201
|
Pierrard J, Dechambre D, Ooteghem GV. Investigation of changes in planning target volume and regression probability of rectal boost using in-silico cone-beam computed tomography-guided online-adaptive radiotherapy. Phys Imaging Radiat Oncol 2025; 34:100757. [PMID: 40231223 PMCID: PMC11994390 DOI: 10.1016/j.phro.2025.100757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/16/2025] Open
Abstract
Background and purpose Radiotherapy boost to the primary tumour may enable organ preservation in locally advanced rectal cancer (LARC). This study evaluated cone-beam computed tomography (CBCT)-guided online-adaptive radiotherapy (ART) to reduce rectal boost planning target volume (PTVBoost) margins and allow dose escalation. Materials and methods Eleven LARC patients were included in this in silico study. Population-based PTVBoost margins were computed for non-adaptive and online-ART using van Herk's formalism. Dose/volume results were compared between: non-adaptive RT with a 25 x 2.16 Gy boost (Non-ART54Gy), ART with a 25 x 2.16 Gy boost (ART54Gy), and ART with an escalated boost of 25 x 2.4 Gy (ART60Gy). Tumour regression probability was compared between each plan using a dose-response model. Results PTVBoost margins for non-adaptive vs. online-ART were 14.2 vs. 3.3 mm in the antero-posterior, 5.0 vs. 3.2 mm in the left-right, and 12.3 vs. 8.7 mm in the supero-inferior axes. PTVBoost and pelvic lymph node PTV coverage (V95%) were significantly improved with ART54Gy and ART60Gy compared to Non-ART54Gy (p < 0.001). High-priority organ-at-risk constraints (priority 1&2) were violated in 26.8 % of cases for Non-ART54Gy, 21.2 % of cases for ART54Gy, and 20.8 % of cases for ART60Gy. Tumour regression probability was superior for ART60Gy (20.8 %) compared to ART54Gy (17.0 %, p < 0.001) and Non-ART54Gy (16.9 %, p < 0.001). Conclusions Online-ART significantly reduce rectal boost PTV margin. It allows better target volume coverage with a similar risk of radiation-induced toxicities, even when escalating the dose. Therefore, online-ART should be considered to perform dose-escalation in LARC patients with the objective of organ preservation.
Collapse
Affiliation(s)
- Julien Pierrard
- UCLouvain, Institut de Recherche Experimentale et Clinique (IREC), Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Brussels, Belgium
- Department of Radiation Oncology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - David Dechambre
- Department of Radiation Oncology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Geneviève Van Ooteghem
- UCLouvain, Institut de Recherche Experimentale et Clinique (IREC), Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Brussels, Belgium
- Department of Radiation Oncology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
202
|
Torres T, Arellano Villanueva E, Alsabawi Y, Fofana D, Tripathi MK. Unraveling Early Onset Disparities and Determinants: An Analysis of Colorectal Cancer Outcomes and Trends in Texas. Cureus 2025; 17:e83124. [PMID: 40438851 PMCID: PMC12119150 DOI: 10.7759/cureus.83124] [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: 03/18/2025] [Accepted: 04/28/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Colorectal cancer (CRC) is the second leading cause of cancer-related deaths in the U.S., with disparities in incidence, survival, and age at diagnosis across racial, ethnic, and socioeconomic groups. The rising incidence of early-onset CRC (<50 years) has amplified concerns regarding access to care, screening disparities, and outcomes, particularly among minorities. This study examines the impact of race, ethnicity, socioeconomic status (SES), and sex on CRC survival and age at diagnosis in Texas from 1995 to 2016. Methods This retrospective cohort study utilized Texas Cancer Registry (TCR) data, including 235,076 CRC cases diagnosed between 1995 and 2016. Kaplan-Meier analysis and log-rank tests assessed 10-year survival by race and ethnicity and time period (1995-2005 vs. 2006-2016). Kruskal-Wallis tests with Bonferroni correction were used to compare survival years between racial/ethnic groups within each period. Age at diagnosis was analyzed by race and ethnicity, and SES using Welch ANOVA and Games-Howell post hoc testing. Welch's t-tests compared intra-race changes across decades. Sex-based differences in diagnosis age were assessed using Mann-Whitney U tests. Results Significant racial, ethnic, and socioeconomic disparities were observed in CRC outcomes. Black: Non-Hispanic and Black: Hispanic patients exhibited some of the lowest median survival times, with minimal overall improvement between the two time periods. Although Black: Hispanic patients exhibited the lowest median survival, the difference was not statistically significant in the 2006-2016 cohort (p = 0.12). Hispanic and Black patients were diagnosed at younger ages compared to White: Non-Hispanic patients. Lower SES was associated with younger age at diagnosis and worse survival. Male patients were consistently diagnosed earlier than female patients across both decades. Despite some improvement in survival for certain groups, disparities persisted, particularly for Black: Non-Hispanic and Black: Hispanic patients. Conclusion Disparities in CRC survival and diagnosis age persist across racial, ethnic, SES, and sex lines in Texas. These findings underscore the need for tailored screening efforts, improved healthcare access, and targeted interventions for high-risk populations. Persistent sex-based differences highlight a need for further research into biological and systemic factors. Addressing social determinants of health may help reduce these disparities.
Collapse
Affiliation(s)
- Tyler Torres
- General Surgery, University of Texas Rio Grande Valley School of Medicine, Edinburg, USA
| | | | - Yossef Alsabawi
- Otolaryngology, University of Texas Rio Grande Valley School of Medicine, Ediburg, USA
| | - Demba Fofana
- Mathematics and Statistics, University of North Carolina Greensboro, Greensboro, USA
| | - Manish K Tripathi
- Cancer and Immunology, Research Faculty, University of Texas Rio Grande Valley, Edinburg, USA
| |
Collapse
|
203
|
Thomas AL, Kulchar RJ, Stephens ES, Mason L, Jackson SS, Harris AR, Ewing AP, Shiels MS, Pichardo CM, McGee-Avila JK, Lawrence WR. County socioeconomic status and premature mortality from cancer in the United States. Cancer Epidemiol 2025; 95:102747. [PMID: 39827619 PMCID: PMC11890931 DOI: 10.1016/j.canep.2025.102747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
INTRODUCTION There are consistent data demonstrating socioeconomic status (SES) is associated with cancer survivorship among older adults, but research on the relationship between area-level SES and risk of premature mortality from cancer remains not well understood. This study investigated the association between county-level SES and premature mortality from cancer. METHODS Demographic characteristics and causes of death were ascertained from the national death certificate data for years 2016-2020. Premature cancer death was defined as cancer mortality between ages 25-64. County SES was calculated using the Yost Index and categorized into distribution-based quintiles (1 =lowest SES, 5=highest SES). To calculate the mortality-adjusted rate ratios (aRR) and corresponding 95 % confidence intervals (95 %CI) for the associations between county SES and cancer, we performed multivariable linear mixed models, adjusting for confounders. RESULTS A total of 3143 counties were included. The age-adjusted mortality rates of all cancers combined were 107.6, 98.4, 88.6, 81.1, and 66.7 per 100,000 population for the 5 SES quintiles, respectively. Compared with high SES counties, low SES counties had a 58 % greater premature cancer mortality rate (aRRquintile 1 vs.5 =1.58, 95 %CI: 1.55-1.60). Similar associations were observed when stratified by sex, though risk was greatest among men ([aRRwomen=1.48, 95 %CI: 1.45-1.52]; [aRRmen=1.66, 95 %CI: 1.62-1.70]). Among leading cancer types, the association was greatest for lung cancer mortality for the lowest SES counties (aRR=2.03; 95 %CI: 1.98-2.08). CONCLUSION Our findings demonstrate that lower SES counties are at greater risk of premature mortality from cancer. Place-based interventions should target the socioeconomic environment across the cancer control continuum.
Collapse
Affiliation(s)
- Aleah L Thomas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, United States; Weill Cornell Medicine, New York, NY, United States.
| | - Rachel J Kulchar
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States; School of Dentistry, University of California Los Angeles, Los Angeles, CA, United States
| | - Erica S Stephens
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Lee Mason
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Sarah S Jackson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Alexandra R Harris
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Aldenise P Ewing
- Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, OH, United States
| | - Meredith S Shiels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Catherine M Pichardo
- Division of Cancer Control and Population Sciences, NCI, NIH, Rockville, MD, United States
| | - Jennifer K McGee-Avila
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Wayne R Lawrence
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
204
|
Spencer A, Bedding C, Nicklin E, Flint H, Gilbert A. Understanding the impact of early onset colorectal cancer on quality of life: a qualitative analysis of online forum data. Qual Life Res 2025; 34:1003-1013. [PMID: 39589667 PMCID: PMC11982157 DOI: 10.1007/s11136-024-03857-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE Early onset colorectal cancer (EOCRC) is rising. The profile of health-related quality of life (HRQOL) impacts may differ in this younger cohort. Online forums are a source of unfiltered information regarding patient experience. This study used a qualitative analysis of online forum messages to elicit the unique HRQOL impacts of EOCRC. METHODS Messages were extracted from an online EOCRC UK forum. Inductive coding (with 10% dual-coding) and thematic analysis were used to describe the impact of diagnosis and treatment on HRQOL. RESULTS Data extraction and analyses were performed over one month; 463 messages (dated 01/04/2019 to 31/03/2024) were included. There was 100% concordance on dual-coding for main themes. Eight themes emerged: (1) diagnostic pathway and barriers; (2) parenthood and effect on children; (3) employment and finances; (4) fertility and early menopause; (5) stoma implications; (6) support systems, relationships and isolation; (7) sport and exercise and (8) mental health. CONCLUSIONS Qualitative thematic analysis of online forum data is a novel and efficient methodology for understanding the impact of cancer on HRQOL. Identified themes overlapped with those published in previous systematic reviews. This study offers new insights into the impact of isolation, early menopause, benefits of parenthood, psychological impact on children and practical and psychological implications of potential infertility in EOCRC. Current understanding of the diagnostic challenges and unique HRQOL impacts of EOCRC raises future research questions regarding how colorectal cancer services should evolve to provide support more in keeping with the needs of this growing younger cohort.
Collapse
Affiliation(s)
- Alice Spencer
- Leeds Institute of Medical Research at St James' Hospital, University of Leeds, Leeds, UK.
| | - Christopher Bedding
- Leeds Institute of Medical Research at St James' Hospital, University of Leeds, Leeds, UK
| | - Emma Nicklin
- Leeds Institute of Medical Research at St James' Hospital, University of Leeds, Leeds, UK
| | | | - Alexandra Gilbert
- Leeds Institute of Medical Research at St James' Hospital, University of Leeds, Leeds, UK
| |
Collapse
|
205
|
Oguz A, Uysal A, Özkan BNS, Oguz M, Yilmaz M. Isatin-modified Calixarene derivatives: A comprehensive study on synthesis, enzyme inhibition, antioxidant, antimicrobial, and Antiproliferative activities. Bioorg Chem 2025; 157:108280. [PMID: 39970757 DOI: 10.1016/j.bioorg.2025.108280] [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: 11/18/2024] [Revised: 01/31/2025] [Accepted: 02/12/2025] [Indexed: 02/21/2025]
Abstract
In this article, a series of calix[4]arenes derivatized with isatin derivatives at the phenolic-O position were synthesized as potential theranostic molecules for antitumor therapy. The cytotoxic mechanism of action of the synthesized compounds was determined by Alamar Blue assay and flow cytometry using MCF-7, MDA-MB-231, DLD1, HeLa and A549 human cancer cell lines and their ability to penetrate into PNT1A healthy epithelial cells. To detect DNA damage, the Comet test was applied after the synthesized compounds interacted with the cells. As a result, it was found that treated cells had abnormal tail nuclei and damaged DNA structures compared with untreated cells. Within the scope of enzyme inhibition experiments, studies were carried out on aromatase and COX-2 enzymes and it was determined that the compounds in the series showed inhibitory activity at varying rates. Especially compounds CLX-A3, CLX-A4, CLX-B3 and CLX-B5 attract attention with their enzyme inhibitor potential. Also, the antioxidant activities of the compounds whose synthesis was completed were also investigated and it was observed that the examined derivatives also had antioxidant activity potential. As a result of the antibacterial and antifungal test performed with broth microdilution, it was observed that the compounds had significant antibacterial and antifungal activity. The lowest MIC values were recorded as 0.006 mg/ml against Sarcina lutea and 0.048 against Candida albicans. In addition, the compound CLX-B3was observed to be effective against all strains including, Klebsiella pneumoniae and Salmonella enteritidis (Gram-negative pathogenic bacteria).
Collapse
Affiliation(s)
- Alev Oguz
- Department of Chemistry, University of Selcuk, Campus, 42031, Konya, Türkiye
| | - Ahmet Uysal
- Selcuk University, Vocational School of Health Services, Department of Medical Services and Techniques, Konya, Turkey
| | | | - Mehmet Oguz
- Department of Chemistry, University of Selcuk, Campus, 42031, Konya, Türkiye
| | - Mustafa Yilmaz
- Department of Chemistry, University of Selcuk, Campus, 42031, Konya, Türkiye.
| |
Collapse
|
206
|
Liu L, Chen D, Wen L, Ma Y, Li J, Zhang G, Hu H, Huang C, Yao X. Efficacy and safety of fruquintinib combined with PD-1 inhibitors in the treatment of refractory metastatic colorectal cancer: a systematic review and meta-analysis. Expert Rev Anticancer Ther 2025; 25:411-421. [PMID: 40035688 DOI: 10.1080/14737140.2025.2474736] [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: 11/04/2024] [Revised: 02/01/2025] [Accepted: 02/23/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Fruquintinib, a VEGFR1-3 tyrosine kinase inhibitor, is approved for treating refractory metastatic colorectal cancer. Recent clinical practice has shown that combining fruquintinib with programmed cell death protein 1 (PD-1) inhibitors can achieve better efficacy.The objective of this study is to assess the efficacy and safety of combining PD-1inhibitors with fruquintinib. METHODS We systematically searched PubMed, Cochrane Library, Embase, Wanfang, and CNKI up to 28 August 2024 for studies comparing fruquintinib combined with PD-1 inhibitors to fruquintinib alone. RevMan software was used to perform meta-analyses of survival data for the included studies. RESULTS A total of 9 retrospective cohort studies and 1 randomized controlled trial were included, involving a total of 716 patients. Compared with the monotherapy group, the combination therapy group had a greater Overall Response Rate [RR = 2.45,95% CI (1.83, 3.56), p < 0.00001], Disease Control Rate [RR = 1.37,95% CI (1.64,4.79), p = 0.0002], and progression-free survival [HR = 0.64,95% CI (0.49, 0.84), p = 0.001]. However, there was no significant difference in overall survival between the two groups. The incidence of adverse effects was identical in both groups. CONCLUSION Fruquintinib combined with PD-1 inhibitors was more effective than fruquintinib alone in the treatment of advanced colorectal cancer, with acceptable safety. REGISTRATION PROSPERO (registration number CRD42024583116).
Collapse
Affiliation(s)
- Linfeng Liu
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Dengzhuo Chen
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Liang Wen
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yongli Ma
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jinghui Li
- Department of General Surgery, Xinfeng County People's Hospital, Xinfeng, Jiangxi, China
| | - Guosheng Zhang
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Hongkai Hu
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Chengzhi Huang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xueqing Yao
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
207
|
Yao X, Deng S, Han X, Huang D, Cao Z, Ning X, Ao W. Deep Learning Algorithm‑Based MRI Radiomics and Pathomics for Predicting Microsatellite Instability Status in Rectal Cancer: A Multicenter Study. Acad Radiol 2025; 32:1934-1945. [PMID: 39289097 DOI: 10.1016/j.acra.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/27/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024]
Abstract
RATIONALE AND OBJECTIVES To develop and validate multimodal deep-learning models based on clinical variables, multiparametric MRI (mp-MRI) and hematoxylin and eosin (HE) stained pathology slides for predicting microsatellite instability (MSI) status in rectal cancer patients. MATERIALS AND METHODS A total of 467 surgically confirmed rectal cancer patients from three centers were included in this study. Patients from center 1 were randomly divided into a training set (242 patients) and an internal validation (invad) set (105 patients) in a 7:3 ratio. Patients from centers 2 and 3 (120 patients) were included in an external validation (exvad) set. HE and immunohistochemistry (IHC) staining were analyzed, and MSI status was confirmed by IHC staining. Independent predictive factors were identified through univariate and multivariate analyses based on clinical evaluations and were used to construct a clinical model. Deep learning with ResNet-101 was applied to preoperative MRI (T2WI, DWI, and contrast-enhanced T1WI sequences) and postoperative HE-stained images to calculate deep-learning radiomics score (DLRS) and deep-learning pathomics score (DLPS), respectively, and to DLRS and DLPS models. Receiver operating characteristic (ROC) curves were plotted, and the area under the curve (AUC) was used to evaluate and compare the predictive performance of each model. RESULTS Among all rectal cancer patients, 82 (17.6%) had MSI. Long diameter (LD) and pathological T stage (pT) were identified as independent predictors and were used to construct the clinical model. After undergoing deep learning and feature selection, a final set of 30 radiomics features and 30 pathomics features were selected to construct the DLRS and DLPS models. A nomogram combining the clinical model, DLRS, and DLPS was created through weighted linear combination. The AUC values of the clinical model for predicting MSI were 0.714, 0.639, and 0.697 in the training, invad, and exvad sets, respectively. The AUCs of DLPS and DLRS ranged from 0.896 to 0.961 across the training, invad, and exvad sets. The nomogram achieved AUC values of 0.987, 0.987, and 0.974, with sensitivities of 1.0, 0.963, and 1.0 and specificities of 0.919, 0.949, and 0.867 in the training, invad, and exvad sets, respectively. The nomogram outperformed the other three models in all sets, with DeLong test results indicating superior predictive performance in the training set. CONCLUSION The nomogram, incorporating clinical data, mp-MRI, and HE staining, effectively reflects tumor heterogeneity by integrating multimodal data. This model demonstrates high predictive accuracy and generalizability in predicting MSI status in rectal cancer patients.
Collapse
Affiliation(s)
- Xiuzhen Yao
- Department of Ultrasound, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China (X.Y.)
| | - Shuitang Deng
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (S.D., Z.C., X.N., W.A.)
| | - Xiaoyu Han
- Department of Pathology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (X.H.)
| | - Danjiang Huang
- Department of Radiology, Huangyan Hospital, Wenzhou Medical University, Taizhou First People's Hospital, Taizhou, Zhejiang Province, China (D.H.)
| | - Zhengyu Cao
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (S.D., Z.C., X.N., W.A.)
| | - Xiaoxiang Ning
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (S.D., Z.C., X.N., W.A.)
| | - Weiqun Ao
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (S.D., Z.C., X.N., W.A.).
| |
Collapse
|
208
|
Chen G, Liu W, Lin Y, Zhang J, Huang R, Ye D, Huang J, Chen J. Predicting bone metastasis risk of colorectal tumors using radiomics and deep learning ViT model. J Bone Oncol 2025; 51:100659. [PMID: 39902382 PMCID: PMC11787686 DOI: 10.1016/j.jbo.2024.100659] [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: 08/19/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 02/05/2025] Open
Abstract
Background Colorectal cancer is a prevalent malignancy with a significant risk of metastasis, including to bones, which severely impacts patient outcomes. Accurate prediction of bone metastasis risk is crucial for optimizing treatment strategies and improving prognosis. Purpose This study aims to develop a predictive model combining radiomics and Vision Transformer (ViT) deep learning techniques to assess the risk of bone metastasis in colorectal cancer patients using both plain and contrast-enhanced CT images. Materials and methods We conducted a retrospective analysis of 155 colorectal cancer patients, including 81 with bone metastasis and 74 without. Radiomic features were extracted from segmented tumors on both plain and contrast-enhanced CT images. LASSO regression was applied to select key features, which were then used to build traditional machine learning models, including Support Vector Machine (SVM), K-Nearest Neighbors (KNN), Random Forest, LightGBM, and XGBoost. Additionally, a dual-modality ViT model was trained on the same CT images, with a late fusion strategy employed to combine outputs from the different modalities. Model performance was evaluated using AUC-ROC, accuracy, sensitivity, and specificity, and differences were statistically assessed using DeLong's test. Results The ViT model demonstrated superior predictive performance, achieving an AUC of 0.918 on the test set, significantly outperforming all traditional radiomics-based models. The SVM model, while the best among traditional models, still underperformed compared to the ViT model. The ViT model's strength lies in its ability to capture complex spatial relationships and long-range dependencies within the imaging data, which are often missed by traditional models. DeLong's test confirmed the statistical significance of the ViT model's enhanced performance, highlighting its potential as a powerful tool for predicting bone metastasis risk in colorectal cancer patients. Conclusion The integration of radiomics with ViT-based deep learning offers a robust and accurate method for predicting bone metastasis risk in colorectal cancer patients. The ViT model's ability to analyze dual-modality CT imaging data provides greater precision in risk assessment, which can improve clinical decision-making and personalized treatment strategies. These findings underscore the promise of advanced deep learning models in enhancing the accuracy of metastasis prediction. Further validation in larger, multicenter studies is recommended to confirm the generalizability of these results.
Collapse
Affiliation(s)
- Guanfeng Chen
- Radiology Department of Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Wenxi Liu
- Radiology Department of The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Yingmin Lin
- Thyroid and Breast Surgery Department of the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Jie Zhang
- Radiology Department of Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Risheng Huang
- Radiology Department of Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Deqiu Ye
- Radiology Department of Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Jing Huang
- Radiology Department of The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Jieyun Chen
- Radiology Department of Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| |
Collapse
|
209
|
Gonzalez APG, Chovwen P, Myers S, Davids JS, Keshinro AO, Hill SS. Diversity, equity, and inclusion in colon and rectal surgery patient populations. Curr Probl Surg 2025; 65:101736. [PMID: 40128008 DOI: 10.1016/j.cpsurg.2025.101736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/13/2025] [Indexed: 03/26/2025]
Affiliation(s)
| | - Praise Chovwen
- Department of Surgery, Cleveland Clinic Foundation, Cleveland, OH
| | - Sara Myers
- Department of Surgery, Boston Medical Center, Boston, MA
| | | | | | - Susanna S Hill
- Department of Surgery, Duke University Medical Center, Durham, NC.
| |
Collapse
|
210
|
Kashif M. Gene expression profiling to uncover prognostic and therapeutic targets in colon cancer, combined with docking and dynamics studies to discover potent anticancer inhibitor. Comput Biol Chem 2025; 115:108349. [PMID: 39813876 DOI: 10.1016/j.compbiolchem.2025.108349] [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: 09/30/2024] [Revised: 12/05/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025]
Abstract
Drug resistance poses a major obstacle to the efficient treatment of colorectal cancer (CRC), which is one of the cancers that kill people most often in the United States. Advanced colorectal cancer patients frequently pass away from the illness, even with advancements in chemotherapy and targeted therapies. Developing new biomarkers and therapeutic targets is essential to enhancing prognosis and therapy effectiveness. My goal in this study was to use bioinformatics analysis of microarray data to find possible biomarkers and treatment targets for colorectal cancer. Using an ArrayExpress database, I examined a dataset on colon cancer to find genes that were differentially expressed (DEGs) in tumor versus healthy tissues. Integration of advanced bioinformatics tools provided robust insights into the identification and analysis of EGFR as a key player. STRING and Cytoscape enabled the construction and visualization of protein-protein interaction networks, highlighting EGFR as a hub gene due to its centrality and interaction profile. Functional enrichment analysis through DAVID revealed EGFR's involvement in critical biological pathways, as identified in GO and KEGG analyses. This underscores the power of combining computational tools to uncover significant biomarkers like EGFR. Autodock Vina screening of the NCI diversity dataset identified two potential EGFR inhibitors, ZINC13597410 and ZINC04896472. MD simulation data revealed that ZINC04896472 could be potential anticancer inhibitor. These findings serve as a basis for the creation of novel therapeutic approaches that target EGFR and other discovered pathways in CRC. The suggested strategy may improve the efficacy of CRC therapy and advance personalized medicine.
Collapse
Affiliation(s)
- Mohammad Kashif
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
211
|
Chen S, Yang S, Jiang J, Wang L. Outcomes of Postchemoradiotherapy Watch-and-Wait Strategy in Patients With Rectal Cancer: A 20-Year, Single-Center Study. J Surg Oncol 2025; 131:899-907. [PMID: 39635915 PMCID: PMC12120379 DOI: 10.1002/jso.28008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/03/2024] [Accepted: 11/09/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND OBJECTIVES The watch-and-wait (WW) strategy is a nonsurgical alternative for patients with rectal cancer exhibiting an excellent response to chemoradiotherapy. Studies on the WW strategy have primarily investigated 5-year oncological outcomes; few have focused on longer-term outcomes or the optimal patient selection approach for this therapeutic strategy. METHODS This retrospective study enrolled patients with locally advanced rectal adenocarcinoma who had achieved complete response after chemoradiotherapy. Patients who achieved pathological complete response were categorized into a control group (n = 95) and those who achieved clinical complete response and were managed using the WW strategy were categorized into a case group (n = 33). Kaplan-Meier estimates were calculated for the between-group comparison of survival. RESULTS The median follow-up duration was 89 months. Compared with the control group, the case group exhibited improved long-term sphincter preservation, particularly for low-lying tumors (p = 0.032), and inferior nonlocal-regrowth disease-free survival (p = 0.007). Within the case group, patients achieving a complete response by positron emission tomography exhibited 5-year survival rates similar to those achieving a complete endoscopic response. CONCLUSION The WW strategy is associated with improved sphincter preservation but worse nonlocal-regrowth disease-free survival. The potential of PET in patient selection for this strategy deserves further investigation.
Collapse
Affiliation(s)
- Shuo‐Fu Chen
- Department of Heavy Particles & Radiation OncologyTaipei Veterans General HospitalTaipeiTaiwan
| | - Shung‐Haur Yang
- Department of SurgeryNational Yang Ming Chiao Tung University HospitalYilanTaiwan
- School of Medicine, College of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Jeng‐Kai Jiang
- School of Medicine, College of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Division of Colon and Rectal Surgery, Department of SurgeryTaipei Veterans General HospitalTaipeiTaiwan
| | - Ling‐Wei Wang
- Department of Heavy Particles & Radiation OncologyTaipei Veterans General HospitalTaipeiTaiwan
- School of Medicine, College of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
212
|
Xu S, Wang H, Yan L, Han X. Isoegomaketone improves radiotherapy efficacy and intestinal injury by regulating apoptosis, autophagy and PI3K/AKT/mTOR signaling in a colon cancer model. Oncol Rep 2025; 53:51. [PMID: 40084689 PMCID: PMC11920779 DOI: 10.3892/or.2025.8884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 12/13/2024] [Indexed: 03/16/2025] Open
Abstract
The current study aimed to investigate the effect of isoegomaketone (IK) as a radiosensitizer for colon cancer and its effect on intestinal injury, and to verify its potential mechanism. A total of 40 BALB/c nude mice were selected to construct a HT‑29 tumor‑bearing mice model with T lymphocyte deficiency. Tumor size was measured every other day, and the survival of mice was counted. Intestinal and tumor tissues of mice were harvested when the experiment ended. The levels of inflammatory factors and markers of oxidative stress in intestinal tissues of different groups of mice were analyzed by ELISA. Western blotting was used to examine the expression of apoptosis‑ and autophagy‑related proteins, and the phosphorylation levels of the PI3K/AKT/mTOR signaling pathway in HT‑29 cells and tumor tissues. Radiotherapy (RT) combined with IK significantly reduced the viability of HT‑29 cells. The optimal dose proportion of RT combined with IK was 8 Gy and 100 µg/ml, and the combination index was <1, suggesting a strong combination effect. In addition, IK could further promote radiation DNA damage in HT‑29 cells by inhibiting the PI3K/AKT/hypoxia inducible factor 1α (HIF‑1α) signaling pathway, while upregulating the expression of proapoptotic and autophagy‑related proteins in HT‑29 cells. In HT‑29 tumor‑bearing mice, RT in combination with IK significantly inhibited the growth of xenografts and improved mouse survival. In addition, the combination of RT and IK significantly upregulated BAX and Beclin‑1 expression, downregulated BCL‑2 expression, and promoted the conversion of LC3 I to LC3 II. Radiation induced an increase in inflammatory cytokine levels as well as oxidative stress marker levels in intestinal tissue. Western blot analysis showed that the combination of RT and IK significantly inhibited the phosphorylation level of the PI3K/AKT/mTOR signaling pathway compared with the control and monotherapy groups. IK could significantly enhance the efficacy of RT by regulating the apoptosis and autophagy of colon cancer tumors, and alleviate inflammation and oxidative stress by regulating the PI3K/AKT/mTOR signaling pathway to alleviate intestinal injury. The present findings suggest that IK can be used as a promising sensitizer and has the potential to enhance the efficacy and safety of RT for colon cancer.
Collapse
Affiliation(s)
- Shufeng Xu
- Department of Radiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Huiyang Wang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Linlin Yan
- Department of Radiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Xiaowei Han
- Department of Radiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| |
Collapse
|
213
|
Soulat A, Mohsenpour T, Roshangar L, Moaddab SY, Soulat F. Innovative Therapeutic Approach Targeting Colon Cancer Stem Cells: Transitional Cold Atmospheric Plasma. ACS OMEGA 2025; 10:12109-12121. [DOI: https:/doi.org/10.1021/acsomega.4c10378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Affiliation(s)
- Abolfazl Soulat
- Department of Atomic and Molecular Physics, Faculty of Sciences
- University of Mazandaran
| | - Taghi Mohsenpour
- Department of Atomic and Molecular Physics, Faculty of Sciences
- University of Mazandaran
| | - Leila Roshangar
- Department of Histology, Faculty of Medicine
- Tabriz University of Medical Sciences
| | | | - Fatemeh Soulat
- Applied Chemistry laboratory, Department of Chemistry, Faculty of Basic Science
- Azarbaijan Shahid Madani University (ASMU)
| |
Collapse
|
214
|
Han H, Wen Z, Yang M, Wang C, Ma Y, Chen Q, Jiang D, Xu Y, Fazal A, Jie W, Lv X, Yin T, Lin H, Lu G, Qi J, Yang Y, Xu G. Shikonin Derivative Suppresses Colorectal Cancer Cells Growth via Reactive Oxygen Species-Mediated Mitochondrial Apoptosis and PI3K/AKT Pathway. Chem Biodivers 2025; 22:e202403291. [PMID: 40022742 DOI: 10.1002/cbdv.202403291] [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: 12/11/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 03/04/2025]
Abstract
Colorectal cancer (CRC) is one of the deadliest cancers globally, ranking as the third most prevalent and second most lethal malignancy worldwide. The standard treatment for CRC typically involves a combination of surgery, radiotherapy, and chemotherapy. Despite advancements in CRC treatment, the prognosis remains unsatisfactory, primarily due to unclear mechanisms underlying tumorigenesis and the aggression of CRC. The aberrant activation of the PI3K/AKT pathway is frequently implicated in the initiation, progression, and metastasis of CRC. Studies have demonstrated that shikonin (SK) exerts anti-cancer effects. In this study, we evaluated the anti-tumor activities of a series of semi-synthesized SK derivatives against CRC cells. Our findings revealed that the SK derivative (M12) significantly inhibited the proliferation and colony formation of CRC cells, reduced cell migration, and induced apoptosis. Mechanistically, M12 enhanced the production of reactive oxygen species and downregulated the mitochondrial membrane potential, ultimately leading to mitochondrial apoptosis. Furthermore, M12 exhibited anti-CRC effects by modulating the PI3K/AKT signaling pathway and significantly suppressed tumorigenicity without causing notable adverse effects in mice. Therefore, targeting the PI3K/AKT pathway could be a promising treatment for CRC. M12 appears to be a promising candidate for the effective and safe treatment of CRC.
Collapse
Affiliation(s)
- Hongwei Han
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Zhongling Wen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Minkai Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Changyi Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Yudi Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Qingqing Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Dexing Jiang
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing, China
| | - Ye Xu
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing, China
| | - Aliya Fazal
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Wencai Jie
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Xiaoran Lv
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Tongming Yin
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Hongyan Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Guihua Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, China
| | - Jinliang Qi
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Yonghua Yang
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Guohua Xu
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
215
|
Garfinkle R, Kyriakopoulos G, Murphy BC, Larson DW, Shawki SF, Merchea A, Mishra N, Mathis KL, Perry W, Behm KT. Robotic-assisted surgery for locally advanced rectal cancer beyond total mesorectal excision planes: the Mayo Clinic experience. Surg Endosc 2025; 39:2498-2505. [PMID: 40000455 DOI: 10.1007/s00464-025-11634-3] [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: 09/27/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND The purpose of this study was to evaluate the surgical and oncological outcomes of robotic-assisted beyond-TME surgery for locally advanced rectal cancer. METHODS Consecutive adult (≥ 18 years old) patients who underwent a robotic-assisted proctectomy beyond-TME planes for primary or recurrent rectal cancer at three Mayo Clinic (USA) hospitals from 2017-2023 were included. Patient demographics and tumor and disease characteristics were obtained by review of the electronic health record. Outcomes of interest included postoperative complications, hospital length of stay, and pathologic and oncologic outcomes. RESULTS In total, 72 patients were included in the final cohort. Thirty-five (48.6%) patients underwent an extended resection without exenteration, while 22 (30.6%) underwent a multi-visceral en bloc exenteration; 20 (36.1%) patients underwent a lateral pelvic lymph node dissection, with or without a concomitant extended resection. Most cases had an advanced T-stage and an involved mesorectal fascia on pre-treatment MRI. The median operative time was 425.0 min (340.5-504.0) and the median estimated blood loss was 150.0 mL (75.0-277.5). Conversion to open surgery was needed in two (2.8%) cases. Nearly half the cohort (48.3%) experienced a postoperative complication and the median postoperative length of stay was 3.5 (3.0-7.0) days. Five cases had a positive margin, resulting in an R0 rate of 93.1%. None of the exenteration cases had a positive margin. After a median follow-up of 22.0 (13.0-45.7) months, 10 patients experienced a local recurrence (13.8%). CONCLUSION Robotic-assisted beyond-TME surgery can be performed safely with favorable postoperative clinical and oncologic outcomes.
Collapse
Affiliation(s)
- Richard Garfinkle
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA.
- Division of Colon and Rectal Surgery, Jewish General Hospital, 3755 Cote Saint-Catherine Road, Montreal, QC, H3T1E2, Canada.
| | | | - Brenda C Murphy
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
| | - David W Larson
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
| | - Sherief F Shawki
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
| | - Amit Merchea
- Division of Colon and Rectal Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Nitin Mishra
- Division of Colon and Rectal Surgery, Mayo Clinic, Phoenix, AZ, USA
| | - Kellie L Mathis
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
| | - William Perry
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
| | - Kevin T Behm
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
216
|
Kelley BS, Carroll CB, Hampton JM, Walker MR, Zafar SN, Hayden D, Schiefelbein A, Vidri RJ, Thompson BB, LoConte NK. Rectal Cancer Disparities Among the American Indian/Alaskan Native Populations. Cancer Med 2025; 14:e70892. [PMID: 40251934 PMCID: PMC12008663 DOI: 10.1002/cam4.70892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025] Open
Abstract
PURPOSE Recent work noted lower overall survival (OS) in American Indian/Alaskan Native (AI/AN) individuals diagnosed with colon cancer compared with non-Hispanic White (NHW) individuals. Rectal cancer demographic profiles at diagnosis and survival outcomes have not been reported. We sought to identify differences in rectal cancer diagnosis and outcomes between AI/AN and White populations. METHODS White and AI/AN patients aged 18 or older, diagnosed between 2004 and 2020 with rectal adenocarcinoma were identified within the National Cancer Database (NCDB). Unadjusted and adjusted analyses were used to evaluate demographic and clinical standardized differences (stddiff) between AI/AN and White patients. Survival analyses of those diagnosed with locally advanced rectal cancer (Stage II/III) were performed using the Kaplan-Meier methods and multivariate Cox-proportional hazards modeling. RESULTS 176,341 eligible cases were identified: 0.6% were AI/AN (N = 992) and 99.4% White (N = 175,349). Compared to the White population, AI/AN patients were younger at diagnosis (mean age 59.9 vs. 64.5 years; stddiff = 0.36) and had more advanced stage disease (44.8% vs. 43.7%; stddiff = 0.15). A higher percentage of AI/AN resided in the areas of the lowest median income (35.5% vs. 15.1%; stddiff = 0.62) per zip code, rural (9.9% vs. 2.2%; stddiff = 0.65), and used Medicaid as their primary payor (14.3% vs. 6.2%; stddiff = 0.63). Adjusted analyses suggest the AI/AN group has an increased hazard of death compared with the White population (HR, 1.14; 95% CI, 1.05-1.25; p = 0.003). CONCLUSIONS AI/AN patients with rectal cancer have a younger age and a more advanced stage at diagnosis. AI/AN race is associated with lower OS compared to White patients in multivariable analyses. Future efforts should focus on increasing colorectal cancer screening and access to treatment for AI/AN populations in an attempt to improve survival outcomes.
Collapse
Affiliation(s)
- Broc S. Kelley
- Department of Medicine, Division of Hematology, Oncology and Palliative CareUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | | | - John M. Hampton
- University of Wisconsin Carbone Cancer CenterMadisonWisconsinUSA
| | - Margaret R. Walker
- Department of Medicine, Division of Hematology, Oncology and Palliative CareUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Syed Nabeel Zafar
- University of Wisconsin Carbone Cancer CenterMadisonWisconsinUSA
- Division of Surgical OncologyDepartment of SurgeryUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Dana Hayden
- University of Wisconsin Carbone Cancer CenterMadisonWisconsinUSA
- Division of Colorectal SurgeryDepartment of SurgeryUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | | | - Roberto J. Vidri
- Division of Surgical OncologyDepartment of SurgeryUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Surgery, Division of Surgical Oncology and Endocrine SurgeryUniversity of North Carolina School of MedicineChapel HillNorth CarolinaUSA
| | - Bret Benally Thompson
- Department of Medicine, Division of Hematology, Oncology and Palliative CareUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- University of Wisconsin Carbone Cancer CenterMadisonWisconsinUSA
| | - Noelle K. LoConte
- Department of Medicine, Division of Hematology, Oncology and Palliative CareUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- University of Wisconsin Carbone Cancer CenterMadisonWisconsinUSA
| |
Collapse
|
217
|
Wu Z, Xiao H, Kloeber JA, Ouyang Y, Yin P, Huang J, Chen B, Zhu S, Lu J, Han Y, Tu X, Dragojevic S, Luo K, Ting AT, Welliver M, Lou Z. Parkin deficiency promotes colorectal tumorigenesis and progression through RIPK3-dependent necroptotic inflammation. Cancer Commun (Lond) 2025; 45:406-410. [PMID: 39754711 PMCID: PMC11999878 DOI: 10.1002/cac2.12648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/06/2025] Open
Affiliation(s)
- Zheming Wu
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
- Department of Radiation OncologyMayo ClinicRochesterUSA
| | - Huaping Xiao
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
- Department of Radiation OncologyMayo ClinicRochesterUSA
| | - Jake A Kloeber
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
- Mayo Clinic Medical Scientist Training ProgramMayo ClinicRochesterUSA
| | - Yaobin Ouyang
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
| | - Ping Yin
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
| | - Jinzhou Huang
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
| | - Bin Chen
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
- Department of Radiation OncologyMayo ClinicRochesterUSA
| | - Shouhai Zhu
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
| | - Jing Lu
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
| | - Yiqun Han
- Department of Radiation OncologyMayo ClinicRochesterUSA
| | - Xinyi Tu
- Department of Radiation OncologyMayo ClinicRochesterUSA
| | | | - Kuntian Luo
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
| | | | - Meng Welliver
- Department of Radiation OncologyMayo ClinicRochesterUSA
| | - Zhenkun Lou
- Division of Oncology ResearchDepartment of OncologyMayo ClinicRochesterUSA
| |
Collapse
|
218
|
Stecko H, Tsilimigras D, Iyer S, Daw J, Zhu H, Huang E, Kalady M, Pawlik TM. Association of non-gain-of-function alterations in exportin-1 with improved overall survival in colorectal cancer. J Gastrointest Surg 2025; 29:101990. [PMID: 39947513 DOI: 10.1016/j.gassur.2025.101990] [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: 12/21/2024] [Revised: 01/23/2025] [Accepted: 02/08/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Upregulation of nuclear export protein exportin-1 (coded by gene XPO1) has been previously demonstrated in multiple cancer subtypes, contributing to pharmacotherapy resistance and increased recurrence rates. This study aimed to explore the effect of non-gain-of-function (GOF) XPO1 alterations in patients with colorectal cancer (CRC). METHODS Patients with colon/rectal/colorectal adenocarcinoma were identified from the Memorial Sloan Kettering Clinicogenomic, Harmonized Oncologic Real-World Dataset using cBioPortal. A subpopulation with alterations in XPO1 was identified. Patients with known amplifications and GOF E571K and R749Q alterations were excluded, as were patients with in situ and stage IV disease. Survival analysis was performed via Kaplan-Meier and Cox proportional hazards analyses, adjusted for patient age and disease stage. RESULTS Among 5543 patients with CRC, 83 (1.5%) had alterations in the XPO1 locus, and 5460 patients (98.5%) did not. Of patients with XPO1 alteration, 66 (79.5%) had non-GOF alterations, and 17 (21.5%) had GOF point mutations or amplifications. Patients with non-GOF XPO1 alteration had a mortality hazard ratio of 0.601 (95% CI, 0.463-0.805; P =.011). When adjusted for patient age and disease stage, XPO1 co-alteration was associated with improved overall survival (OS) in patients with alterations in TP53, APC, FBXW7, SMAD4, and BRAF genes (all P <.01). CONCLUSION XPO1 alterations were associated with improved OS in patients with CRC. Associated survival benefits persisted when co-alterations were present, particularly in co-alterations with intranuclear tumor suppressor proteins.
Collapse
Affiliation(s)
- Hunter Stecko
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States; The Ohio State University College of Medicine, Columbus, OH, United States
| | - Diamantis Tsilimigras
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Sidharth Iyer
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States; The Ohio State University College of Medicine, Columbus, OH, United States
| | - Jad Daw
- The Ohio State University College of Medicine, Columbus, OH, United States
| | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Emily Huang
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Matthew Kalady
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States.
| |
Collapse
|
219
|
Akram U, Ahmed S, Fatima E, Ahmad E, Ashraf H, Hassan SA, Qureshi Z, Altaf F, Buckles D, Iqbal J, Mohamed Ahmed KAH. Efficacy and safety of oral sulfate solution versus polyethylene glycol for colonoscopy: A systematic review and meta-analysis of randomized controlled trials. DEN OPEN 2025; 5:e70113. [PMID: 40248440 PMCID: PMC12003215 DOI: 10.1002/deo2.70113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/19/2025]
Abstract
Background Colonoscopy is the gold standard for early detection and monitoring of colorectal cancer. Procedural effectiveness is dependent on optimal bowel preparation. Traditional polyethylene glycol (PEG) solutions are difficult to tolerate, whereas newer low-volume alternatives, including PEG with ascorbic acid and oral sulfate solutions (OSS), offer improved efficacy and tolerability. The meta-analysis was performed to evaluate the efficacy and safety of OSS compared to PEG for bowel preparation in colonoscopy. Methods Studies were identified by searching PubMed, Embase, Cochrane CENTRAL, and clinicaltrials.gov from inception until June 2024. Only randomized controlled trials comparing OSS with PEG were included. Data was analyzed using R version 4.4.0 using a random effects model to calculate risk ratios (RRs) and mean differences (MDs) with 95% confidence intervals (CIs). Results Twenty-one studies with 6346 participants met the inclusion criteria. OSS significantly improved adenoma detection (RR, 1.13; 95% CI, 1.04-1.22; p-value <0.01; I2 = 0%) and polyp detection rates (RR, 1.16; 95% CI, 1.06-1.26; p-value <0.01; I2 = 0%), and had a higher Boston Bowel Preparation Scale (BBPS) score (MD, 0.31; 95% CI, 0.13-0.50; p-value <0.01; I2 = 81%). PEG was associated with more sleep disturbances (RR, 0.45; 95% CI, 0.25-0.82; p-value = 0.03; I2 = 0%). However, other adverse effects were similar between both solutions. Conclusion OSS demonstrated superior adenoma and polyp detection rates. When compared to PEG, patients utilizing OSS achieved higher BBPS scores. Data gleaned support enhanced cleansing efficacy and safety of OSS as a bowel preparation regimen.
Collapse
Affiliation(s)
- Umar Akram
- Department of MedicineAllama Iqbal Medical CollegeLahorePakistan
| | - Shahzaib Ahmed
- Department of MedicineFatima Memorial Hospital College of Medicine and DentistryLahorePakistan
| | - Eeshal Fatima
- Department of MedicineServices Institute of Medical SciencesLahorePakistan
| | - Eeman Ahmad
- Department of MedicineFatima Memorial Hospital College of Medicine and DentistryLahorePakistan
| | - Hamza Ashraf
- Department of MedicineAllama Iqbal Medical CollegeLahorePakistan
| | - Syed Adeel Hassan
- Division of Digestive Diseases and NutritionUniversity of KentuckyLexingtonUSA
| | - Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac UniversityBridgeportUSA
| | - Faryal Altaf
- Department of Internal MedicineIcahn School of Medicine at Mount Sinai/BronxCare Health SystemNew YorkUSA
| | - Daniel Buckles
- Division of Gastroenterology and HepatologyThe University of Kansas Medical CenterKansas CityUSA
| | - Javed Iqbal
- Nursing Department Hamad Medical CorporationDohaQatar
| | | |
Collapse
|
220
|
Mills K, Figueroa F, Knight R, Ekpo E, Lee LC, Baldo L, Xu C, Wang S, Adelman RM, Pemu P, Levin T, Shaukat A, Liu JJ. Fact or Myth? Black Patients Do Not Want to Participate in Clinical Trials. Clin Transl Gastroenterol 2025; 16:e00826. [PMID: 39878425 PMCID: PMC12020698 DOI: 10.14309/ctg.0000000000000826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025] Open
Abstract
INTRODUCTION To assess strategies for optimizing participation of underserved minorities in a blood-based early colorectal cancer detection test study (PREEMPT CRC; NCT04369053) at a hospital serving primarily Black individuals. METHODS Culturally sensitive, racially congruent research staff approached individuals undergoing average-risk screening colonoscopy. Consent/study procedures were synchronized with clinical appointments. Enrolled and not-enrolled patient characteristics were compared. Recruitment was compared with other study sites. RESULTS In total, 247 of 509 eligible individuals were enrolled; most were identified as Black (88.7%). No baseline characteristics were associated with participation. Recruitment was high compared with other sites (11th centile). DISCUSSION Recruitment barriers for Black individuals can be overcome when easy, culturally sensitive access is facilitated.
Collapse
Affiliation(s)
- Krystal Mills
- Department of Medicine, Division of Gastroenterology, Morehouse School of Medicine, Atlanta, Georgia, USA
- Department of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | | | - RaKetra Knight
- Department of Medicine, Division of Gastroenterology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Emem Ekpo
- Department of Medicine, Division of Gastroenterology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | | | - Lance Baldo
- Freenome Inc, South San Francisco, California, USA
| | - Chuanbo Xu
- Freenome Inc, South San Francisco, California, USA
| | - Siqi Wang
- Department of Sociology, University at Buffalo, Buffalo, New York, USA
| | - Robert M. Adelman
- Department of Sociology, University at Buffalo, Buffalo, New York, USA
| | - Priscilla Pemu
- Department of Medicine, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Theodore Levin
- Kaiser Permanente Division of Research, Pleasanton, California, USA
| | - Aasma Shaukat
- Department of Medicine, Division of Gastroenterology, NYU Grossman School of Medicine, New York, New York, USA
| | - Julia J. Liu
- Department of Medicine, Division of Gastroenterology, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
221
|
Qiu Q, Ding Y, Guo X, Han J, Zhang J, Liu Y, She J, Chen Y. Extrachromosomal circular DNA as a novel biomarker for the progression of colorectal cancer. Mol Med 2025; 31:123. [PMID: 40165080 PMCID: PMC11960012 DOI: 10.1186/s10020-025-01164-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Extrachromosomal circular DNA (eccDNA) has potential in tumor diagnosis, particularly for improving diagnostic accuracy and early cancer detection; however, many challenges remain in its application to clinical practice. METHODS We conducted a Circle-Seq analysis on clinical samples at different stages of colorectal cancer progression to examine the dynamic changes of eccDNA during the progression of colorectal cancer. We used breakpoint-specific PCR to verify candidate eccDNAs identified by Circle-Seq. The results were further validated using the AOM/DSS-induced colorectal cancer model. RESULTS There was an increase in the abundance of eccDNA with the progression of colorectal cancer. The genes associated with these eccDNA molecules were primarily related to signaling pathways involved in tumor development and metastasis. Our analysis also revealed that eccDNA abundance positively correlates with gene expression, and eccDNA derived from specific genes has potential value for the early diagnosis of tumors. CONCLUSIONS This study revealed a connection between eccDNA and colorectal cancer progression and highlights the clinical potential of eccDNA for the early diagnosis of colorectal cancer.
Collapse
Affiliation(s)
- Quanpeng Qiu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yi Ding
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaolong Guo
- Center for Gut Microbiome Research, Med-X Institute, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jing Han
- Center for Gut Microbiome Research, Med-X Institute, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiaqi Zhang
- Center for Gut Microbiome Research, Med-X Institute, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yaping Liu
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Center for Gut Microbiome Research, Med-X Institute, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, China.
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Yinnan Chen
- Center for Gut Microbiome Research, Med-X Institute, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, China.
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Hubei Province Key Laboratory of Precision Radiation Oncology, Wuhan, China.
| |
Collapse
|
222
|
Maida M, Vitello A, Zullo A, Ramai D, Facciorusso A, Vassallo R. Gender Differences in Quality of Bowel Preparation for Colonoscopy: Post Hoc Analysis of a Randomized Controlled Trial. J Clin Gastroenterol 2025; 59:344-349. [PMID: 38847811 DOI: 10.1097/mcg.0000000000002024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/21/2024] [Indexed: 07/23/2024]
Abstract
OBJECTIVE Bowel cleansing is a key element for a quality colonoscopy. Despite recent advances, not all predictors of successful cleansing are fully known. This post hoc analysis of an RCT aims to explore gender differences in bowel cleansing quality. METHODS The "OVER" trial was a multicenter phase-4 RCT including 478 patients randomized 1:1 to receive split-dose 1L polyethylene glycol plus ascorbate (PEG+ASC) or 4L-PEG. In this post hoc analysis, multivariable logistic regression models were designed to assess predictors of cleansing success (CS) and adenoma detection rate (ADR) by gender. RESULTS Of the 478 randomized patients, 50.2% were males and 49.8% females.Overall, CS was comparable between females and males (87.1% vs 88.4, P = 0 .6), whereas CS in the right (95.7% vs 90.9, P = 0.049) and transverse colon (98.6% vs 93.9, P =0.011) was significantly higher in females.At multivariable regression analysis for CS outpatient setting (OR = 5.558) and higher withdrawal time (OR = 1.294) were independently associated with CS in females, whereas screening/surveillance indication (OR = 6.776) was independently associated with CS in males.At multivariable regression analysis for ADR, running time <5 hours (OR = 3.014) and higher withdrawal time (OR = 1.250) were independently associated with ADR in females, whereas older age (OR = 1.040) and higher withdrawal time (OR = 1.093) were independently associated with ADR in males. CONCLUSIONS This study showed different results in bowel preparation quality and different predictors of CS and ADR by gender. These findings suggest the need for further research to explore gender-specific approaches for bowel preparation.
Collapse
Affiliation(s)
- Marcello Maida
- Department of Medicine and Surgery, University of Enna 'Kore', Enna
- Gastroenterology Unit, Umberto I Hospital, Enna, Italy
| | | | - Angelo Zullo
- Gastroenterology Unit, Nuovo Regina Margherita Hospital, Roma
| | - Daryl Ramai
- Division of Gastroenterology, Hepatology, and Nutrition, University of Utah Health, Salt Lake City, UT
| | - Antonio Facciorusso
- Department of Medical Sciences, Gastroenterology Unit, University of Foggia, Foggia, Italy
| | | |
Collapse
|
223
|
Vallicelli C, Morezzi D, Perrina D, Fugazzola P, Pinson J, Vigutto G, Ghaly A, Viganò J, Tomasoni M, Ansaloni L, Tuech JJ, Catena F. Colon and rectal peritoneal carcinomatosis: are we mixing apples with oranges? A propensity score-matched analysis. Updates Surg 2025; 77:277-285. [PMID: 39827438 DOI: 10.1007/s13304-025-02104-5] [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: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Rectal cancer is universally considered a different disease entity as compared to colon cancer, except when dealing with colorectal peritoneal carcinomatosis (PC), in which the two cancers are deemed as the same one. The present study aims to investigate the influence of primary tumor location (colon vs. rectum) on oncologic outcomes in patients undergoing cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) for colorectal peritoneal metastases. Data from three referral centers undergoing CRS plus HIPEC for PC of colorectal origin were prospectively collected. The primary outcomes were overall survival (OS) and disease-free survival (DFS) according to primary tumor location (colic vs. rectal). Univariate and multivariate analyses were performed using the Cox proportional hazard model first on the total number of patients. Then, a propensity score matching using the nearest-neighbour method with a 1:1 ratio was performed. The study included 167 patients: 126 colic and 41 rectal PC. After propensity score matching, rectal primary tumor location was independently predictive of a lower DFS (HR 1.91; 95%CI 1.06-3.45; p = 0.031) but not of a lower OS (HR 1.12; 95%CI 0.57-2.21; p = 0.73). Post-matching 3-year DFS rates were 49.2% (95%CI 34,3-70,5%) and 19.4% (95%CI 9,4-40,2%) for colic and rectal PC, respectively. The present study shows a significantly worse DFS for rectal cancer PC undergoing CRS and HIPEC compared to colon cancer PC, suggesting a possible need for dedicated pathways for rectal PC patients and posing a question for rectal PC to be considered as a unique disease entity.
Collapse
Affiliation(s)
- Carlo Vallicelli
- General, Emergency and Trauma Surgery, Maurizio Bufalini Hospital, viale Ghirotti 286, 47521, Cesena, Italy.
| | - Daniele Morezzi
- General, Emergency and Trauma Surgery, Maurizio Bufalini Hospital, viale Ghirotti 286, 47521, Cesena, Italy
- Department of Digestive Surgery, Rouen University Hospital, Rouen, France
| | - Daniele Perrina
- General, Emergency and Trauma Surgery, Maurizio Bufalini Hospital, viale Ghirotti 286, 47521, Cesena, Italy
| | - Paola Fugazzola
- General and Emergency Surgery Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Jean Pinson
- Department of Digestive Surgery, Rouen University Hospital, Rouen, France
| | - Gabriele Vigutto
- General, Emergency and Trauma Surgery, Maurizio Bufalini Hospital, viale Ghirotti 286, 47521, Cesena, Italy
| | - Ahmed Ghaly
- General and Emergency Surgery Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Jacopo Viganò
- General and Emergency Surgery Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Matteo Tomasoni
- General and Emergency Surgery Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luca Ansaloni
- General and Emergency Surgery Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- University of Pavia, Pavia, Italy
| | - Jean-Jacques Tuech
- Department of Digestive Surgery, Rouen University Hospital, Rouen, France
| | - Fausto Catena
- General, Emergency and Trauma Surgery, Maurizio Bufalini Hospital, viale Ghirotti 286, 47521, Cesena, Italy
- Alma Mater Studiorum Bologna University, Bologna, Italy
| |
Collapse
|
224
|
Pei J, Li L, Li C, Li Z, Wu Y, Kuang H, Ma P, Huang L, Liu J, Tian G. Dumbbell probe-bridged CRISPR/Cas13a and nicking-mediated DNA cascade reaction for highly sensitive detection of colorectal cancer-related microRNAs. Biosens Bioelectron 2025; 273:117190. [PMID: 39862677 DOI: 10.1016/j.bios.2025.117190] [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: 09/28/2024] [Revised: 12/30/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths globally, necessitating the development of sensitive and minimally invasive diagnostic approaches. In this study, we present a novel diagnostic strategy by integrating dumbbell probe-mediated CRISPR/Cas13a with nicking-induced DNA cascade reaction (DP-bridged Cas13a/NDCR) for highly sensitive microRNA (miRNA) detection. Target miRNA triggers Cas13a-mediated cleavage of the dumbbell probe, releasing an intermediate strand that hybridizes with a methylene blue-labeled hairpin probe on the electrode surface. Nicking enzyme cleaves the formed duplex DNA, triggering a cascade reaction that amplifies the electrochemical signal. Under optimized conditions, the method demonstrates a detection limit of 8.26 fM for miRNA-21, with reliable specificity and long-term stability. Furthermore, integration with machine learning models using multiple miRNA markers improved diagnostic accuracy, differentiating CRC from colorectal polyps and healthy controls with 100% accuracy in clinical validation cohorts. This study highlights the potential of DP-bridged Cas13a/NDCR as a sensitive and accurate diagnostic tool for CRC.
Collapse
Affiliation(s)
- Jingwen Pei
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Lan Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Chang Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Zongying Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yu Wu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Haiyang Kuang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Pan Ma
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Li Huang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China.
| | - Gang Tian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, 646000, China.
| |
Collapse
|
225
|
Soulat A, Mohsenpour T, Roshangar L, Moaddab SY, Soulat F. Innovative Therapeutic Approach Targeting Colon Cancer Stem Cells: Transitional Cold Atmospheric Plasma. ACS OMEGA 2025; 10:12109-12121. [PMID: 40191350 PMCID: PMC11966581 DOI: 10.1021/acsomega.4c10378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025]
Abstract
Transitional cold atmospheric plasma (TCAP) represents a novel technique for generating plasma remotely from a primary source. It consists of a partially nonthermal ionized gas mixture containing charged and neutral particles, photons, and free radicals. In recent years, TCAP has attracted considerable attention in biomedical applications. In order to evaluate colon cancer stem cells' (CCSCs) proliferation, apoptotic induction, inflammatory response, and survival, TCAP was utilized both directly and indirectly in this study. Using argon and helium gases, TCAP was continuously delivered in two stages during the experiment. For direct state, TCAP was irradiated onto CCSCs for 3 and 5 min. In the indirect technique, Matrigel was treated with TCAP for 5 min before the introduction of cells. In vitro assays demonstrated that TCAP exposure significantly reduced the viability of CCSCs; helium gas and direct application had greater impacts than argon. Numerous investigations confirmed the induction of apoptosis, showing that the treated groups had more apoptotic cells and altered cellular structures than controls (****p < 0.0001). A substantial increase in the Bax/Bcl-2 ratio was found by analyzing the expression of the Bax and Bcl-2 genes, indicating increased susceptibility to apoptosis (*p = 0.0177 and ***p = 0.0004). The higher efficacy of the direct helium mode was further highlighted by inflammatory marker analysis, which showed a significant reduction in interleukin-6 and interleukin-8 expression in cells directly treated with TCAP-helium compared to TCAP-argon (**p = 0.0015 and ***p = 0.0007). Lastly, the proliferation test, which relies on K i-67 expression, demonstrated a noteworthy decline in all TCAP-treated groups, with the direct helium group exhibiting the most robust impact (**p = 0.0014). Overall, the findings highlight the potential of TCAP, particularly with helium, as a promising approach for selectively targeting CCSCs and providing insights into its therapeutic mechanisms for cancer treatment. TCAP, therefore, emerges as a unique therapeutic strategy with potential applications in cancer stem cell-targeted therapies.
Collapse
Affiliation(s)
- Abolfazl Soulat
- Department of Atomic and Molecular Physics, Faculty of Sciences, University of Mazandaran, 4741613534 Babolsar, Iran
| | - Taghi Mohsenpour
- Department of Atomic and Molecular Physics, Faculty of Sciences, University of Mazandaran, 4741613534 Babolsar, Iran
| | - Leila Roshangar
- Department of Histology, Faculty of Medicine, Tabriz University of Medical Sciences, 5166614766 Tabriz, Iran
| | - Seyyed Yaghoub Moaddab
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, 5166614766 Tabriz, Iran
| | - Fatemeh Soulat
- Applied Chemistry laboratory, Department of Chemistry, Faculty of Basic Science, Azarbaijan Shahid Madani University (ASMU), 5375171379 Tabriz, Iran
| |
Collapse
|
226
|
Cao KY, Zhang D, Bai LB, Yan TM, Chen Y, Jiang YY, Jiang ZH. Targeting NUCKS1 with a fragment of tRNA Asn(GUU) of Chinese yew for the treatment of colorectal cancer. Noncoding RNA Res 2025; 11:38-47. [PMID: 39736854 PMCID: PMC11683283 DOI: 10.1016/j.ncrna.2024.11.002] [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: 08/24/2024] [Revised: 10/22/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
Despite the discovery of numerous oncogenes in colorectal cancer (CRC), the development of associated drugs is limited, posing a significant challenge for CRC treatment. Identification of novel druggable targets is therefore crucial for the therapeutic development of CRC. Here, we report the first investigation on therapeutics targeting the potent oncogene NUCKS1 to suppress cancer progression. NUCKS1-orientated bioinformatics screening of NUCKS1 inhibitors from our library of tRNA fragments originated from medicinal plants identified tRF-T36, a 5' tRNA fragment of tRNAAsn(GUU) of Chinese yew (Taxus chinensis), exhibiting stronger inhibitory effects than taxol against CRC progression. Mechanistically, tRF-T36 binds directly to the 3' UTR of NUCKS1 mRNA to downregulate its expressions via RNAi pathway. High-throughput RNA sequencing indicated that the downregulated NUCKS1 induced by tRF-T36 further inhibits PI3K/Akt pathway, as verified by the significantly efficacy decrease of tRF-T36 mimic in co-treatment with 740Y-P, an agonist of PI3K/Akt pathway. Collectively, our findings emphasize the importance of NUCKS1 as a promising druggable target for CRC. Furthermore, the present study provides the first siRNA sequence, tRF-T36 mimic, as small RNA drug candidate, thereby shedding light on CRC therapeutics.
Collapse
Affiliation(s)
- Kai-Yue Cao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Da Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Long-Bo Bai
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Tong-Meng Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Yan Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Yu-Yang Jiang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Zhi-Hong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| |
Collapse
|
227
|
Van Den Puttelaar R, Shi KS, Smith R, Zhao J, Ogongo MK, Harlass M, Hahn AI, Zauber AG, Yabroff KR, Lansdorp-Vogelaar I. Implications of the initial Braidwood v. Becerra ruling for colorectal cancer outcomes: a modeling study. J Natl Cancer Inst 2025; 117:790-794. [PMID: 39361402 PMCID: PMC11972676 DOI: 10.1093/jnci/djae244] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/18/2024] [Accepted: 09/26/2024] [Indexed: 04/08/2025] Open
Abstract
The Affordable Care Act (ACA) eliminated patient cost-sharing for United States Preventive Service Task Force (USPSTF) recommended services. However, if the US Court of Appeals for the Fifth Circuit fully upheld a US District Court ruling in Braidwood Management v. Becerra, 666 F. Supp. 3d 613 (N.D. Tex 2023), cost-sharing for USPSTF recommendations made after ACA passage would have been reinstated for more than 150 million people. The case would have reinstated cost-sharing for colorectal cancer (CRC) screening for ages 45-49 years and for polyp removal during (diagnostic) colonoscopy across all ages. Using the MISCAN-Colon model, we simulated the potential impact on CRC outcomes, assuming early-onset CRC trends and lower screening participation. An 8-percentage-point decline in screening participation could increase CRC incidence by 5.1% and CRC mortality by 9.1%, with slightly lower costs due to increased cost-sharing. Larger decreases in screening participation can result in higher costs from increased incidence and delayed diagnoses.
Collapse
Affiliation(s)
- Rosita Van Den Puttelaar
- Department of Public Health, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands
| | - Kewei Sylvia Shi
- Surveillance and Health Equity Science Department, American Cancer Society, Atlanta, GA 30303, USA
| | - Robert Smith
- Center for Early Cancer Detection Science, American Cancer Society, Atlanta, GA 30303, USA
| | - Jingxuan Zhao
- Surveillance and Health Equity Science Department, American Cancer Society, Atlanta, GA 30303, USA
| | - Margaret Katana Ogongo
- Surveillance and Health Equity Science Department, American Cancer Society, Atlanta, GA 30303, USA
| | - Matthias Harlass
- Department of Public Health, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands
| | - Anne I Hahn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ann G Zauber
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - K Robin Yabroff
- Surveillance and Health Equity Science Department, American Cancer Society, Atlanta, GA 30303, USA
| | - Iris Lansdorp-Vogelaar
- Department of Public Health, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands
| |
Collapse
|
228
|
Zhang L, Ramesh P, Atencia Taboada L, Roessler R, Zijlmans DW, Vermeulen M, Picavet-Havik DI, van der Wel NN, Vaz FM, Medema JP. UGT8 mediated sulfatide synthesis modulates BAX localization and dictates apoptosis sensitivity of colorectal cancer. Cell Death Differ 2025; 32:657-671. [PMID: 39580596 PMCID: PMC11982410 DOI: 10.1038/s41418-024-01418-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
Elevated de novo lipid synthesis is a remarkable adaptation of cancer cells that can be exploited for therapy. However, the role of altered lipid metabolism in the regulation of apoptosis is still poorly understood. Using thermal proteome profiling, we identified Manidipine-2HCl, targeting UGT8, a key enzyme in the synthesis of sulfatides. In agreement, lipidomic analysis indicated that sulfatides are strongly reduced in colorectal cancer cells upon treatment with Manidipine-2HCl. Intriguingly, this reduction led to severe mitochondrial swelling and a strong synergism with BH3 mimetics targeting BCL-XL, leading to the activation of mitochondria-dependent apoptosis. Mechanistically, Manidipine-2HCl enhanced mitochondrial BAX localization in a sulfatide-dependent fashion, facilitating its activation by BH3 mimetics. In conclusion, our data indicates that UGT8 mediated synthesis of sulfatides controls mitochondrial homeostasis and BAX localization, dictating apoptosis sensitivity of colorectal cancer cells.
Collapse
Affiliation(s)
- Le Zhang
- LEXOR, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Prashanthi Ramesh
- LEXOR, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Lidia Atencia Taboada
- LEXOR, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Rebecca Roessler
- LEXOR, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Dick W Zijlmans
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
- Oncode Institute, Nijmegen, The Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
- Oncode Institute, Nijmegen, The Netherlands
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daisy I Picavet-Havik
- Medical Biology - MB Core Facility, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicole N van der Wel
- Medical Biology - MB Core Facility, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Amsterdam UMC location University of Amsterdam, Department of Laboratory Medicine and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Inborn errors of metabolism, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Paul Medema
- LEXOR, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Oncode Institute, Amsterdam, The Netherlands.
| |
Collapse
|
229
|
Chou CL, Lin CY, Li WS, Lee SW, Yang CC, Tian YF, Shiue YL, Tsai HH, Lai HY. Low CXCL11 expression is indicative of poor prognosis in rectal cancer patients undergoing preoperative chemoradiotherapy: a retrospective cohort study. Virchows Arch 2025; 486:803-815. [PMID: 39592484 PMCID: PMC12018498 DOI: 10.1007/s00428-024-03974-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024]
Abstract
INTRODUCTION Neoadjuvant concurrent chemoradiotherapy (CCRT) is routinely used before surgery in patients with locally advanced rectal cancer to reduce tumor size and decrease the risk of local recurrence. However, the disease-specific survival has not improved in most cases due to distant metastases. In selected individuals exhibiting a clinical complete response, non-operative management may be allowed; however, those who presented no or little response tend to have an inferior prognosis. Consequently, refined molecular characterization could aid in predicting which patients would benefit from neoadjuvant chemoradiotherapy. METHODS The mRNA level (by transcriptomic profiling) and protein expression (by immunohistochemical staining) of C-X-C motif chemokine ligand 11 (CXCL11) were integrated to predict neoadjuvant chemoradiotherapy efficacy. For survival analysis, clinicopathological features and CXCL11 immunoreactivity that were statistically significant in univariate analysis were included in multivariate analysis using the Cox proportional hazards regression model. RESULTS We identified that the CXCL11 level exhibits the most significant downregulation among neoadjuvant chemoradiotherapy non-responders. Using tumor samples from our rectal cancer cohort (n = 343) with immunohistochemistry validation, we demonstrated that low CXCL11 immunoexpression shows significant correlations with advanced disease and positive lymph nodes both prior to and following CCRT (all p < 0.001), vascular and perineural invasion (p < 0.001 and p = 0.006), and poor response to CCRT (p < 0.001). Moreover, low CXCL11 immunoexpression was an independent adverse prognostic factor significantly associated with patient survival. Additionally, we further identified pyroptotic cell death as an unrevealed role of CXCL11 in rectal cancer through bioinformatic analysis. CONCLUSION CXCL11 expression may serve as an early predictor of clinical outcomes and aid in therapeutic decision-making by identifying individuals likely to respond to neoadjuvant chemoradiotherapy in rectal cancer.
Collapse
Affiliation(s)
- Chia-Lin Chou
- Department of Medical Technology, Chung Hwa University of Medical Technology, Tainan, 717, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, 710, Taiwan
| | - Cheng-Yi Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, 710, Taiwan
| | - Wan-Shan Li
- Department of Medical Technology, Chung Hwa University of Medical Technology, Tainan, 717, Taiwan
- Department of Pathology, Chi Mei Medical Center, Tainan, 710, Taiwan
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Sung-Wei Lee
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
- Department of Radiation Oncology, Chi Mei Medical Center, Liouying, 736, Taiwan
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, 710, Taiwan
- College of Pharmacy and Science, Chia Nan University, Tainan, 717, Taiwan
| | - Yu-Feng Tian
- Division of Colon and Rectal Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, 710, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
- Institute of Precision Medicine, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Hsin-Hwa Tsai
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Hong-Yue Lai
- Department of Pharmacology, School of Medicine, College of Medicine, China Medical University, Taichung, 404, Taiwan.
| |
Collapse
|
230
|
Zhang C, Tian C, Zhu R, Chen C, Jin C, Wang X, Sun L, Peng W, Ji D, Zhang Y, Sun Y. CircSATB1 Promotes Colorectal Cancer Liver Metastasis through Facilitating FKBP8 Degradation via RNF25-Mediated Ubiquitination. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406962. [PMID: 39921520 PMCID: PMC11967755 DOI: 10.1002/advs.202406962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/14/2024] [Indexed: 02/10/2025]
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide and liver metastasis is the leading reason for its mortality. Circular RNAs (circRNAs) are conclusively associated with the progression of various cancers, rendering the exploration of its specific mechanisms in colorectal cancer liver metastasis(CRLM) highly valuable. Combined with GEO (Gene Expression Omnibus) databases and clinical data in our center, we found that high expression of circSATB1 is closely related to the progression of CRLM. Functionally, circSATB1 could significantly promote the metastatic ability of CRC cells in vitro and in vivo. Mechanistically, circSATB1 facilitated the RNF25-mediated ubiquitylation and degradation of FKBP8, releasing its inhibitory effects on mTOR signaling. In this process, circSATB1 acted as a scaffold for RNF25-FKBP8 complexes. Additionally, circSATB1 could be packaged in exosomes and secreted from the CRC primary tumors into plasma. In conclusion, this study uncovered a new circSATB1 that acts as a potent promoter of CRLM and offers novel insights into the precision therapeutic strategies for CRLM.
Collapse
Affiliation(s)
- Chuan Zhang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Chuanxin Tian
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Renzhong Zhu
- Institute of Translational Medicine, Medical CollegeYangzhou UniversityYangzhou225000China
| | - Chen Chen
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Chi Jin
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Xiaowei Wang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Lejia Sun
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Wen Peng
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Dongjian Ji
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Yue Zhang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Yueming Sun
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| |
Collapse
|
231
|
Li D, Morgan DR, Corral JE, Montgomery EA, Riquelme A, Shah SC. Gastric Cancer Screening in the United States: A Review of Current Evidence, Challenges, and Future Perspectives. Am J Gastroenterol 2025; 120:765-777. [PMID: 40072512 DOI: 10.14309/ajg.0000000000003301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 12/18/2024] [Indexed: 03/14/2025]
Abstract
Gastric cancer remains a leading cause of cancer-related mortality worldwide. In the United States, gastric cancer incidence and mortality are substantially higher among non-White racial and ethnic groups and new immigrants from high-incidence countries. This is in large part related to the higher prevalence of Helicobacter pylori -associated gastric premalignant changes in these populations. Apart from primary prevention, early detection of gastric cancer is the principal strategy to reduce gastric cancer mortality and improve survival. Extensive evidence in Asian countries has demonstrated the benefits of endoscopic screening in detecting early-stage gastric cancer and reducing gastric cancer-related mortality. By contrast, direct, high-quality US-based data, such as from large clinical trials or observational studies, on important outcomes of gastric cancer screening are still lacking. In this review, we evaluate and summarize the latest global evidence on the epidemiology and predisposing factors of gastric cancer as well as the efficacy, benefits vs. risks, and cost-effectiveness of gastric cancer screening. We further discuss the critical knowledge gaps and challenges in promoting gastric cancer screening in the United States. Dedicated research is urgently needed to enrich the US-based data on gastric cancer primary and secondary prevention to inform clinical practice and reduce gastric cancer-related morbidity and mortality in a cost and resource efficient manner.
Collapse
Affiliation(s)
- Dan Li
- Department of Gastroenterology, Kaiser Permanente Medical Center, Santa Clara, California, USA
- Kaiser Permanente Northern California Division of Research, Oakland, California, USA
| | - Douglas R Morgan
- Division of Gastroenterology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Juan E Corral
- Division of Gastroenterology, Prisma Health, Greenville, South Carolina, USA
| | - Elizabeth A Montgomery
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Arnoldo Riquelme
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Center for Control and Prevention of Cancer (CECAN), Santiago, Chile
| | - Shailja C Shah
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
- Gastroenterology Section, Jennifer Moreno Department of Veterans Affairs Medical Center, La Jolla, California, USA
| |
Collapse
|
232
|
Emile SH, Horesh N, Garoufalia Z, Gefen R, Dourado J, Wignakumar A, Wexner SD. Household income is independently associated with overall and cancer-specific survival after proctectomy for rectal cancer: A surveillance, epidemiology, and end results-based analysis. Am J Surg 2025; 242:116191. [PMID: 39823655 DOI: 10.1016/j.amjsurg.2025.116191] [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: 11/05/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
BACKGROUND We assessed association among household income, overall survival (OS), and cancer-specific survival (CSS) after proctectomy for rectal cancer. METHODS Population-based cohort study included stage I-III rectal adenocarcinoma patients who underwent proctectomy (2010-2020), subdivided by household income at diagnosis [low (<$50,000), average ($50,000-74,999), above-average (≥$75,000)] and compared. RESULTS Of 39,185 patients (59 % male; mean age 60.4), 12.5 % had low, 48.1 % had average, and 39.4 % had above-average income. Low-income patients were more often Black, rural dwellers, and undergone total proctectomy (OR: 1.49, p < 0.001). Income <$50,000 patients had shorter restricted mean OS (p < 0.001) and CSS (p < 0.001) than the other groups. 5-year OS (70.5 % vs. 73.6 % vs. 82.3 %, p < 0.001) and CSS (78.3 % vs. 80.6 % vs. 87.2 %, p < 0.001) were significantly lower in the low-income group than the average and above-average income groups. Adjusted for other factors, low-income was an independent predictor of OS (HR: 1.31, 95%CI: 1.22-1.41) and CSS (HR: 1.31, 95%CI: 1.21-1.43), compared to above-average. CONCLUSIONS Black patients and rural dwellers more often had <$50,000 income. Low-income increased odds of undergoing non-restorative surgery for rectal cancer and reduced OS and CSS.
Collapse
Affiliation(s)
- Sameh Hany Emile
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL, USA; Colorectal Surgery Unit, General Surgery Department, Mansoura University Hospitals, Mansoura, Egypt
| | - Nir Horesh
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL, USA; Department of Surgery and Transplantation, Sheba Medical Center and Faculty of Medicine, Ramat-Gan, Israel
| | - Zoe Garoufalia
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL, USA
| | - Rachel Gefen
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL, USA; Department of General Surgery, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Justin Dourado
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL, USA
| | - Anjelli Wignakumar
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL, USA
| | - Steven D Wexner
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL, USA.
| |
Collapse
|
233
|
Shao H, Liu M, Jiang H, Zhang Y. Polysaccharide-based drug delivery targeted approach for colon cancer treatment: A comprehensive review. Int J Biol Macromol 2025; 302:139177. [PMID: 39798740 DOI: 10.1016/j.ijbiomac.2024.139177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/10/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025]
Abstract
Colon cancer is a leading cause of cancer-related morbidity and mortality worldwide, necessitating advancements in therapeutic strategies to improve outcomes. Current treatment modalities, including surgery, chemotherapy, and radiation, are limited by systemic toxicity, low drug utilization rates, and off-target effects. Colon-targeted drug delivery systems (CDDS) offer a promising alternative by leveraging the colon's unique physiology, such as near-neutral pH and extended transit time, to achieve localized and controlled drug release. Polysaccharide-based CDDS, utilizing natural polymers like chitosan, cyclodextrin, pectin, guar gum, alginate, hyaluronic acid, dextran, chondroitin sulfate, and inulin, have emerged as innovative approaches for improving the specificity and efficacy of colon cancer treatments. These biocompatible and biodegradable polymers enable site-specific drug delivery, enhance tumor apoptosis, reduce systemic side effects, and improve patient compliance. This review evaluates recent advancements in polysaccharide-based CDDS, detailing their drug release mechanisms, therapeutic potential, and challenges in overcoming gastrointestinal transit and pH variability. Studies highlight the successful formulation of nanoparticles, microspheres, and other delivery systems, demonstrating targeted drug delivery, improved bioavailability, and enhanced cytotoxicity against colon cancer cells in-vitro and in-vivo. The review underscores the need for continued research on polysaccharide-based CDDS for colon cancer treatment, offering a path toward more effective, patient-centered oncological care.
Collapse
Affiliation(s)
- Hua Shao
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China
| | - Minghua Liu
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China
| | - Hongfang Jiang
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China.
| | - Ying Zhang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
234
|
Emerson B, Reiter PL, Klatt M, Gray DM, Hussan H, Chakraborty S, Katz ML. Development of a Brief Online Mindfulness-Based Intervention to Reduce Patient Anxiety Before a First-Time Screening Colonoscopy. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2025:10.1007/s13187-025-02608-z. [PMID: 40163313 DOI: 10.1007/s13187-025-02608-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/14/2025] [Indexed: 04/02/2025]
Abstract
To describe the development of an online mindfulness-based intervention (MBI) to reduce anxiety before a first-time screening colonoscopy among average-risk patients. A qualitative study used an iterative process guided by health behavior and mindfulness theories and feedback from a convenience sample of patients, endoscopy medical staff, and community members. Patient and medical staff (n = 18) were included in formative interviews (30-45 min), eight helped during intervention development sessions (15-90 min), and four community members reviewed the MBI in individual sessions (60 min). Interviews and sessions were recorded, transcribed verbatim, and analyzed using NVivo qualitative data software. Two themes emerged from the study: (1) both patients and medical staff reported that average-risk patients have pre-procedural anxiety before a first-time screening colonoscopy, and (2) using stakeholder-engaged strategies in an iterative process with both patients and medical staff is important so the developed intervention is acceptable to the priority population and to ensure medical accuracy and avoid disruption of workflow. Using an iterative process with key stakeholders is essential to develop interventions that are feasible and acceptable. The MBI developed through this process is being compared to usual care in a pilot randomized controlled trial to determine intervention feasibility and patient acceptability and to collect preliminary efficacy data. If efficacious, the developed MBI has the potential to reduce pre-procedural anxiety which may improve patient behaviors (e.g., bowel prep adherence and quality), patient satisfaction, and clinic workflow by reducing cancellation/no-shows, the amount of sedation required, and procedural time.
Collapse
Affiliation(s)
- Brent Emerson
- Division of Health Behavior and Health Promotion, College of Public Health, The Ohio State University, Columbus, OH, USA.
| | - Paul L Reiter
- Division of Health Behavior and Health Promotion, College of Public Health, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Maryanna Klatt
- Center for Integrative Health, Department of Family and Community Medicine, College Medicine, The Ohio State University, Columbus, OH, USA
| | - Darrell M Gray
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Hisham Hussan
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Subhankar Chakraborty
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Mira L Katz
- Division of Health Behavior and Health Promotion, College of Public Health, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
235
|
Zhang J, Sun Z, Li G, Ding L, Wang Z, Liu M. Discovering biomarkers associated with infiltration of CD8 + T cells and tumor-associated fibrosis in colon adenocarcinoma using single-cell RNA sequencing and gene co-expression network. Front Immunol 2025; 16:1496640. [PMID: 40230854 PMCID: PMC11994618 DOI: 10.3389/fimmu.2025.1496640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
Background Colorectal adenocarcinoma (COAD) is a prevalent malignant tumor associated with a high mortality rate. Within the tumor microenvironment, CD8+ T cells play a pivotal role in the anti-tumor immune response within the human body. Fibrosis directly and indirectly affects the therapeutic response of tumor immunotherapy. However, the significance of regulatory genes associated with tumor-associated fibrosis and CD8+ T cell infiltration remains uncertain. Therefore, it is imperative to identify biomarkers with prognostic value and elucidate the precise role of CD8+ T cells and tumor-associated fibrosis. Methods We performed a single-cell transcriptome analysis of COAD samples from the GEO database. To evaluate immune infiltration in COAD samples, we utilized CIBERSORT and ESTIMATE. Furthermore, we analyzed the correlation between CD8+ T cells and immune infiltration. To analyze COAD expression's quantitative immune cell composition data, we conducted a Weighted Gene Correlation Network Analysis and utilized a deconvolution algorithm. The data for these analyses were obtained from the GEO database. We utilized univariate Cox regression and LASSO analysis to create a prognostic model. The predictive model was assessed through Kaplan-Meier analysis, and a survival prediction nomogram was created. Additionally, we analyzed the correlation between the prognostic model and chemotherapy drug sensitivity. To estimate the expression of hub genes, we employed immunohistochemistry, real-time PCR, and western blot techniques. Results Single-cell transcriptome analysis has indicated a higher prevalence of CD8+ T cells in COAD tumor samples. The connection between COAD and CD8+ T cells was further confirmed by WGCNA and deconvolution analysis using the GEO database. The Protein-Protein Interaction network analysis revealed three hub genes: LARS2, SEZ6L2, and SOX7. A predictive model was subsequently created using LASSO and univariate COX regression, which included these three genes. Two of these hub genes (LARS2 and SEZ6L2) were found to be upregulated in COAD cell lines and tissues, while SOX7 was observed to be downregulated. The prognostic model demonstrated a significant association with CD8+ T cells, suggesting that these genes could serve as potential biomarkers and targets for gene therapy in treating COAD. Conclusion This study has identified three key genes associated with CD8+ T cells and the prognosis of COAD, providing new prognostic biomarkers for diagnosing and treating COAD.
Collapse
Affiliation(s)
- Jinning Zhang
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziquan Sun
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Heilongjiang Province Key Laboratory of Digestive Surgery and Nutrition & Metabolism, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Guodong Li
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Heilongjiang Province Key Laboratory of Digestive Surgery and Nutrition & Metabolism, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lixian Ding
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zitong Wang
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ming Liu
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Heilongjiang Province Key Laboratory of Digestive Surgery and Nutrition & Metabolism, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
236
|
Richiardone E, Giolito MV, Al Roumi R, Ambroise J, Boidot R, Drotleff B, Ghesquière B, Lupo B, Trusolino L, Bardelli A, Arena S, Feron O, Corbet C. Acidosis overrides molecular heterogeneity to shape therapeutically targetable metabolic phenotypes in colon cancers. Cancer Lett 2025; 613:217512. [PMID: 39900217 DOI: 10.1016/j.canlet.2025.217512] [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: 11/04/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
Colorectal cancer (CRC) represents a prototypical example of a cancer type for which inter- and intra-tumor heterogeneities remain major challenges for the clinical management of patients. Besides genotype-mediated phenotypic alterations, tumor microenvironment (TME) conditions are increasingly recognized to promote intrinsic diversity and phenotypic plasticity and sustain disease progression. In particular, acidosis is a common hallmark of solid tumors, including CRC, and it is known to induce aggressive cancer cell phenotypes. In this study, we report that long-term adaptation to acidic pH conditions is associated with common metabolic alterations, including a glycolysis-to-respiration switch and a higher reliance on the activity of phosphoglycerate dehydrogenase (PHGDH), in CRC cells initially displaying molecularly heterogeneous backgrounds. Pharmacological inhibition of PHGDH activity or mitochondrial respiration induces greater growth-inhibitory effects in acidosis-exposed CRC cells in 2D and 3D culture conditions, and in patient-derived CRC organoids. These data pave the way for drugs targeting the acidic tumor compartment as a "one-size-fits-all" therapeutic approach to delay CRC progression.
Collapse
Affiliation(s)
- Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Maria Virginia Giolito
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Rim Al Roumi
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Jérôme Ambroise
- Centre des Technologies Moléculaires Appliquées (CTMA), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 54, B-1200, Brussels, Belgium
| | - Romain Boidot
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges-François Leclerc Cancer Center-UNICANCER, 21079, Dijon, France
| | | | - Bart Ghesquière
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Barbara Lupo
- Department of Oncology, University of Torino, Candiolo (TO), Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo (TO), Italy
| | - Livio Trusolino
- Department of Oncology, University of Torino, Candiolo (TO), Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo (TO), Italy
| | - Alberto Bardelli
- Department of Oncology, Molecular Biotechnology Center, University of Torino, Torino, Italy; IFOM ETS, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Sabrina Arena
- Department of Oncology, University of Torino, Candiolo (TO), Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo (TO), Italy
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium; WEL Research Institute, Avenue Pasteur 6, B-1300, Wavre, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium; WEL Research Institute, Avenue Pasteur 6, B-1300, Wavre, Belgium.
| |
Collapse
|
237
|
Vonica RC, Butuca A, Morgovan C, Pumnea M, Cipaian RC, Frum A, Dobrea CM, Vonica-Tincu AL, Pacnejer AM, Ghibu S, Batar F, Gligor FG. Bevacizumab-Insights from EudraVigilance Database on the Assessments of the Safety Profile of Monoclonal Antibodies Used as Targeted Cancer Treatment. Pharmaceuticals (Basel) 2025; 18:501. [PMID: 40283938 PMCID: PMC12030381 DOI: 10.3390/ph18040501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/22/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Worldwide, colon cancer is a major cause of cancer-related mortality, with an increasing incidence influenced by genetic, environmental, and lifestyle factors. Despite advances in diagnosis and personalized treatments, challenges remain in improving patient prognosis, particularly in metastatic colorectal cancer (mCRC). Bevacizumab (BEV), a monoclonal antibody, is widely used in colorectal cancer treatment. This study aimed to analyze adverse events associated with BEV compared with other therapies based on data from the EudraVigilance (EV) database. Methods: A descriptive and disproportionality analysis was conducted on signals reported in the EV database related to BEV. The study included comparisons with other antineoplastic treatments, such as chemotherapy, targeted therapy, and immunotherapy. Patient demographics, severity of adverse drug reactions (ADRs), and distribution patterns were analyzed to assess the safety profile of BEV in colorectal cancer treatment. Results: The majority of the signals for BEV were from patients aged 18-64 years (39.42%) and 65-85 years (34.08%). Hypertension, thromboembolism, proteinuria, and gastrointestinal disorders have been the most frequently reported. Serious ADRs, including gastrointestinal perforations, hemorrhage, and arterial thromboembolism, were observed in 93.74% of Individual Case Safety Reports. BEV was associated with a higher likelihood of vascular and endocrine disorders compared with chemotherapy and other targeted therapies. Immunotherapy was linked to increased immunological ADRs, while BEV demonstrated fewer immune-related toxicities. Conclusions: Continuous monitoring is necessary to optimize patient management, particularly in elderly patients or those with cardiovascular comorbidities. Understanding BEV's safety profile allows for better personalization of treatment strategies, minimizing risks while enhancing therapeutic outcomes.
Collapse
Affiliation(s)
- Razvan Constantin Vonica
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Anca Butuca
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Manuela Pumnea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Remus Calin Cipaian
- Clinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania;
- County Clinical Emergency Hospital of Sibiu, 2–4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Adina Frum
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Carmen Maximiliana Dobrea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Andreea Loredana Vonica-Tincu
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Aliteia-Maria Pacnejer
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania
| | - Steliana Ghibu
- Department of Pharmacology, Physiology and Pathophysiology, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Florina Batar
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Felicia Gabriela Gligor
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| |
Collapse
|
238
|
Cregg J, Edwards AV, Chang S, Lee BJ, Knox JE, Tomlinson ACA, Marquez A, Liu Y, Freilich R, Aay N, Wang Y, Jiang L, Jiang J, Wang Z, Flagella M, Wildes D, Smith JAM, Singh M, Wang Z, Gill AL, Koltun ES. Discovery of Daraxonrasib (RMC-6236), a Potent and Orally Bioavailable RAS(ON) Multi-selective, Noncovalent Tri-complex Inhibitor for the Treatment of Patients with Multiple RAS-Addicted Cancers. J Med Chem 2025; 68:6064-6083. [PMID: 40056080 DOI: 10.1021/acs.jmedchem.4c02314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Oncogenic RAS mutations are among the most common in human cancers. To target the active, GTP-bound state of RAS(ON) directly, we employed an innovative tri-complex inhibitor (TCI) modality. Formation of a complex with an intracellular chaperone protein CypA, an inhibitor, and a target protein RAS blocks effector binding, inhibiting downstream RAS signaling and tumor cell proliferation. Herein, we describe the structure-guided SAR journey that led to the discovery of daraxonrasib (RMC-6236), a noncovalent, potent tri-complex inhibitor of multiple RAS mutant and wild-type (WT) variants. This orally bioavailable bRo5 macrocyclic molecule occupies a unique composite binding pocket comprising CypA and SWI/SWII regions of RAS(ON). To achieve broad-spectrum RAS isoform activity, we deployed an SAR campaign that focused on interactions with residues conserved between mutants and WT RAS isoforms. Concurrent optimization of potency and drug-like properties led to the discovery of daraxonrasib (RMC-6236), currently in clinical evaluation in RAS mutant advanced solid tumors (NCT05379985; NCT06040541; NCT06162221; NCT06445062; NCT06128551).
Collapse
Affiliation(s)
- James Cregg
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Anne V Edwards
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Stephanie Chang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Bianca J Lee
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - John E Knox
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | | | - Abby Marquez
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Yang Liu
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Rebecca Freilich
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Naing Aay
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Yingyun Wang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Lingyan Jiang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Jingjing Jiang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Zhican Wang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Michael Flagella
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - David Wildes
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | | | - Mallika Singh
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Zhengping Wang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Adrian L Gill
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Elena S Koltun
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| |
Collapse
|
239
|
Neira M, Mena C, Torres K, Simón L. The Potential Benefits of Curcumin-Enriched Diets for Adults with Colorectal Cancer: A Systematic Review. Antioxidants (Basel) 2025; 14:388. [PMID: 40298630 PMCID: PMC12024020 DOI: 10.3390/antiox14040388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/30/2025] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. Conventional treatments such as chemotherapy and radiotherapy are often associated with severe side effects and limited effectiveness. Curcumin, a polyphenol derived from Curcuma longa, has demonstrated anti-inflammatory and anticancer properties. A systematic review of the recent scientific literature followed PRISMA guidelines to evaluate the benefits of a curcumin-enriched diet for adults with colorectal cancer. Articles published between 2018 and 2024 were retrieved from PubMed, SciELO, Google Scholar, and Scopus. Studies meeting the inclusion criteria focused on curcumin, adults, and colorectal cancer outcomes. The administration of curcumin-containing products was associated with improved survival rates, enhanced quality of life, tumor reduction, and anti-inflammatory effects. A curcumin-enriched diet shows potential as an effective adjunct therapy for CRC patients, though its limited bioavailability and potential side effects, such as gastrointestinal discomfort, pose challenges. Addressing these limitations through larger cohorts, extended study durations, and improved formulations to enhance bioavailability is essential. Such efforts could enable the development of personalized dietary recommendations for CRC management.
Collapse
Affiliation(s)
| | | | - Keila Torres
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501015, Chile; (M.N.); (C.M.)
| | - Layla Simón
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501015, Chile; (M.N.); (C.M.)
| |
Collapse
|
240
|
Chang L, Qin C, Chu Y, Guan M, Deng X. Migrasome-Related Genes as Potential Prognosis and Immunotherapy Response Predictors for Colorectal Cancer. Biomedicines 2025; 13:799. [PMID: 40299331 PMCID: PMC12024535 DOI: 10.3390/biomedicines13040799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Studies highlight the role of migrasomes as mediators of intercellular communication and signaling, critical in influencing tumorigenesis and progression. Yet migrasome-related genes and their potential role in colorectal cancer prognosis remain unexplored. Methods: Differentially expressed gene set A (DEG set A) was identified in the TCGA-CRC dataset, and Weighted Gene Co-expression Network Analysis (WGCNA) was performed to identify the most important modules associated with migrasome-related gene (MRG) scores. Single-cell RNA-seq dataset GSE231559 DEG set B was determined. Candidate migrasome-related genes were filtered by intersecting DGE set A, key module genes, and DEG set B. Prognostic genes were subsequently screened through regression analysis, and a risk model was developed. Patients with CRC in the TCGA cohort were stratified into high- and low-risk groups based on the optimal cutoff of the risk score. Immunotherapy response-related analyses were then performed. Finally, cell-to-cell communication analysis was carried out for key cells identified based on prognostic gene expression analysis in annotated cells. Results: The six candidate migrasome-related genes were identified through the overlap of 5158 DEG set A, 1960 key module genes, and 146 DEG set B. Further screening led to the selection of T1MP1, CXCL8, and MGP as potential prognostic biomarkers. Immune-related analysis indicated that the high-risk group exhibited a better response to immunotherapy. Notably, the prognostic genes showed elevated expression levels in monocytes and tissue stem cells, thereby designating them as key cell types. Conclusions: We conducted bioinformatic analysis of migrasome-related genes and identified significant involvement of T1MP1, CXCL8, and MGP in influencing CRC prognosis and immunotherapy response. Our research provides novel insights into the role of migrasomes in CRC biology.
Collapse
Affiliation(s)
- Lu Chang
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai 200040, China; (L.C.); (C.Q.)
| | - Chao Qin
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai 200040, China; (L.C.); (C.Q.)
| | - Yimin Chu
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200050, China;
| | - Ming Guan
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai 200040, China; (L.C.); (C.Q.)
| | - Xuan Deng
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai 200040, China; (L.C.); (C.Q.)
| |
Collapse
|
241
|
Tao XY, Li QQ, Dong SS, Wang H, Yang YQ, Yang X, Zeng Y. Long noncoding HOXD-AS1: a crucial regulator of malignancy. Front Cell Dev Biol 2025; 13:1543915. [PMID: 40206400 PMCID: PMC11979210 DOI: 10.3389/fcell.2025.1543915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) play a crucial role in the occurrence and progression of various cancers. HOXD-AS1, an antisense RNA 1 of the lncRNA HOXD cluster, (also known as HAGLR, MIR7704HG, Mdgt, and STEEL), is located at human chromosome 2q31.1. Recent studies have demonstrated that the abnormal expression of HOXD-AS1 is significantly correlated with the clinicopathological features of patients with various tumors. The expression of HOXD-AS1 is abnormal in various tumors, affecting tumor cell proliferation, apoptosis, metastasis, invasion, metabolism, and drug resistance. HOXD-AS1 is important for cancer diagnosis and prognosis evaluation. Detecting its expression level helps judge cancer progression and predict patient survival. It is a therapeutic target and biomarker for early diagnosis and prognosis, with good clinical application prospects. This article reviews the role, molecular mechanisms, and potential clinical value of HOXD-AS1 in malignant tumor development.
Collapse
Affiliation(s)
- Xiang-Yuan Tao
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qian-Qian Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shan-Shan Dong
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hui Wang
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yu-Qing Yang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xi Yang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yong Zeng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
242
|
Li T, Li T, Liang Y, Yuan Y, Liu Y, Yao Y, Lei X. Colorectal cancer cells-derived exosomal miR-188-3p promotes liver metastasis by creating a pre-metastatic niche via activation of hepatic stellate cells. J Transl Med 2025; 23:369. [PMID: 40134019 PMCID: PMC11938777 DOI: 10.1186/s12967-025-06334-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/01/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND/AIM Metastasis is the leading cause of mortality for colorectal cancer (CRC). Cancer-derived exosomes are widely recognized as the primary catalysts behind the development of pre-metastasis niche (PMN) in distal sites. However, the exact mechanism behind this process in CRC remains elusive. This study aimed to investigate the function and mechanisms underlying the role of exosomal miR-188-3p in activating hepatic stellate cells (HSCs) to develop the PMN and promote liver metastasis. METHODS We extracted exosomes from CRC cells using ultracentrifugation. Exosomes were identified using transmission electron microscopy, nanoparticle tracking analysis, and Western blot. Exosome uptake was assessed using fluorescence tracing, exosome PKH67 staining, and real-time quantitative PCR. The effects of CRC cell-derived exosomes on HSCs migration were evaluated using Transwell migration and wound healing assays. Key differentially expressed miRNAs were screened from the GEO database, and bioinformatics prediction along with dual-luciferase reporter assays were used to identify downstream target genes of miR-188-3p. Downstream related proteins of the target genes were detected by Western blot. In vivo, the distribution of exosomes and activation of HSCs in the liver were explored by tail vein injection of exosomes into nude mice. Further, the impact of exosomal miR-188-3p on liver metastasis was investigated using a spleen injection liver metastasis model. Finally, the expression levels of miR-188-3p in exosomes from CRC patient plasma were determined by real-time quantitative PCR, and the relationship between the expression of miR-188-3p in plasma exosomes and CRC prognosis was analyzed. RESULTS The expression level of miR-188-3p within plasma exosomes demonstrated a statistically significant increase in CRC with liver metastasis compared to those without liver metastases. We also demonstrated the transferability of miR-188-3p from CRC cells to HSCs cells via the exosomes. Exosomal miR-188-3p plays a pivotal role in orchestrating the establishment of PMN through targeting PHLPP2 to activate HSCs before tumor metastasis. Exosomal miR-188-3p was found to actively foster in vivo metastasis of CRC. Additionally, plasma exosomal miR-188-3p potentially serves as a viable blood-based biomarker for CRLM. CONCLUSION Exosomal miR-188-3p derived from CRC cells can promote liver metastasis by activating HSCs to form a PMN through targeting PHLPP2 to activate the AKT/mTOR pathway. These results offer a new perspective on the mechanisms driving CRLM.
Collapse
Affiliation(s)
- Tao Li
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Taiyuan Li
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yahang Liang
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yuli Yuan
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yang Liu
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yao Yao
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiong Lei
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
243
|
Quiroz-Troncoso J, Alegría-Aravena N, Sáenz de Mierae B, Sánchez-Díez M, González-Martos R, Gavira-O’Neill CE, González EJ, González-Miquel M, Valdés Vergara C, González-Silva G, Bensadon-Naeder L, Galeano J, Ramírez-Castillejo C. Anticancer Potential, Phenolic Profile, and Antioxidant Properties of Synsepalum dulcificum (Miracle Berry) in Colorectal Tumor Cell Lines. Antioxidants (Basel) 2025; 14:381. [PMID: 40298626 PMCID: PMC12024304 DOI: 10.3390/antiox14040381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/14/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Polyphenols, recognized for their antioxidant capacity, have shown potential in improving the response treatment of various diseases, including cancer. In this context, polyphenols have the ability to induce cytotoxicity in tumor cells, making them possible complementary agents to current treatments. The present study aims to evaluate the effect of the aqueous extract of Synsepalum dulcificum, using the commercial product DMB®, on the proliferation of colorectal tumor cells. An aqueous extract of DMB® was obtained, and 12 compounds were identified through high-performance liquid chromatography (HPLC), with protocatechuic acid, gallic acid, and catechin being the most prominent. Regarding cytotoxicity, the extracts reduced cell viability in the DLD-1, HT29, SW480, and SW620 cell lines, with IC50 values of 7, 11, 13, and 15 mg/mL, respectively. The combination of oxaliplatin with the DMB® extract reduced the resistant population by up to 50% in the DLD-1 and SW620 cell lines, affecting the G2/M and S phases of the cell cycle, respectively. Additionally, treatment with the DMB® extract induced an increase in the expression of BCL2, CASP3, and CASP9, suggesting a mechanism of action associated with apoptosis. The aqueous extract of Synsepalum dulcificum (DMB®) exhibited cytotoxicity in colorectal cancer cells, enhancing the effect of oxaliplatin and activating apoptotic pathways, suggesting its potential as an adjuvant in anticancer therapies.
Collapse
Affiliation(s)
- Josefa Quiroz-Troncoso
- HST Group, Centro de Tecnología Biomédica (CTB), Departamento Biotecnología-B.V. ETSIAAB, Universidad Politécnica de Madrid, 28223 Madrid, Spain; (M.S.-D.); (R.G.-M.); (C.E.G.-O.)
- Departamento de Oncología, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain;
| | - Nicolás Alegría-Aravena
- Departamento de Oncología, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain;
- Grupo de Biología y Producción de Cérvidos, Instituto de Desarrollo Regional, Universidad de Castilla La Mancha, 02006 Albacete, Spain
- Asociación Española Contra el Cáncer (AECC)—Fundación Científica AECC, 28045 Madrid, Spain
| | - Blanca Sáenz de Mierae
- Departamento de Ingeniería Química Industrial y del Medio Ambiente, ETSI Industriales, Universidad Politécnica de Madrid, 28006 Madrid, Spain; (B.S.d.M.); (E.J.G.); (M.G.-M.)
| | - Marta Sánchez-Díez
- HST Group, Centro de Tecnología Biomédica (CTB), Departamento Biotecnología-B.V. ETSIAAB, Universidad Politécnica de Madrid, 28223 Madrid, Spain; (M.S.-D.); (R.G.-M.); (C.E.G.-O.)
- Departamento de Oncología, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain;
| | - Raquel González-Martos
- HST Group, Centro de Tecnología Biomédica (CTB), Departamento Biotecnología-B.V. ETSIAAB, Universidad Politécnica de Madrid, 28223 Madrid, Spain; (M.S.-D.); (R.G.-M.); (C.E.G.-O.)
- Departamento de Oncología, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain;
| | - Clara E. Gavira-O’Neill
- HST Group, Centro de Tecnología Biomédica (CTB), Departamento Biotecnología-B.V. ETSIAAB, Universidad Politécnica de Madrid, 28223 Madrid, Spain; (M.S.-D.); (R.G.-M.); (C.E.G.-O.)
- Departamento de Oncología, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain;
- Instituto de Investigación de Enfermedades Raras (IIER-AGH), Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Emilio J. González
- Departamento de Ingeniería Química Industrial y del Medio Ambiente, ETSI Industriales, Universidad Politécnica de Madrid, 28006 Madrid, Spain; (B.S.d.M.); (E.J.G.); (M.G.-M.)
| | - Maria González-Miquel
- Departamento de Ingeniería Química Industrial y del Medio Ambiente, ETSI Industriales, Universidad Politécnica de Madrid, 28006 Madrid, Spain; (B.S.d.M.); (E.J.G.); (M.G.-M.)
| | - Cristian Valdés Vergara
- Centro de Investigación de Estudios Avanzados del Maule, Universidad Católica de Chile, Talca 3460000, Chile;
| | - Gloria González-Silva
- Laboratorio de Microbiología Aplicada, Escuela de Ingeniería en Biotecnología, Centro de Biotecnología de los Recursos Naturales (CenBio), Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Talca 3466706, Chile;
| | | | - Javier Galeano
- Grupo de Sistemas Complejos, Departamento de Ingeniería Agroforestal, ETSIAAB, Universidad Politécnica de Madrid, 28040 Madrid, Spain;
| | - Carmen Ramírez-Castillejo
- HST Group, Centro de Tecnología Biomédica (CTB), Departamento Biotecnología-B.V. ETSIAAB, Universidad Politécnica de Madrid, 28223 Madrid, Spain; (M.S.-D.); (R.G.-M.); (C.E.G.-O.)
- Departamento de Oncología, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain;
| |
Collapse
|
244
|
Gao Y, Fu S, Peng Y, Zhou Y, Zhu J, Zhang X, Cai C, Han Y, Shen H, Zeng S. HMBOX1 reverses autophagy mediated 5-fluorouracil resistance through promoting HACE1-induced ubiquitination and degradation of ATG5 in colorectal cancer. Autophagy 2025:1-22. [PMID: 40126194 DOI: 10.1080/15548627.2025.2477443] [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: 09/06/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
Chemotherapy remains the primary treatment for unresectable or advanced postoperative colorectal cancers. However, its effectiveness is compromised by chemoresistance, which adversely affects patient outcomes. Dysregulated macroautophagy/autophagy is a proposed mechanism behind this resistance, with ubiquitination playing a key regulatory role. In this study, we identify the transcription factor HMBOX1 (homeobox containing 1) as a critical regulator of chemoresistance in colorectal cancer. RNA sequencing revealed that HMBOX1 is downregulated in drug-resistant colorectal cancer cells and tissues, with its low expression linked to poor prognosis. An integrated analysis of genes associated with autophagy and 5-fluorouracil (5-FU) resistance was conducted, verified in the colorectal cancer tissues of patients by single-cell RNA sequencing and immunostaining. Mass-spectrometry-based proteomics and RNA sequencing were used to elucidate the underlying molecular mechanisms. Functionally, upregulation of HMBOX1 enhances the sensitivity of colorectal cancer cells to the first-line treatment with 5-FU by inhibiting autophagy. Mechanistically, HMBOX1 promotes the transcription of the E3 ubiquitin ligase HACE1, which in turn enhances ATG5 K63-ubiquitination and subsequent proteasome-mediated degradation. This results in decreased ATG5 levels, inhibiting autophagy and thus reducing 5-FU resistance in colorectal cancer cells both in vitro and in vivo. Furthermore, we confirm that HMBOX1 expression positively correlates with HACE1 expression and inversely correlates with autophagy levels in clinical colorectal cancer tissues. Our findings suggest that HMBOX1 downregulation drives 5-FU resistance through autophagy enhancement in colorectal cancer, highlighting HMBOX1 as a potential target for improving chemosensitivity and patient prognosis.Abbreviation: 3-MA: 3-methyladenine; 5-FU: 5-fluorouracil; ATG: autophagy related; CASP3: caspase 3; C-CASP3: cleaved caspase 3; C-PARP: cleaved PARP; CCK8: cell counting kit-8; ChIP: chromatin immunoprecipitation; CHX: cycloheximide; CNV: copy number variation; co-IP: co-immunoprecipitation; COAD: colorectal adenocarcinoma; CQ: chloroquine; CRC: colorectal cancer; CR: complete response; FHC: fetal human colon; GEO: Gene Expression Omnibus; HACE1: HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1; HMBOX1: homeobox containing 1; IHC: immunohistochemistry; LC-MS/MS: liquid chromatography-tandem mass spectrometry; mIHC: multiplexed immunohistochemistry; MUT: mutant; NC: negative control; OS: overall survival; PBS: phosphate-buffered saline; PD: progressive disease; PFA: paraformaldehyde; PFS: progression-free survival; PR: partial response; qPCR: quantitative polymerase chain reaction; RAPA: rapamycin; SD: stable disease; TCGA: The Cancer Genome Atlas; TEM: transmission electron microscopy; TF: translation factor; USP22: ubiquitin specific peptidase 22; WT: wild type.
Collapse
Affiliation(s)
- Yan Gao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Shenao Fu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yinghui Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yulai Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Jiang Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangyang Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
245
|
Tang Z, Zhou G, Xu Y, Zhang Y. Survival analysis and prediction of early-onset colorectal cancer patients post-chemotherapy: an analysis based on the SEER database. Int J Colorectal Dis 2025; 40:74. [PMID: 40118983 PMCID: PMC11928432 DOI: 10.1007/s00384-025-04853-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2025] [Indexed: 03/24/2025]
Abstract
BACKGROUND The incidence of Early-Onset Colorectal Cancer (EOCRC) has risen markedly in recent years, garnering widespread attention due to its distinctive clinical and biological features. However, systematic research on prognostic risk factors and long-term survival prediction for EOCRC patients undergoing postoperative chemotherapy remains scarce. This study seeks to pinpoint critical prognostic factors for EOCRC patients receiving postoperative chemotherapy and to devise a survival prediction tool employing a Nomogram model. METHODS Patients diagnosed with EOCRC between 2010 and 2015, who underwent postoperative chemotherapy, were extracted from the SEER (Surveillance, Epidemiology, and End Results) database. Only those meeting the inclusion criteria were included. Univariate and multivariate Cox regression analyses were performed to determine independent risk factors influencing overall survival (OS). A Nomogram model was then developed using significant variables. The model's predictive accuracy and clinical utility were assessed through the concordance index (C-index), calibration curves, receiver operating characteristic (ROC) curves, and decision curve analysis (DCA). RESULTS A cohort of 9,205 patients was analyzed, with 6,445 randomly allocated to the training group and 2,760 to the validation group from the SEER database. Independent prognostic factors, including gender, race, marital status, primary tumor location, histological type, TNM stage, CEA levels, bone metastasis, liver metastasis, and lung metastasis, were identified through univariate and multivariate Cox regression analyses. A Nomogram model constructed from these factors yielded a C-index of 0.76 (0.75, 0.77) in the training group and 0.76 (0.75, 0.78) in the validation group, reflecting robust discriminative ability and consistency. The area under the curve (AUC) for predicting 1-year OS was calculated as 0.84 (0.81, 0.86) in the training group and 0.82 (0.78, 0.85) in the validation group. For 3-year OS, AUCs were recorded at 0.83 (0.82, 0.84) and 0.82 (0.80, 0.84), respectively, while for 5-year OS, AUCs reached 0.81 (0.80, 0.82) and 0.82 (0.80, 0.84). Calibration curves demonstrated close alignment between predicted and observed survival rates. Additionally, DCA affirmed the model's clinical decision-making value. CONCLUSION Prognostic risk factors for EOCRC patients receiving postoperative chemotherapy were systematically evaluated in this study, leading to the development of a Nomogram-based survival prediction model. This tool offers a robust scientific foundation for tailoring individualized treatment and guiding follow-up strategies.
Collapse
Affiliation(s)
- Zhiguo Tang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Guojia Zhou
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Yu Xu
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Yinxu Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China.
| |
Collapse
|
246
|
Guo J, Zhang Z. Integrated multi-omics unveils novel immune signature for predicting prognosis in colon cancer patients. Sci Rep 2025; 15:9788. [PMID: 40118975 PMCID: PMC11928561 DOI: 10.1038/s41598-025-85390-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 01/02/2025] [Indexed: 03/24/2025] Open
Abstract
Colon cancer, a prevalent malignancy, is subject to intricate immune modulation, which substantially affects both treatment efficacy and prognostic outcomes. Furthermore, colon cancer is highly heterogeneous, and our comprehensive understanding of its immune microenvironment has not yet been fully realized. There is still ample opportunity for in-depth investigation into the composition and interactions of immune cells within colon cancer, as well as their implications for disease prognosis. In this study, we employed single-cell data from colon cancer to distinguish immune cells from non-immune cells through cluster analysis. Furthermore, we conducted an in-depth analysis of myeloid and T cells, which were categorized into 20 distinct cell subpopulations. Functional enrichment analysis revealed T cells' active involvement in the Fatty Acid Metabolism and Adipogenesis pathways, while immune checkpoint-associated genes (ICGs) were notably upregulated in CD8+ T cells. Subsequent analysis involved calculating gene scores to characterize cell subpopulations, which, when combined with patient survival time analysis, revealed a significant association between the gene characterization score (referred to as "imm-score") and the survival of colon cancer patients. Specifically, the presence of CD8+-ANXA1hi-T cells was linked to shortened overall survival in the high imm-score subgroup. Subsequently, combined with genomic analysis, patients in the high imm-score subgroup exhibited elevated tumor mutation burden (TMB) and heightened activity in both the epithelial-mesenchymal transition (EMT) and Notch signaling pathway. Finally, according to our new algorithm, scores calculated predicted the effectiveness of immunotherapy for patients. The results revealed that patients with lower scores could achieve better therapeutic outcomes with immunotherapy. This study offers an extensive analysis of the interplay between T cells and myeloid cells within colon cancer tissues, exploring their impact on the survival and prognosis of colon cancer patients. Additionally, it unveils the potential significance of the imm-score in colon cancer, potentially indicating a poor prognosis and providing novel insights into the immune-regulatory mechanisms underlying the disease.
Collapse
Affiliation(s)
- Jing Guo
- Department of Gastrointestinal and Anorectal Surgery, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
| | - Zili Zhang
- Department of Gastrointestinal and Anorectal Surgery, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China.
| |
Collapse
|
247
|
Dong Y, Sheng G, Chen W. TPX2 knockdown mediates p53 activation to induce autophagy and apoptosis for anti-colorectal cancer effects. J Recept Signal Transduct Res 2025:1-13. [PMID: 40116489 DOI: 10.1080/10799893.2025.2470180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 03/23/2025]
Abstract
Colorectal cancer (CRC) exhibits high morbidity and mortality worldwide. Targeting protein for Xenopus kinesin-like protein 2 (TPX2) impacts various cancers; however, mechanism of TPX2 in CRC remains unclear. Xenograft nude mouse models were constructed by subcutaneous injection of HCT116 cells with sh-NC, sh-TPX2, OE-NC, and OE-TPX2 transfection. Following the test of tumor growth, immunohistochemistry and TUNEL staining were done. In vitro, HCT116, RKO, and SW480 cells were divided into sh-NC, sh-TPX2, and sh-TPX2 + 3-methyladenine (3-MA, autophagy inhibitor) groups. Further, sh-p53 and rapamycin (RA, autophagy agonist) were added in HCT116 cells. EdU staining, flow cytometry, transparent electron microscopy, and Western blot were performed. Comparing with sh-NC group, sh-TPX2 inhibited tumor growth and Ki67 expression, and increased LC3-II expression and apoptosis, whereas OE-TPX2 group presented an opposite trend. In vitro, HCT116 and RKO cells in sh-TPX2 group enhanced apoptosis and LC3 II/LC3 I expression, and inhibited proliferation and P62 expression, which were reversed after further 3-MA intervention. The above results were not found in SW480 cells. Moreover, compared to sh-TPX2 group, sh-TPX2 + RA group enhanced apoptosis and autophagy, and suppressed the proliferation of HCT116 cells, which were reversed following further sh-p53 intervention. Therefore, sh-TPX2 mediated p53 activation to induce autophagy for anti-CRC effects, providing new ideas for CRC treatment.
Collapse
Affiliation(s)
- Yunfei Dong
- Department of Anorectal, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Guixian Sheng
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenbin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
248
|
Kozak Y, Finiuk N, Czarnomysy R, Gornowicz A, Pinyazhko R, Lozynskyi A, Holota S, Klyuchivska O, Karkhut A, Polovkovych S, Klishch M, Stoika R, Lesyk R, Bielawski K, Bielawska A. Juglone-Bearing Thiopyrano[2,3-d]thiazoles Induce Apoptosis in Colorectal Adenocarcinoma Cells. Cells 2025; 14:465. [PMID: 40136714 PMCID: PMC11941218 DOI: 10.3390/cells14060465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Colorectal cancer is a major global health challenge, with current treatments limited by toxicity and resistance. Thiazole derivatives, known for their bioactivity, are emerging as promising alternatives. Juglone (5-hydroxy-1,4-naphthoquinone) is a naturally occurring compound with known anticancer properties, and its incorporation into thiopyrano[2,3-d]thiazole scaffolds may enhance their therapeutic potential. This study examined the cytotoxicity of thiopyrano[2,3-d]thiazoles and their effects on apoptosis in colorectal cancer cells. Les-6547 and Les-6557 increased the population of ROS-positive HT-29 cancer cells approximately 10-fold compared with control cells (36.3% and 38.5% vs. 3.8%, respectively), potentially contributing to various downstream effects. Elevated ROS levels were associated with cell cycle arrest, inhibition of DNA biosynthesis, and reduced cell proliferation. A significant shift in the cell cycle distribution was observed, with an increase in S-phase (from 17.3% in the control to 34.7% to 51.3% for Les-6547 and Les-6557, respectively) and G2/M phase (from 24.3% to 39.9% and 28.8%). Additionally, Les-6547 and Les-6557 inhibited DNA biosynthesis in HT-29 cells, with IC50 values of 2.21 µM and 2.91 µM, respectively. Additionally, ROS generation may initiate the intrinsic apoptotic pathway. Les-6547 and Les-6557 activated both intrinsic and extrinsic apoptotic pathways, demonstrated by notable increases in the activity of caspase 3/7, 8, 9, and 10. This study provides a robust basis for investigating the detailed molecular mechanisms of action and therapeutic potential of Les-6547 and Les-6557.
Collapse
Affiliation(s)
- Yuliia Kozak
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine; (N.F.); (O.K.); (M.K.); (R.S.)
| | - Nataliya Finiuk
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine; (N.F.); (O.K.); (M.K.); (R.S.)
| | - Robert Czarnomysy
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, Kilinskiego 1, 15-089 Białystok, Poland; (R.C.); (K.B.)
| | - Agnieszka Gornowicz
- Department of Biotechnology, Faculty of Pharmacy, Medical University of Bialystok, Kilinskiego 1, 15-089 Białystok, Poland;
| | - Roman Pinyazhko
- Department of Normal Physiology, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine;
| | - Andrii Lozynskyi
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (A.L.); (S.H.); (R.L.)
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (A.L.); (S.H.); (R.L.)
| | - Olga Klyuchivska
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine; (N.F.); (O.K.); (M.K.); (R.S.)
| | - Andriy Karkhut
- Department of Technology of Biologically Active Substances, Pharmacy and Biotechnology, Lviv Polytecnic National University, Bandera 12, 79013 Lviv, Ukraine; (A.K.); (S.P.)
| | - Svyatoslav Polovkovych
- Department of Technology of Biologically Active Substances, Pharmacy and Biotechnology, Lviv Polytecnic National University, Bandera 12, 79013 Lviv, Ukraine; (A.K.); (S.P.)
| | - Mykola Klishch
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine; (N.F.); (O.K.); (M.K.); (R.S.)
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine; (N.F.); (O.K.); (M.K.); (R.S.)
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (A.L.); (S.H.); (R.L.)
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszów, Sucharskiego 2, 35-225 Rzeszów, Poland
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, Kilinskiego 1, 15-089 Białystok, Poland; (R.C.); (K.B.)
| | - Anna Bielawska
- Department of Biotechnology, Faculty of Pharmacy, Medical University of Bialystok, Kilinskiego 1, 15-089 Białystok, Poland;
| |
Collapse
|
249
|
Zheng P, Xu M, Ma D, Feng L, Qin J, Gao X. Survival benefit and impact of adjuvant chemotherapy following neoadjuvant therapy in patients with locally advanced rectal cancer. Updates Surg 2025:10.1007/s13304-025-02175-4. [PMID: 40108054 DOI: 10.1007/s13304-025-02175-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025]
Abstract
The controversy surrounding the continuation of postoperative adjuvant chemotherapy (AC) for locally advanced rectal cancer patients who underwent neoadjuvant therapy (NAT) still existed. The study aimed to identify the individuals that would benefit from AC from those with stage ypII/III rectal cancer. Data for this retrospective study were obtained from the Surveillance, Epidemiology, and End Results (SEER) database and the local database. Subgroup differentiation of the beneficiary population by classification and regression tree analysis. The primary endpoint was overall survival (OS). 15,671 patients were included from the SEER database and 508 patients from local database. The proportions receiving AC were 41.9% in the SEER database and 77.6% in local database, respectively. Analysis results illustrated that the AC benefited population in the SEER database was characterized as: stage ypT4/N + patients (HR 0.75, 95% CI 0.69-0.82, p < 0.001); stage ypT3N0 patients aged 70 years or older (HR 0.69, 95% CI 0.56-0.83, p < 0.001). Moreover, stage ypT4/N + patients also significantly benefited from AC in local database (HR 0.48, 95% CI 0.31-0.74, p < 0.001). The analysis of the two databases showed that stage ypT3N0 patients aged < 70 years could not significantly benefit from AC (HR 0.90, p = 0.114 in the SEER database; HR 0.90, p = 0.960 in local database). Postoperative adjuvant chemotherapy provides a significant benefit in patients with stage ypT4/N + rectal cancer following neoadjuvant therapy. Our study discovered that locally advanced rectal cancer patients with aggressive tumors might benefit from postoperative adjuvant chemotherapy and prolonged the survival.
Collapse
Affiliation(s)
- Pengwen Zheng
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Mengzhen Xu
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China
| | - Dening Ma
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China
| | - Longhai Feng
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Jing Qin
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China.
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China.
| | - Xinyi Gao
- Department of Radiology, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, China.
| |
Collapse
|
250
|
Siqin S, Nikitina E, Rahbari M, Ernst C, Krunic D, Birgin E, Tessmer C, Hofmann I, Rahbari N, Bund T. Bovine Meat and Milk Factor (BMMF) Protein Is Expressed in Macrophages Spread Widely over the Mucosa of Colorectal Cancer Patients. Cells 2025; 14:455. [PMID: 40136704 PMCID: PMC11940877 DOI: 10.3390/cells14060455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Red meat consumption is considered a risk factor for colorectal cancer (CRC) development and stimulated isolation of plasmid-like DNA molecules from bovine serum and milk, termed bovine meat and milk factors (BMMFs). BMMFs encode a conserved replication protein (Rep). Increased populations of Rep-expressing macrophages have been identified in the peritumor of CRC patients and pre-cancerous tissues when compared to the tissues of healthy individuals. This supports the concept that BMMFs increase cancer risk by indirect carcinogenesis, upon induction of chronic inflammation. However, the spread of Rep+ immune cells in tissues at greater distances from primary tumors has not yet been assessed. Here, we immunohistologically analyzed the presence of Rep+ immune cells in sets of tumor, peritumor and, additionally, distant tissues of CRC patients (n = 13). We identified consistently high numbers of BMMF-positive macrophages in mucosal tissues at distances of as much as 25 cm away from the primary tumors, at levels comparable to peritumors and associated with M2-like macrophage polarization. The broad distribution of BMMFs suggests that BMMF+ macrophages might already exist at stages of pre-cancerous dysplasia or before. Quantification of BMMF tissue expression during colonoscopy might help to preventively stratify individuals at risk of developing polyps/CRC and recommend them for enhanced surveillance and/or changes in dietary lifestyle.
Collapse
Affiliation(s)
- Sumen Siqin
- Division of Episomal-Persistent DNA in Cancer- and Chronic Diseases, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ekaterina Nikitina
- Division of Episomal-Persistent DNA in Cancer- and Chronic Diseases, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mohammad Rahbari
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, University of Tuebingen, 72076 Tuebingen, Germany
| | - Claudia Ernst
- Division of Episomal-Persistent DNA in Cancer- and Chronic Diseases, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Damir Krunic
- Light Microscopy Facility, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Emrullah Birgin
- Department of Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Clinic for General and Visceral Surgery, University Hospital Ulm, 89081 Ulm, Germany
| | - Claudia Tessmer
- Core Facility Antibodies, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ilse Hofmann
- Core Facility Antibodies, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Nuh Rahbari
- Department of Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Clinic for General and Visceral Surgery, University Hospital Ulm, 89081 Ulm, Germany
| | - Timo Bund
- Division of Episomal-Persistent DNA in Cancer- and Chronic Diseases, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|