mocciaro
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Carla Carnovale
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Viterbo 01100, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy.
| |
Collapse
|
38
|
Donati F, Cervelli R, Boraschi P. Rare pancreatic cystic neoplasms: A pictorial review. Eur J Radiol Open 2025; 14:100620. [PMID: 39811581 PMCID: PMC11730956 DOI: 10.1016/j.ejro.2024.100620] [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: 09/23/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 01/05/2025] Open
Abstract
Since rare pancreatic cystic tumors may differ from common pancreatic cystic neoplasms in terms of treatment plan and prognosis, the differential diagnosis of these diseases is clinically relevant. Various imaging tests play an important role in the differential diagnosis of rare cystic pancreatic tumors, but accurately distinguishing these diseases solely on the basis of imaging findings is challenging. The purpose of this pictorial review is to present CT and in particular MR imaging features of rare pancreatic cystic tumors and discuss potential elements for differential diagnosis.
Collapse
Affiliation(s)
- Francescamaria Donati
- Department of Radiological Nuclear and Laboratory Medicine - Pisa University Hospital, Via Paradisa 2, Pisa 56124, Italy
| | - Rosa Cervelli
- Department of Radiological Nuclear and Laboratory Medicine - Pisa University Hospital, Via Paradisa 2, Pisa 56124, Italy
| | - Piero Boraschi
- Department of Radiological Nuclear and Laboratory Medicine - Pisa University Hospital, Via Paradisa 2, Pisa 56124, Italy
| |
Collapse
|
39
|
Ma J, Liao L, Du X. Discussion on surgical approaches for giant, well‑differentiated liposarcomas of the esophagus: Report of two cases. Oncol Lett 2025; 29:294. [PMID: 40271006 PMCID: PMC12016007 DOI: 10.3892/ol.2025.15040] [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: 01/23/2025] [Accepted: 03/13/2025] [Indexed: 04/25/2025] Open
Abstract
Giant liposarcoma of the esophagus is an exceedingly rare esophageal tumor with complex treatment options. This study investigated the treatment modalities and reported on the clinical outcomes of two cases involving giant, well-differentiated liposarcoma of the esophagus, providing a reference for the management of similar cases. Both tumors measured >20 cm in length and had diameters exceeding 4 cm; one case exhibited a lobulated appearance with visibly expanded blood vessels on its surface. Following discussions within a multidisciplinary team, under general anesthesia with tracheal intubation, the endoscopy team conducted endoscopic submucosal dissection, collaborating with the thoracic surgery team to manage potential bleeding risks. Both patients successfully underwent endoscopic tumor removal with postoperative pathology confirming the presence of well-differentiated liposarcoma and no observed complications. For patients with giant and complex well-differentiated liposarcomas, endoscopic dissection in conjunction with multidisciplinary collaboration represents a safe and effective treatment option, ensuring complete tumor removal while minimizing surgical trauma and enhancing patient prognosis.
Collapse
Affiliation(s)
- Jian Ma
- Department of Thoracic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Lihang Liao
- Department of Thoracic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Xiaojun Du
- Department of Thoracic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
40
|
Li D, Wang J, Li X, Wang Z, Yu Q, Koh SB, Wu R, Ye L, Guo Y, Okoli U, Pati-Alam A, Mota E, Wei W, Yoo KH, Cho WC, Feng D, Heavey S. Interactions between radiotherapy resistance mechanisms and the tumor microenvironment. Crit Rev Oncol Hematol 2025; 210:104705. [PMID: 40107436 DOI: 10.1016/j.critrevonc.2025.104705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Resistance to radiotherapy (RT) presents a significant clinical challenge in management of cancer. Recent evidence points to specific mechanisms of resistance within the tumor microenvironment (TME), which we aim to discuss, with the aim of overcoming the clinical challenge. METHODS We performed the narrative review using PubMed and Web of Science databases to identify studies that reported the regulative network and treatments of RT resistance from TME perspectives. RESULTS RT significantly changes the immune TME of cancers, which is closely appearing to play a key role in RT resistance (RTR) by modulating immune cell infiltration and function. Various phenotypes are involved in the development of RTR, such as autophagy, senescence, oxidative stress, cell polarization, ceramide metabolism, and angiogenesis in the TME. Key genes and pathways are also implicated in RTR, including immune and inflammatory cytokines, TGF-β, P53, the NF-κB pathway, the cGAS/STING pathway, the ERK and AKT pathway, and the STAT pathway. Based on the mechanism of RTR in the TME, many proposed routes to overcome RTR, several specifically target the TME including targeting fibroblast activation protein, exosomes management, nanomedicine, and immunotherapy. Many challenges in RT resistance still need to be further explored with emerging investigative methods, such as artificial intelligence, genetic technologies, and bioengineering. CONCLUSIONS The complex interactions between RT and TME significantly affect the efficiency of RT. Novel approaches to overcome this clinical difficulty are promising, which needs future work to further explore and identify better treatment strategies.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinrui Li
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, Zhejiang 315211, China
| | - Siang Boon Koh
- Faculty of Health and Life Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yiqing Guo
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Uzoamaka Okoli
- Division of Surgery & Interventional Science, University College London, London, UK; Basic and Translational Cancer Research Group, Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria, Eastern part of Nigeria, Nsukka, Enugu, Nigeria
| | - Alisha Pati-Alam
- Division of Surgery & Interventional Science, University College London, London, UK
| | - Eduardo Mota
- Division of Surgery & Interventional Science, University College London, London, UK
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, South Korea
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region of China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China; Division of Surgery & Interventional Science, University College London, London, UK.
| | - Susan Heavey
- Division of Surgery & Interventional Science, University College London, London, UK.
| |
Collapse
|
41
|
Wang C, Liang W, Zhong J, Liu J, Zhou C, Ma C, Liao Y, Gao Y, Zhao J, He Y. m6A-mediated regulation of CPSF6 by METTL3 promotes oxaliplatin resistance in colorectal cancer through enhanced glycolysis. Cell Signal 2025; 130:111676. [PMID: 40010558 DOI: 10.1016/j.cellsig.2025.111676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/16/2024] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Oxaliplatin resistance poses a significant challenge in colorectal cancer (CRC) treatment. Recent studies have implicated CPSF6 in cancer progression and drug resistance, although its role in chemotherapy resistance and regulatory mechanisms is unclear. This study investigates CPSF6's involvement in oxaliplatin resistance in CRC and its regulation via m6A methylation by METTL3. We assessed CPSF6 expression in oxaliplatin-resistant (OxR) CRC cell lines (HT29-OxR and HCT116-OxR) compared to sensitive counterparts using qRT-PCR and Western blotting. CPSF6 was significantly upregulated in OxR cells, and its knockdown reduced cell viability, colony formation, and glycolytic activity while increasing apoptosis. m6A modification of CPSF6 mRNA was elevated in OxR cells, correlating with increased METTL3 expression. METTL3 knockdown decreased CPSF6 levels and m6A enrichment, enhancing mRNA degradation, while its overexpression stabilized CPSF6 mRNA, promoting resistance. Xenograft experiments showed that CPSF6 knockdown suppressed tumor growth and glycolysis. Thus, CPSF6 is identified as a mediator of oxaliplatin resistance in CRC, regulated by the METTL3/m6A axis, suggesting potential therapeutic targets to overcome resistance.
Collapse
Affiliation(s)
- Chengxing Wang
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Weijun Liang
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jietao Zhong
- Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China; Department of Gastroenterology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jiachen Liu
- Department of Nuclear Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong 510000, China
| | - Chaorong Zhou
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Changyi Ma
- Department of Radiology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Yuehua Liao
- Department of Pathology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Yuan Gao
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jinglin Zhao
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China.
| | - Yaoming He
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China.
| |
Collapse
|
42
|
Li Z, Bai Y, Wu H, Feng Y, Wang X, Zhao C, Wang X. PTEN/PI3K/AKT pathway activation with hypoxia-induced human umbilical vein endothelial cell exosome for angiogenesis-based diabetic skin reconstruction. Mater Today Bio 2025; 32:101651. [PMID: 40177380 PMCID: PMC11964554 DOI: 10.1016/j.mtbio.2025.101651] [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: 01/08/2025] [Revised: 02/06/2025] [Accepted: 03/08/2025] [Indexed: 04/05/2025] Open
Abstract
Diabetic skin, a major clinical challenge due to impaired wound healing, is often exacerbated by a hypoxic microenvironment at the wound site. Exosomes have been proven to have excellent biological activities and applied to solve many bioengineering problems. However, the wide application of exosomes is still limited by their short in vitro lifetime and low yield. To overcome these application limitations, this study specifically enhances the pro-angiogenic biological efficacy of exosomes through hypoxic treatment and achieves sustained release using hydrogel loading. In vitro, hypoxia-induced exosomes (Hp-Exo) significantly enhanced endothelial cell migration, proliferation, and angiogenic capacity. In vivo, Gelman hydrogels loaded with Hp-Exo accelerated wound closure, promoted collagen deposition, and increased vascularization in diabetic mice. miRNA sequencing of Hp-Exo revealed that exosomes induced under hypoxic conditions contain various miRNAs, which enhance vascular endothelial cell proliferation, migration, and angiogenesis through the PTEN/PI3K/AKT pathway. These results highlight that hypoxia-induced exosomes, when delivered through a biocompatible hydrogel platform, provide potential therapeutic approach to improve diabetic wound healing by stimulating angiogenesis and tissue regeneration.
Collapse
Affiliation(s)
- Zhenming Li
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Yuhao Bai
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Hao Wu
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Yisheng Feng
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xinxi Wang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Cancan Zhao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
- State Key Laboratory of Molecular Engineering of Polymers (Fudan University), Shanghai, 200438, China
| | - Xudong Wang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| |
Collapse
|
43
|
Manjili DA, Babaei FN, Younesirad T, Ghadir S, Askari H, Daraei A. Dysregulated circular RNA and long non-coding RNA-Mediated regulatory competing endogenous RNA networks (ceRNETs) in ovarian and cervical cancers: A non-coding RNA-Mediated mechanism of chemotherapeutic resistance with new emerging clinical capacities. Arch Biochem Biophys 2025; 768:110389. [PMID: 40090441 DOI: 10.1016/j.abb.2025.110389] [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: 11/11/2024] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Cervical cancer (CC) and ovarian cancer (OC) are among the most common gynecological cancers with significant mortality in women, and their incidence is increasing. In addition to the prominent role of the malignant aspect of these cancers in cancer-related women deaths, chemotherapy drug resistance is a major factor that contributes to their mortality and presents a clinical obstacle. Although the exact mechanisms behind the chemoresistance in these cancers has not been revealed, accumulating evidence points to the dysregulation of non-coding RNAs (ncRNAs), particularly long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), as key contributors. These ncRNAs perform the roles of regulators of signaling pathways linked to tumor formation and chemoresistance. Strong data from various recent studies have uncovered that the main mechanism of these ncRNAs in the induction of chemoresistance of CC and OC is done through a dysregulated miRNA sponge activity as competing endogenous RNA (ceRNA) in the competing endogenous RNA networks (ceRNETs), where a miRNA regulating a messenger RNA (mRNA) is trapped, thereby removing its inhibitory effect on the desired mRNA. Understanding these mechanisms is essential to enhancing treatment outcomes and managing the problem of drug resistance. This review provides a comprehensive overview of lncRNA- and circRNA-mediated ceRNETs as the core process of chemoresistance against the commonly used chemotherapeutics, including cisplatin, paclitaxel, oxaliplatin, carboplatin, and docetaxel in CC and OC. Furthermore, we highlight the clinical potential of these ncRNAs serving as diagnostic indicators of chemotherapy responses and therapeutic targets.
Collapse
Affiliation(s)
- Danial Amiri Manjili
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Naghdi Babaei
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Tayebeh Younesirad
- Department of Medical Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Sara Ghadir
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hamid Askari
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
44
|
Monton O, Kopecky K, Gurau A, Farber ON, Lilley EJ, Greer JB, Johnston FM. Trends and Predictors of Palliative Therapy Use in Young Adults with Advanced Gastrointestinal Cancer: A National Cancer Database Study. Ann Surg Oncol 2025; 32:4261-4271. [PMID: 40032735 PMCID: PMC12049391 DOI: 10.1245/s10434-025-17074-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/07/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Young adults (YAs) with advanced gastrointestinal (GI) cancer have unique care needs, which may be addressed through palliative therapy. OBJECTIVES The aims of this study were to describe temporal trends and identify predictors of palliative therapy utilization in YAs with advanced GI cancer. METHODS We conducted a retrospective cohort study using the National Cancer Database. YAs (18-39 years of age) diagnosed with advanced GI cancer from 2004 to 2020 were identified. We performed a trend analysis followed by univariable and multivariable logistic regression analyses. RESULTS A total of 43,616 YAs with advanced GI cancer were identified, of whom 3820 (8.76%) were treated with palliative therapy. The proportion of patients who received palliative therapy increased significantly, from 5.33% in 2004 to 12.36% in 2020 (ptrend < 0.05). Patients of non-White/non-Black race (odds ratio [OR] 1.23, 95% confidence interval [CI] 1.09-1.40), with no insurance (OR 1.35, 95% CI 1.20-1.53), and with a median income of less than $63,000 (OR 1.20, 95% CI 1.08-1.34) were more likely to receive palliative therapy. Multiple comorbidities (OR 1.59, 95% CI 1.24-2.06), stage IV disease (OR 8.28, 95% CI 7.33-9.34), and cancers of the esophagus (OR 2.26, 95% CI 1.88-2.71), liver (OR 2.19, 95% CI 1.88-2.56), pancreas (OR 2.20, 95% CI 1.53-3.16), and biliary tract (OR 2.12, 95% CI 1.54-2.91) were also predictors of palliative therapy utilization. CONCLUSIONS Palliative therapy utilization in YAs with advanced GI cancer increased significantly over the study period, however major gaps remain in the provision of this care. Further work is needed to understand the barriers to access among YAs.
Collapse
Affiliation(s)
- Olivia Monton
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Kimberly Kopecky
- Division of Surgical Oncology, Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Andrei Gurau
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Orly N Farber
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
- Center for Surgery and Public Health at Brigham and Women's Hospital, Boston, MA, USA
| | - Elizabeth J Lilley
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
- Center for Surgery and Public Health at Brigham and Women's Hospital, Boston, MA, USA
- Department of Psychological Oncology and Palliative Care, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jonathan B Greer
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Fabian M Johnston
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
45
|
Kanaan S, Altamimi A, Qattous H, Rbeihat H. Enhanced non-invasive machine learning approach for early colorectal cancer detection: Predictive modeling and validation in a Jordanian cohort. Comput Biol Med 2025; 191:110184. [PMID: 40249989 DOI: 10.1016/j.compbiomed.2025.110184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 01/16/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) ranks as the third most prevalent cancer worldwide, posing significant public health challenges. Late-stage detection often results in poor treatment outcomes, elevating mortality rates. The economic and psychological burdens of CRC treatment underscore the need for early detection. OBJECTIVE This study aims to enhance the early detection of colorectal cancer by employing machine learning (ML) algorithms on non-invasive features. The focus is on constructing a comprehensive dataset, analyzing non-invasive features, and developing predictive models to minimize the necessity for invasive procedures such as colonoscopy. By focusing on non-invasive, easily accessible data, the study aims to develop a model that can be widely applied without the associated risks of invasive procedures. METHODS A retrospective dataset of 400 patients was sourced from the colorectal cancer unit of Royal Medical Services (2021-2022). The dataset included demographic data, imaging reports, laboratory results, and clinical evaluations. The study involved three experiments, training ML models (K-Nearest Neighbors (KNN), Super Vector Machine (SVM), Random Forest (RF), Decision Tree (DT), and Naïve Bayes (NB)) on the collected dataset and a public dataset to validate generalizability. The first experiment used 35 features across the ML algorithms. The second experiment focused on the most informative features. The third experiment validated the models using a public dataset, with Phase I including all data and Phase II excluding missing values. RESULTS The Random Forest (RF) algorithm consistently outperformed other models, achieving an accuracy of 95.8 % in the first experiment, increasing to 96.5 % in the second experiment. For the public dataset, RF accuracy was 66.0 % in Phase I and 68.9 % in Phase II. Conversely, the KNN algorithm exhibited the lowest accuracy across all experiments. CONCLUSION This study highlights the effectiveness of ML in early CRC detection using non-invasive techniques. The RF model demonstrated superior accuracy, suggesting its potential application in clinical settings. The research contributes valuable insights into CRC detection within the local context and emphasizes the broader applicability of ML in improving cancer diagnosis and personalized treatment.
Collapse
Affiliation(s)
- Soha Kanaan
- Princess Sumaya University for Technology,(PSUT), Amman, Jordan.
| | - Ahmad Altamimi
- Department of Software Engineering, Princess Sumaya University for Technology (PSUT), Amman, Jordan
| | - Hazem Qattous
- Department of Software Engineering, Princess Sumaya University for Technology (PSUT), Amman, Jordan
| | | |
Collapse
|
46
|
Yan C, Wang G. Advances in research on flavonoids in tumor immunotherapy (Review). Mol Med Rep 2025; 31:150. [PMID: 40211703 PMCID: PMC11995692 DOI: 10.3892/mmr.2025.13515] [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: 09/03/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
Cancer immunotherapy is an approach used in anti‑tumor treatment; however, its efficacy is limited to specific tumor types that are inherently sensitive to immune system modulation. Expanding the scope of indications and enhancing the efficacy of cancer immunotherapy are key goals for continued advancement. Flavonoids modulate the tumor‑immunosuppressive microenvironment. Integrating flavonoids with immunotherapeutic modalities, including cancer vaccines, immune checkpoint inhibitors and adoptive immune‑cell therapy, has potential in terms of augmenting the therapeutic efficacy of immunotherapy. The present review aimed to summarize flavonoids that enhance cancer immunotherapy, focusing on their underlying mechanisms and the application of nanotechnology to overcome inherent limitations such as poor solubility, low bioavailability, rapid metabolism, and instability under physiological conditions, thereby highlighting the potential of flavonoids in advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Chaoguang Yan
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| | - Guangchun Wang
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| |
Collapse
|
47
|
Vyas A, Kumar K, Sharma A, Verma D, Bhatia D, Wahi N, Yadav AK. Advancing the frontier of artificial intelligence on emerging technologies to redefine cancer diagnosis and care. Comput Biol Med 2025; 191:110178. [PMID: 40228444 DOI: 10.1016/j.compbiomed.2025.110178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Artificial Intelligence (AI) is capable of revolutionizing cancer therapy and advancing precision oncology via integrating genomics data and digitized health information. AI applications show promise in cancer prediction, prognosis, and treatment planning, particularly in radiomics, deep learning, and machine learning for early cancer diagnosis. However, widespread adoption requires comprehensive data and clinical validation. While AI has demonstrated advantages in treating common malignancies like lung and breast cancers, challenges remain in managing rare tumors due to limited datasets. AI's role in processing multi-omics data and supporting precision oncology decision-making is critical as genetic and health data become increasingly digitized. METHOD This review article presents current knowledge on AI and associated technologies, which are being utilized in the diagnosis and therapy of cancer. The applications of AI in radiomics, deep learning, and machine learning for cancer screening and treatment planning are examined. The study also explores the capabilities and limitations of predictive AI in diagnosis and prognosis, as well as generative AI, such as advanced chatbots, in patient and provider interactions. RESULTS AI can improve the early diagnosis and treatment of high-incidence cancers like breast and lung cancer. However, its application in rare cancers is limited by insufficient data for training and validation. AI can effectively process large-scale multi-omics data from DNA and RNA sequencing, enhancing precision oncology. Predictive AI aids in risk assessment and prognosis, while generative AI tools improve patient-provider communication. Despite these advancements, further research and technological progress are needed to overcome existing challenges. CONCLUSIONS AI holds transformative potential for cancer therapy, particularly in precision oncology, early detection, and personalized treatment planning. However, challenges such as data limitations in rare cancers, the need for clinical validation, and regulatory considerations must be addressed. Future advancements in AI could significantly improve decision-support systems in oncology, ultimately enhancing patient care and quality of life. The review highlights both the opportunities and obstacles in integrating AI into cancer diagnostics and therapeutics, calling for continued research and regulatory oversight.
Collapse
Affiliation(s)
- Akanksha Vyas
- Academy of Scientific and Innovative Research, Ghaziabad, 201002, India
| | - Krishan Kumar
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Ayushi Sharma
- College of Medicine, Taipei Medical University, Taipei City, 110, Taiwan
| | - Damini Verma
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Dhiraj Bhatia
- Department of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Near Palaj, Gandhinagar, Gujarat, 382355, India
| | - Nitin Wahi
- Department of Biotechnology, LNCT University, Kolar Road, Shirdipuram, Bhopal, Madhya Pradesh, 462042, India
| | - Amit K Yadav
- Department of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Near Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
48
|
Nasrah R, Kanbalian M, Van Der Borch C, Dewar K, Chevalier S, Jagoe R. Stool Microbiome Features and Weight Change Response to Treatment for cancer cachexia. J Cachexia Sarcopenia Muscle 2025; 16:e13816. [PMID: 40325598 PMCID: PMC12052804 DOI: 10.1002/jcsm.13816] [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: 08/09/2024] [Revised: 03/06/2025] [Accepted: 03/19/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND AND AIMS Cancer cachexia is characterised by significant weight loss and muscle wasting that adversely affects patient outcomes. Nutritional interventions in cancer cachexia leads to improved outcomes, including improved weight change. However, there are wide variations in weight response to dietary interventions. Thus, it remains difficult to predict response to a given increase in dietary intake at an individual patient level. This study aimed to identify gut microbiome features that could serve as potential predictive biomarkers for response to individualized dietary intervention in patients with cancer cachexia attending the McGill Cancer Nutrition-Rehabilitation Program at the Jewish General Hospital (CNR-JGH). METHODS Participants were recruited from CNR-JGH clinic. Interventions included individualized nutritional counselling by a registered dietitian, to increase energy and protein intake to meet recommended levels. Stool DNA samples were collected at baseline (V1) and visit 2 (V2), and gut microbiome profiles were analysed to assess microbial diversity and identify differentially abundant genera in patients who lost weight (WL, N = 8) vs. maintained/gained weight (WSG, N = 29) at subsequent CNR-JGH clinic visits. RESULTS Greater alpha-diversity and higher Lachnospira genus abundance at baseline predicted higher likelihood that patients would have good response to CNR-JGH intervention (WSG at V2). Though predictors of poor response to nutritional intervention (WL at V2) were not identified, subjects in the WL group exhibited lower alpha-diversity and greater microbial population instability after CNR-JGH interventions. CONCLUSIONS In this cohort of patients with cancer-related weight loss attending a cancer cachexia clinic, certain gut microbiome features were associated with response to dietary interventions. Patients who lost weight after CNR-JGH intervention also developed a less diverse and less stable gut microbiome. Lachnospira genus abundance is a potential predictor of positive weight change response to dietary intervention as part of multimodal care for cancer cachexia, and further confirmatory studies are warranted. In addition, targeted dietary approaches to maintain diversity and gut microbiome population stability may have a role in improving the response to dietary interventions in cancer cachexia.
Collapse
Affiliation(s)
- Rima Nasrah
- McGill Cancer Nutrition Rehabilitation ProgramJewish General Hospital (CNR‐JGH)MontrealCanada
- Peter Brojde Lung Cancer CentreSegal Cancer Centre, Jewish General HospitalMontrealCanada
- Division of Experimental Medicine, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQCCanada
| | - Mary Kanbalian
- McGill Cancer Nutrition Rehabilitation ProgramJewish General Hospital (CNR‐JGH)MontrealCanada
| | - Christina Van Der Borch
- McGill Cancer Nutrition Rehabilitation ProgramJewish General Hospital (CNR‐JGH)MontrealCanada
| | - Ken Dewar
- McGill Centre for Microbiome ResearchMcGill UniversityMontrealQCCanada
- Department of Human GeneticsMcGill UniversityMontrealQCCanada
| | - Stéphanie Chevalier
- Research Institute of the McGill University Health CentreMontréalQCCanada
- School of Human NutritionMcGill UniversitySte‐Anne‐de‐BellevueQCCanada
- Department of MedicineMcGill UniversityMontrealQCCanada
| | - R. Thomas Jagoe
- McGill Cancer Nutrition Rehabilitation ProgramJewish General Hospital (CNR‐JGH)MontrealCanada
- Peter Brojde Lung Cancer CentreSegal Cancer Centre, Jewish General HospitalMontrealCanada
- Division of Experimental Medicine, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQCCanada
- Department of MedicineMcGill UniversityMontrealQCCanada
| |
Collapse
|
49
|
Vats M, Rathod D, Patel H, Richards T, Patel K. Self-emulsifying Nano-PND oral delivery systems of PND1186: In silico modeling for bioavailability estimation. J Mol Liq 2025; 426:127161. [PMID: 40322757 PMCID: PMC12048016 DOI: 10.1016/j.molliq.2025.127161] [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] [Indexed: 05/08/2025]
Abstract
Focal adhesion kinase (FAK) inhibitors have proven to aid the therapeutic potential of anti-cancer agents. PND1186 (PND) is a FAK inhibitor disrupting the oncogenic processes such as cell survival, proliferation, adhesion, migration and angiogenesis, as well as remodeling of tumor microenvironment. However, the pharmacological potential of PND is limited by its poor solubility and bioavailability due to rapid precipitation of weakly basic PND in the intestinal milieu. As a solution, we have developed a self-nanoemulsifying PND oral delivery system (NanoPODS) for rapid dissolution of PND while Soluplus containing system (NanoPODS-S) was prepared to prevent the precipitation of PND. Optimized NanoPODS-S depicted a particle size of 107.0 ± 3.6 nm, PDI of 0.223 ± 0.016, and a surface potential of -4.2 ± 0.007 mV, along with > 70% PND released at pH 6.8. In silico pharmacokinetics predicted 99% oral bioavailability for NanoPODS-S. This study evaluates the efficacy of NanoPODS and NanoPODS-S for improved oral bioavailability with better cytotoxicity efficacy on Pancreatic Ductal Adenocarcinoma (PDAC) cell lines. NanoPODS-S is the first of its kind, self-nanoemulsifying system containing a polymeric precipitation inhibitor mimicking a "spring-parachute effect". It will be a novel platform technology for rapid and enhanced dissolution of poorly soluble molecules.
Collapse
Affiliation(s)
- Mukti Vats
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Drishti Rathod
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Henis Patel
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Terjahna Richards
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| |
Collapse
|
50
|
Schabl L, Duraes LC, Erozkan K, Alipouriani A, Steele SR, Kessler H. Surgical and oncological outcomes in transverse colon carcinoma: does tumor sublocation make a difference? Langenbecks Arch Surg 2025; 410:156. [PMID: 40343536 DOI: 10.1007/s00423-025-03665-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/03/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Transverse colon carcinomas are often treated as a single entity in medical literature, despite differences in embryology, anatomy, physiology, genetics, and surgical treatment. We hypothesized that tumor sublocation affects demographics, oncological, surgical and quality of life outcomes. METHODS A retrospective analysis of patients who underwent surgery for transverse colon carcinomas between 2000 and 2018 was conducted. Tumor localization was determined by operative, pathological and imaging reports, and procedures were defined by the extent of vascular resection. RESULTS The study included 273 patients aged 69 years (SD 12.3), of whom 44% were female. The BMI was 28.8 kg/m2 (SD 6.2), and 61% were ASA class 3. Carcinomas were in the proximal (22%), the mid (42%), and the distal transverse colon (36%). Mid-transverse carcinomas exhibited the highest prevalence among female patients (53% vs. 35% proximal vs. 39% distal, p < 0.03). Proximal transverse carcinomas presented with a higher proportion of pathological stage II than mid and distal transverse carcinomas (68.3% vs. proximal vs. 44.3% mid-vs. 49% distal, p = 0.006). On multivariate analysis, anemia was more likely in mid than proximal (p = 0.009) and distal (p = 0.002) transverse colon cancers. Obstruction occurred more often in proximal than mid (p = 0.003), and hematochezia in distal than in mid (p < 0.001) and proximal (p = 0.014) transverse colon carcinomas. The 30-day mortality and morbidity and 5-year overall and disease-free survival rates were similar between the tumor sublocations. CONCLUSION Sublocations of the transverse colon carcinomas affect the symptoms of patients. Tumor sublocation does not impact the intraoperative, postoperative, or oncological outcomes and quality of life.
Collapse
Affiliation(s)
- L Schabl
- Department of Colorectal Surgery, Digestive Disease Institute, Cleveland Clinic, 9500 Euclid Ave, A30, Cleveland, OH, 44122, USA.
- Department of General, Visceral and Thoracic Surgery, University Hospital of Salzburg, Salzburg, Austria.
| | - L C Duraes
- Department of Colorectal Surgery, Digestive Disease Institute, Cleveland Clinic, 9500 Euclid Ave, A30, Cleveland, OH, 44122, USA
| | - K Erozkan
- Department of Colorectal Surgery, Digestive Disease Institute, Cleveland Clinic, 9500 Euclid Ave, A30, Cleveland, OH, 44122, USA
| | - A Alipouriani
- Department of Colorectal Surgery, Digestive Disease Institute, Cleveland Clinic, 9500 Euclid Ave, A30, Cleveland, OH, 44122, USA
| | - S R Steele
- Department of Colorectal Surgery, Digestive Disease Institute, Cleveland Clinic, 9500 Euclid Ave, A30, Cleveland, OH, 44122, USA
| | - H Kessler
- Department of Colorectal Surgery, Digestive Disease Institute, Cleveland Clinic, 9500 Euclid Ave, A30, Cleveland, OH, 44122, USA
| |
Collapse
|