101
|
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
|
102
|
Xu Y, Fu G, Chen X, He R, Fang Q, Tan X, Xu K, Li H, Tan J, Fang M, Fu K, Huang Q, Xiao S. Integrated single-nuclear RNA sequencing analysis reveals distinct characteristics of mucinous adenocarcinoma in right-sided colon cancer. Int J Biol Macromol 2025; 309:142744. [PMID: 40180102 DOI: 10.1016/j.ijbiomac.2025.142744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Mucinous adenocarcinoma (MAC) is a unique histological subtype of colorectal cancer (CRC), which usually occurs in the right-sided colon with poor prognosis. Our previous studies reveled unique clinicopathological characteristic of MAC, but the distinct molecular features and tumor microenvironment (TME) characteristics remain clarify systematically. In this study, we conducted a single nuclear RNA sequencing (snRNA-seq) analysis for CRC tissues with different histological subtypes, including MAC, partial mucinous adenocarcinoma (pMAC) and non-specific adenocarcinoma (AC). Our results show that MAC has a unique transcriptome profile and a distinct single-cell characteristic. It exhibited a higher degree of tumor purity but a lower composition of TME. MAC had a distinct cell-cell communication microenvironment and a particular evolutionary trajectory. The EpithelialCells of MAC closely interacted with fibroblasts, endothelial cells, and mural cells clusters. Furthermore, molecular functional characteristics analysis revealed that MAC enriched EpithelialCells, Fibroblasts, and Endothelial cells clusters have a high active in glycolysis, inflammation, and angiogenesis respectively. This comprehensive study first demonstrated that MAC is a unique histological subtype of right-sided CRC in the single cell level, and elucidated its tumor microenvironment composition and biological function characteristics, which will help us better understand the intrinsic feature of MAC.
Collapse
Affiliation(s)
- Yunhua Xu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Institute of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Guang Fu
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Xiguang Chen
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Rongfang He
- The First Affiliated Hospital, Department of pathology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Qing Fang
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Xiangwen Tan
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The Second Affiliated Hospital, Department of hepatological Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Kunming Xu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Hongming Li
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jie Tan
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Min Fang
- The First Affiliated Hospital, Department of Education and Training, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Kai Fu
- Institute of Molecular Precision Medicine, Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China.
| | - Qiulin Huang
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| | - Shuai Xiao
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Institute of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| |
Collapse
|
103
|
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
|
104
|
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
|
105
|
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
|
106
|
Zhang X, Di Y, Wang Y, Qin J, Ye L, Wen X, Ke Z, Wang Z, He W. SIRT5-mediated desuccinylation of PPA2 enhances HIF-1alpha-dependent adaptation to hypoxic stress and colorectal cancer metastasis. EMBO J 2025:10.1038/s44318-025-00416-1. [PMID: 40164945 DOI: 10.1038/s44318-025-00416-1] [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: 10/08/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Metastasis is the primary cause of death in patients with colorectal cancer (CRC). Hypoxia is a hallmark of solid tumors that promotes cellular metabolic adaptation and dissemination. However, the mechanisms linking hypoxia-regulated metabolic adaptation to CRC metastasis remain unclear. Here, we found that inorganic pyrophosphatase 2 (PPA2) suppresses metastatic progression of CRC via its phosphatase function. PPA2 expression levels are reduced in CRC specimen and correlate with enhanced response to hypoxia by promoting hypoxia-inducible factor-1 (HIF-1) signaling to promote CRC cell glycolysis and dissemination. Mechanistically, PPA2 decreases HIF-1alpha stability through non-canonical ubiquitin-mediated proteasomal degradation via recruitment of E3 ligase NEDD4. Furthermore, PPA2 directly dephosphorylates NEDD4 at threonine 758 residue, resulting in its activation. Under hypoxic stress, NAD-dependent protein deacetylase sirtuin-5 promotes the dissociation of PPA2 and NEDD4 by inducing PPA2 desuccinylation at lysine 176, contributing to the improved stability of HIF-1alpha under hypoxic conditions. Our findings reveal a tumor-suppressive role of PPA2 in HIF-1alpha-dependent colorectal cancer, providing a potential therapeutic target and prognostic strategy.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yuqin Di
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Youpeng Wang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jiale Qin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Lvlan Ye
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiangqiong Wen
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Medical College of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zunfu Ke
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Ziyang Wang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Weiling He
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China.
| |
Collapse
|
107
|
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
|
108
|
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
|
109
|
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
|
110
|
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
|
111
|
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] [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
|
112
|
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] [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
|
113
|
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
|
114
|
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] [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
|
115
|
Birgersson M, Holm M, Gallardo-Dodd CJ, Chen B, Stepanauskaitė L, Hases L, Kutter C, Archer A, Williams C. Intestinal estrogen receptor beta modulates the murine colon tumor immune microenvironment. Cancer Lett 2025:217661. [PMID: 40120798 DOI: 10.1016/j.canlet.2025.217661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Chronic inflammation contributes to the development of colorectal cancer, partly through its regulation of the microenvironment and antitumor immunity. Interestingly, women have a lower incidence of colorectal cancer, and estrogen treatment has been shown to reduce the occurrence of colorectal tumors. While intestinal estrogen receptor beta (ERβ, Esr2) can protect against colitis and colitis-induced cancer in mice, its role in shaping the tumor microenvironment remains unknown. In this study, we performed RNA sequencing to analyze the transcriptome of colonic epithelia and tumors from azoxymethane/dextran sulfate sodium-treated wild-type and intestinal ERβ knockout (ERβKOVil) mice and vehicle-treated controls. This revealed significant differences in gene expression and enriched biological processes influenced by sex and genotype, with immune-related responses being overrepresented. Deconvolution supported differential immune cell abundance and immunostaining showed that tumors from ERβKOVil mice displayed significantly increased macrophage infiltration, decreased T cell infiltration, and impaired natural killer cell infiltration. Further, ERβ mRNA levels in clinical colorectal tumors correlated with immune signaling profiles and better survival. Our findings indicate that intestinal ERβ promotes an antitumor microenvironment and could potentially affect the effectiveness of immunotherapy. These insights highlight the importance of ERβ in modulating antitumor immunity and underscore its therapeutic potential in colorectal cancer.
Collapse
Affiliation(s)
- Madeleine Birgersson
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Matilda Holm
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Carlos J Gallardo-Dodd
- Department of Microbiology, Tumor and Cell Biology, SciLifeLab, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Baizhen Chen
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden
| | - Lina Stepanauskaitė
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Linnea Hases
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Claudia Kutter
- Department of Microbiology, Tumor and Cell Biology, SciLifeLab, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Amena Archer
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Cecilia Williams
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83 Huddinge, Sweden.
| |
Collapse
|
116
|
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
|
117
|
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] [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
|
118
|
Pandey P, Lakhanpal S, Mahmood D, Baldaniya L, Kang HN, Hwang S, Kang S, Choi M, Moon S, Pandey S, Chaudhary K, Khan F, Kim B. Recent Update of Natural Compounds as HIF-1α Inhibitors in Colorectal Carcinoma. Drug Des Devel Ther 2025; 19:2017-2034. [PMID: 40124557 PMCID: PMC11929541 DOI: 10.2147/dddt.s511406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/08/2025] [Indexed: 03/25/2025] Open
Abstract
Hypoxia-inducible factor (HIF)-1 is a transcription factor that regulates the expression of target genes associated with oxygen homeostasis under hypoxic conditions, thereby contributing to tumor development and progression. Accumulating evidence has demonstrated that HIF-1α mediates different biological processes, including tumor angiogenesis, metastasis, metabolism, and immune evasion. Thus, overexpression of HIF-1α is strongly associated with poor prognosis in cancer patients. Natural compounds are important sources of anticancer drugs and studies have emphasized the decisive role of these mediators in modulating HIF-1α. Therefore, the pharmacological targeting of HIF-1α has emerged as a novel cancer therapeutic approach in recent years. The novelty of this review is that it summarizes natural products targeting HIF-1α in colorectal cancer that have not been presented earlier. We studied research publications related to the HIF-1α domain in cancer from 2010 to 2024. However, our main focus was to identify a better targeted approach for colorectal carcinoma management. Our review described HIF-1α role in tumor progression, summarizes the natural compounds employed as HIF-1α inhibitors, and discusses their potential in the development of natural compounds as HIF-1α inhibitors for colorectal cancer treatment.
Collapse
Affiliation(s)
- Pratibha Pandey
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
- Chitkara Centre for Research and Development, Chitkara University, Baddi, Himanchal Pradesh, 174103, India
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Danish Mahmood
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Han Na Kang
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Sungho Hwang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul, 05253, Republic of Korea
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul, 05253, Republic of Korea
| | - Min Choi
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul, 05253, Republic of Korea
| | - Seungjoon Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul, 05253, Republic of Korea
| | - Shivam Pandey
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248007, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, Rajasthan, India
| | - Fahad Khan
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Bonglee Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul, 05253, Republic of Korea
| |
Collapse
|
119
|
Zhang Z, Liu X, Peng C, Du R, Hong X, Xu J, Chen J, Li X, Tang Y, Li Y, Liu Y, Xu C, Liu D. Machine Learning-Aided Identification of Fecal Extracellular Vesicle microRNA Signatures for Noninvasive Detection of Colorectal Cancer. ACS NANO 2025; 19:10013-10025. [PMID: 40033785 DOI: 10.1021/acsnano.4c16698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Colorectal cancer (CRC) remains a formidable threat to human health, with considerable challenges persisting in its diagnosis, particularly during the early stages of the malignancy. In this study, we elucidated that fecal extracellular vesicle microRNA signatures (FEVOR) could serve as potent noninvasive CRC biomarkers. FEVOR was first revealed by miRNA sequencing, followed by the construction of a CRISPR/Cas13a-based detection platform to interrogate FEVOR expression across a diverse spectrum of clinical cohorts. Machine learning-driven models were subsequently developed within the realms of CRC diagnostics, prognostics, and early warning systems. In a cohort of 38 CRC patients, our diagnostic model achieved an outstanding accuracy of 97.4% (37/38), successfully identifying 37 of 38 CRC cases. This performance significantly outpaced the diagnostic efficacy of two clinically established biomarkers, CEA and CA19-9, which showed accuracies of mere 26.3% (10/38) and 7.9% (3/38), respectively. We also examined the expression levels of FEVOR in several CRC patients both before and after surgery, as well as in patients with colorectal adenomas (CA). Impressively, the results showed that FEVOR could serve as a robust prognostic indicator for CRC and a potential predictor for CA. This endeavor aimed to harness the predictive power of FEVOR for enhancing the precision and efficacy of CRC management paradigms. We envision that these findings will propel both foundational and preclinical research on CRC, as well as clinical studies.
Collapse
Affiliation(s)
- Zhaowei Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xuyang Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Chuanyue Peng
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin Institute of Coloproctology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Rui Du
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaoqin Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jia Xu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jiaming Chen
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaomin Li
- Medical and Hygienic Materials Research Institute, SINOPEC (Beijing) Research Institute of Chemical Industry Co., Ltd., Beijing 100013, China
| | - Yujing Tang
- Medical and Hygienic Materials Research Institute, SINOPEC (Beijing) Research Institute of Chemical Industry Co., Ltd., Beijing 100013, China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin Institute of Coloproctology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yang Liu
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Department of Hepatobiliary and Pancreatic Oncology, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin 300308, China
| | - Chen Xu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin Institute of Coloproctology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Dingbin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
120
|
Gu C, Sha G, Zeng B, Cao H, Cao Y, Tang D. Therapeutic potential of fecal microbiota transplantation in colorectal cancer based on gut microbiota regulation: from pathogenesis to efficacy. Therap Adv Gastroenterol 2025; 18:17562848251327167. [PMID: 40104324 PMCID: PMC11915259 DOI: 10.1177/17562848251327167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related deaths worldwide, with its progression intricately linked to gut microbiota dysbiosis. Disruptions in microbial homeostasis contribute to tumor initiation, immune suppression, and inflammation, establishing the microbiota as a key therapeutic target. Fecal microbiota transplantation (FMT) has emerged as a transformative approach to restore microbial balance, enhance immune responses, and reshape the tumor microenvironment. This review explores the mechanisms underlying FMT's therapeutic potential, evaluates its advantages over other microbiota-based interventions, and addresses challenges such as donor selection, safety concerns, and treatment standardization. Looking forward, the integration of FMT into personalized CRC therapies requires robust clinical trials and the identification of predictive biomarkers to optimize its efficacy and safety.
Collapse
Affiliation(s)
- Chen Gu
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Gengyu Sha
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Binbin Zeng
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Herong Cao
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yibo Cao
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dong Tang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou 225000, China
- The Yangzhou Clinical College of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, 221000, China
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, 225000, China
- Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| |
Collapse
|
121
|
Atarere J, Annor E, Bilalaga MM, Egbo O, Gaddipati GN, Vasireddy R, Mensah B, Roberts L. Social media use and the relationship with colorectal cancer screening among foreign-born populations in the United States. Cancer Causes Control 2025:10.1007/s10552-025-01985-6. [PMID: 40100525 DOI: 10.1007/s10552-025-01985-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Social media (SM) has emerged as a tool for health-related usage among US adults, including cancer screening promotion. Here, we aimed to assess the differences in health-related SM use between US and foreign-born adults and the relationship between health-related SM use and colorectal (CRC) screening practices. METHODS Using data from the fifth edition of the National Cancer Institute's Health Information National Trends Survey (HINTS 5), cycle 2, we compared the differences in health-related SM use between US and foreign-born adults and the effects of SM use on CRC screening by country of birth. We included adults aged 50-75 and excluded participants with a history of CRC. The primary endpoint was CRC screening, which was determined by self-reported CRC screening using colonoscopy, sigmoidoscopy, or stool occult blood testing. RESULTS Our study included 1,812 adults, of whom 236 (13.0%) were foreign-born. Most participants (72.1%) reported undergoing CRC screening. Interestingly, we found no discernible difference in health-related SM use [odds ratio [OR] 0.91; 95% CI (0.49, 1.69)] between US and foreign-born adults. Furthermore, our analysis revealed that SM use did not influence CRC screening practices among either group (US-born: 0.88 [95% CI: 0.50, 1.52], foreign-born 0.52 [0.10, 2.51]). CONCLUSION Contrary to previous studies, which showed a positive relationship between SM use and satisfactory health-related practices, we found that although foreign-born adults use SM as much as US-born adults, there was no significant relationship between SM use and CRC screening.
Collapse
Affiliation(s)
- Joseph Atarere
- MedStar Health, MedStar Union Memorial Hospital, 201 E University Pkwy, Baltimore, MD, 21218, USA
| | - Eugene Annor
- University of Illinois College of Medicine, Peoria, IL, USA
| | - Mariah Malak Bilalaga
- MedStar Health, MedStar Union Memorial Hospital, 201 E University Pkwy, Baltimore, MD, 21218, USA.
| | - Olachi Egbo
- Department of Medicine, Aurora Medical Center, Oshkosh, WI, USA
| | - Greeshma N Gaddipati
- MedStar Health, MedStar Union Memorial Hospital, 201 E University Pkwy, Baltimore, MD, 21218, USA
| | - Ramya Vasireddy
- MedStar Health, MedStar Union Memorial Hospital, 201 E University Pkwy, Baltimore, MD, 21218, USA
| | - Boniface Mensah
- MedStar Health, MedStar Union Memorial Hospital, 201 E University Pkwy, Baltimore, MD, 21218, USA
| | - Lewis Roberts
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
122
|
Xu C, Mannucci A, Esposito F, Oliveres H, Alonso-Orduña V, Yubero A, Fernández-Martos C, Salud A, Gallego J, Martín-Richard M, Fernández-Plana J, Guillot M, Aparicio J, Fakih M, Kopetz S, Feliu J, Maurel J, Goel A. An Exosome-Based Liquid Biopsy Predicts Depth of Response and Survival Outcomes to Cetuximab and Panitumumab in Metastatic Colorectal Cancer: The EXONERATE Study. Clin Cancer Res 2025; 31:1002-1015. [PMID: 39820673 PMCID: PMC11913580 DOI: 10.1158/1078-0432.ccr-24-1934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/21/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE The EXOsome and cell-free miRNAs of anti-EGFR ResistAnce (EXONERATE) study was an open-label, biomarker interventional study designed to develop, test, and validate a liquid biopsy predictive of progression-free survival (PFS), overall survival (OS), and objective response rate (ORR) for first-line EGFR inhibitors in metastatic colorectal cancer (mCRC). PATIENTS AND METHODS Patients with newly diagnosed RAS wild-type, chemotherapy-naïve mCRC, both right- and left-sided, were enrolled in two nationwide trials to receive cetuximab or panitumumab along with chemotherapy. The primary endpoint was 12-month PFS, which was hierarchically tested in left- and right-sided mCRCs to predict PFS, OS, and ORR. RESULTS Genome-wide small RNA sequencing identified 12 cell-free and 14 exosomal candidates that were differentially expressed in both plasma and tumor tissue of good versus poor responders (based on PFS <12 months). The 8 and 9 best performing candidates, respectively, were used to generate the EXONERATE assay. In left-sided mCRC, 65% were EXONERATE-high, correlating with shorter median PFS (9.5 vs. 18.5 months; P < 0.001). In the independent right-sided mCRC cohort, 80.8% were EXONERATE-high and experienced a similarly shorter median PFS (8.6 vs. 41.2 months; P = 0.0004). In the right-sided group, EXONERATE predicted PFS ≥12 months with 100% sensitivity. A linear relationship existed between EXONERATE values and response depth. Multivariate analysis revealed that EXONERATE predicts PFS and OS independently of tumor sidedness. CONCLUSIONS The EXONERATE assay robustly predicted PFS and OS outcomes in patients with mCRC, both right- and left-sided, before they received either panitumumab or cetuximab. It stratified PFS, OS, and ORR better than a right versus left approach.
Collapse
Affiliation(s)
- Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Alessandro Mannucci
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Francis Esposito
- Department of Medical Oncology, Hospital Clinic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Helena Oliveres
- Department of Medical Oncology, Hospital Clinic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | - Alfonso Yubero
- Medical Oncology Service, Hospital Universitario Lozano Blesa, Zaragoza, Spain
| | | | - Antonieta Salud
- Medical Oncology Service, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Javier Gallego
- Medical Oncology Service, Hospital General Universitario of Elche, Elche, Spain
| | - Marta Martín-Richard
- Medical Oncology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Mónica Guillot
- Medical Oncology Service, Hospital Son Espases, Palma, Spain
| | - Jorge Aparicio
- Medical Oncology Department, Hospital La Fe de Valencia, Valencia, Spain
| | | | - Scott Kopetz
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jaime Feliu
- Medical Oncology Service, Hospital Universitario La Paz, CIBERONC, Madrid, Spain
| | - Joan Maurel
- Department of Medical Oncology, Hospital Clinic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, IDIBAPS, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California
- City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
123
|
Takahashi H, Hanaoka K, Wada H, Kojima D, Watanabe M. The Current Status of T Cell Receptor (TCR) Repertoire Analysis in Colorectal Cancer. Int J Mol Sci 2025; 26:2698. [PMID: 40141338 PMCID: PMC11943327 DOI: 10.3390/ijms26062698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
The rapid increase in colorectal cancer (CRC) cases recently has highlighted the need to use predictive biomarkers to guide therapeutic approaches. Current studies have focused on the tumor-infiltrating lymphocytes present in the tumor microenvironment (TME), in which cytotoxic T cell activation and the amount are associated with CRC patient prognosis. The T cell receptor (TCR) is essential for antigen recognition and T cell identification, playing a central role in cancer immunotherapy. The T cell status reflects TCR diversity or clonality, known as the TCR repertoire. Accordingly, analyzing the TCR repertoire dynamics may help predict the immunological circumstances of the TME in a timely way. In this review, we summarize the TCR repertoire-related knowledge, including its potential use as predictive biomarkers in CRC. The intratumoral TCR repertoire is restricted in CRC patients compared with healthy individuals, as well as in peripheral blood. Patients with deficient mismatch repair display more restriction than those with proficient mismatch repair. Importantly, a higher TCR diversity before treatment and a decrease following treatment may indicate a good response and a better clinical outcome in CRC patients. The future use of TCR repertoire sequencing technology combined with artificial intelligence-based analysis is a potential strategy for CRC therapeutic decision making.
Collapse
Affiliation(s)
- Hiroyuki Takahashi
- Department of Surgery, Fukuoka University Chikushi Hospital, 1-1-1 Zokumyoin, Chikushino 818-8502, Fukuoka, Japan; (K.H.); (H.W.); (D.K.); (M.W.)
| | | | | | | | | |
Collapse
|
124
|
Liu Z, Chang S, Chen S, Gu R, Guo S. A novel prognostic signature based on m5C‑related LncRNAs and its immunological characteristics in colon adenocarcinoma. Discov Oncol 2025; 16:332. [PMID: 40095128 PMCID: PMC11914420 DOI: 10.1007/s12672-025-02081-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Colon adenocarcinoma (COAD) has high mortality rates due to frequent resistance to treatment. 5-methylcytosine (m5C) is a crucial epigenetic modification of RNA, closely associated with tumorigenesis in various cancers. This study focuses on developing an m5C-related long non-coding RNA (lncRNA) signature to predict prognosis and explore potential therapeutic targets. METHODS Using data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO), we analyzed 18 m5C regulatory genes and their associated lncRNAs in COAD samples. Prognostic lncRNAs were identified through univariate Cox regression, and a risk model was constructed through LASSO regression analyses. Kaplan-Meier survival and receiver operating characteristic analyses were employed to validate the prognostic ability of the signature. Additionally, functional enrichment and immune infiltration analyses were conducted to investigate underlying biological pathways and immune characteristics of the risk groups. Tumor mutation burden and drug sensitivity analyses were also performed. Functional validation of NR2F2-AS1 was conducted through in vitro experiments. RESULTS We established a risk score signature comprising six lncRNAs associated with m5C regulators. Patients were classified into high- and low-risk groups based on the median risk score. This prognostic signature demonstrated significant accuracy and was independent of other clinical features. Immune cell infiltration analysis revealed correlations between the risk signature and various immune cell subtypes. Drug sensitivity analysis indicated the potential therapeutic value of our prognostic signature. Functional experiments confirmed that NR2F2-AS1 acts as a risk factor in the proliferation of colon cancer cells. CONCLUSIONS The m5C-related lncRNA signature serves as a reliable prognostic indicator for colon adenocarcinoma and provides new insights into the tumor immune microenvironment.
Collapse
Affiliation(s)
- Zihe Liu
- Department of Gastroenterology, Jiangsu Funing People's Hospital, Funing, China
| | - Sheng Chang
- Department of Gastroenterology, Jiangsu Funing People's Hospital, Funing, China
| | - Shouguo Chen
- Department of Gastroenterology, Jiangsu Funing People's Hospital, Funing, China
| | - Rong Gu
- Department of Gastroenterology, Jiangsu Funing People's Hospital, Funing, China
| | - Shaoyong Guo
- Department of Gastroenterology, Jiangsu Funing People's Hospital, Funing, China.
| |
Collapse
|
125
|
Zhao Q, Cai D, Xu H, Gao Y, Zhang R, Zhou X, Chen X, Chen S, Wu J, Peng W, Yuan S, Li D, Li G, Nan A. o8G-modified circPLCE1 inhibits lung cancer progression via chaperone-mediated autophagy. Mol Cancer 2025; 24:82. [PMID: 40098195 PMCID: PMC11912650 DOI: 10.1186/s12943-025-02283-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Lung cancer poses a serious threat to human health, but its molecular mechanisms remain unclear. Circular RNAs (circRNAs) are closely associated with tumour progression, and the important role of 8-oxoguanine (o8G) modification in regulating the fate of RNA has been gradually revealed. However, o8G modification of circRNAs has not been reported. We identified circPLCE1, which is significantly downregulated in lung cancer, and further investigated the o8G modification of circPLCE1 and the related mechanism in lung cancer progression. METHODS We identified differentially expressed circRNAs by RNA high-throughput sequencing and then conducted methylated RNA immunoprecipitation (MeRIP), immunofluorescence (IF) analysis, crosslinking immunoprecipitation (CLIP) and actinomycin D (ActD) assays to explore circPLCE1 o8G modification. The biological functions of circPLCE1 in vivo and in vitro were clarified via establishing a circPLCE1 silencing/overexpression system. Tagged RNA affinity purification (TRAP), RNA Immunoprecipitation (RIP) and coimmunoprecipitation (Co-IP) assays, and pSIN-PAmCherry-KFERQ-NE reporter gene were used to elucidate the molecular mechanism by which circPLCE1 inhibits lung cancer progression. RESULTS This study revealed that reactive oxygen species (ROS) can induce circPLCE1 o8G modification and that AUF1 can mediate a decrease in circPLCE1 stability. We found that circPLCE1 significantly inhibited lung cancer progression in vitro and in vivo and that its expression was associated with tumour stage and prognosis. The molecular mechanism was elucidated: circPLCE1 targets the HSC70 protein, increases its ubiquitination level, regulates ATG5-dependent macroautophagy via the chaperone-mediated autophagy (CMA) pathway, and ultimately inhibits lung cancer progression. CONCLUSION o8G-modified circPLCE1 inhibits lung cancer progression through CMA to inhibit macroautophagy and alter cell fate. This study provides not only a new theoretical basis for elucidating the molecular mechanism of lung cancer progression but also potential targets for lung cancer treatment.
Collapse
Affiliation(s)
- Qingyun Zhao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Dunyu Cai
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Haotian Xu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Yihong Gao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Ruirui Zhang
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xiaodong Zhou
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xingcai Chen
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Sixian Chen
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Jiaxi Wu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Wenyi Peng
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Shengyi Yuan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Deqing Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Gang Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| | - Aruo Nan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
126
|
Eid M, Trizuljak J, Taslerova R, Gryc M, Vlazny J, Vilmanova S, Jelinkova M, Homolova A, Tucek S, Hlavsa J, Grolich T, Kala Z, Kral Z, Slaby O. Incidental germline findings during comprehensive genomic profiling of pancreatic and colorectal cancer: single-centre, molecular tumour board experience. Mutagenesis 2025; 40:20-29. [PMID: 38773787 PMCID: PMC11911010 DOI: 10.1093/mutage/geae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/14/2024] [Indexed: 05/24/2024] Open
Abstract
Multidisciplinary molecular tumor boards (MTB) are already well established in many comprehensive cancer centers and play an important role in the individual treatment planning for cancer patients. Comprehensive genomic profiling of tumor tissue based on next-generation sequencing is currently performed for diagnostic and mainly predictive testing. If somatic genomic variants are identified, which are suspected to be pathogenic germline variants (PGVs), MTB propose genetic counseling and germline DNA testing. Commonly used comprehensive genomic profiling approaches of tumor tissue do not include a matched germline DNA control. Therefore, the detection of PGVs could be only predicted based on the content of tumor cells (CTC) in selected tumor area (%) and variant allele frequency score (%). For conclusion, the role of a medical geneticist is essential in these cases. The overall prevalence of PGVs in patients with pancreatic ductal adenocarcinoma (PDAC) and colorectal cancer (CRC) is approximately 10%. In this single-center study, we present 37 patients with PDAC and 48 patients with CRC who were presented at MTB and tested using the large combined DNA/RNA sequencing panel. Content of tumor cells and variant allele frequency scores were evaluated in all tested patients. In case of suspicion of PGV and no previous genetic testing based on the standard guidelines, genetic counseling was recommended regardless of age, sex, and family history. In the PDAC subgroup, five patients were recommended by MTB for genetic counseling based on suspicious genetic findings. Based on a medical geneticist's decision, germline DNA sequencing was performed in four of these cases, and all of them tested positive for PGV in the following genes: ATM, ATM, BRCA1, and BRCA2. In the CRC subgroup, no PGV was confirmed in the two patients genetically tested based on the MTB recommendations. Furthermore, we present data from our center's registry of patients with PDAC and CRC who underwent genetic counseling and germline DNA testing based on the standard screening criteria. Our data confirm that comprehensive genomic profiling of tumor tissue can identify patients with hereditary forms of PDAC, who could remain unidentified by standard screening for hereditary forms of cancer.
Collapse
Affiliation(s)
- Michal Eid
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jakub Trizuljak
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Renata Taslerova
- Department of Pathology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martin Gryc
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jakub Vlazny
- Department of Pathology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sara Vilmanova
- Department of Pathology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martina Jelinkova
- Department of Pathology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Alena Homolova
- Department of Pathology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Stepan Tucek
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Hlavsa
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tomas Grolich
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zdenek Kala
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zdenek Kral
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Ondrej Slaby
- Department of Pathology, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
127
|
Gu J, Wang Y, Zhang H, Gu H, Zhu H. SIGLEC1 has the potential to be an immune-related prognostic indicator in colon adenocarcinoma: a study based on transcriptomic data and Mendelian randomization analysis. Discov Oncol 2025; 16:324. [PMID: 40088346 PMCID: PMC11910455 DOI: 10.1007/s12672-025-02093-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/07/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Colonic adenocarcinoma (COAD) is the most common pathological type of colon cancer. Tumor microenvironment (TME) plays an important role in the occurrence and development of COAD. There are currently no specific studies indicating the mechanism of action of TME in COPD patients. METHODS The percentage of tumor-infiltrating immune cells (TIC) in 512 COAD cases from The Cancer Genome Atlas (TCGA) database was calculated using CIBERSORT and ESTIMATE. Weighted gene coexpression network analysis (WGCNA) was performed to find modules of differentially expressed genes (DEGs) with high correlations followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses to determine the function of distant metastasis (M)-stage-related modules. Pathway enrichment analysis, protein-protein interaction (PPI) network, Cox regression analysis, and Kaplan-Meier survival analysis were performed on DEGs to select the most critical genes. The correlation between SIGLEC1 expression in COAD and TME status and between immune checkpoints and SIGLEC1 was examined using gene set enrichment analysis (GSEA) and Pearson correlation coefficients. RESULTS A WGCNA screen was performed to obtain 12,342 DEGs and 209 key genes associated with M stage between tumor and normal samples. GO and KEGG analysis revealed that the DEGs primarily engaged in pathways such as Th1 and Th2 cell differentiation and cell adhesion molecules. SIGELEC1 gene was identified by univariate Cox regression, PPI network construction, and survival analysis. GSEA showed that the genes in the high-expression SIGLEC1 group were mainly enriched in immune-related activities. In the low-expression SIGLEC1 group, the genes were enriched in MYC targets. CIBERSORT analysis of the proportion of TICs showed that SIGLEC1 was positively correlated with macrophages (M0, M2), T-cell CD8 and immune checkpoint-related genes, suggesting that SIGLEC1 may be responsible for maintaining the immune dominance of TME. Immunohistochemical and prognostic analysis showed that the group with higher SIGLEC1 expression had more severe lesions and a worse prognosis than the group with lower SIGLEC1 expression. CONCLUSIONS SIGLEC1 gene is a distant metastasis-related gene that affects the survival prognosis of COAD patients and provides additional insight into the treatment of COAD.
Collapse
Affiliation(s)
- Jue Gu
- Cardiovascular Department Affiliated Hospital of Nantong University, Nantong, China
| | - Yaxuan Wang
- Cancer Research Center Nantong, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Hui Zhang
- Cancer Research Center Nantong, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Haijuan Gu
- Cancer Research Center Nantong, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong, China.
| | - Haixia Zhu
- Cancer Research Center Nantong, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong, China.
| |
Collapse
|
128
|
Zhu G, Kang Y, Luo M, Ju L, Sun Y, Chen L. Identification of a glycolysis-associated lncRNA signature to predict survival of patients with colorectal cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2025; 18:110-122. [PMID: 40226108 PMCID: PMC11982773 DOI: 10.62347/rvly4737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/16/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVE Colorectal cancer (CRC) still has a poor prognosis and is one of the most common malignancies worldwide. Recently, a close correlation between glycolysis and the progression of CRC has been reported. Hence, explorations of the prognostic value of glycolysis-associated long noncoding RNAs in CRC patients are urgently needed. This study aimed to investigate the role of glycolysis-associated lncRNAs for predicting the prognosis and treatment response of CRC, thereby identifying more biomarkers for CRC. METHODS RNA sequencing (RNA-seq) data for CRC from The Cancer Genome Atlas database were used. A glycolysis-associated long noncoding RNA (lncRNA) signature was estimated by Cox regression analysis, and its predictive capacity was assessed by constructing a receiver operating characteristic (ROC) curve and performing a gene set enrichment analysis. RESULTS One of our constructed glycolysis-related clusters was strongly correlated with an immunosuppressive tumor environment. Moreover, a signature consisting of 14 glycolysis-associated lncRNAs was used as a prognostic model, and CRC patients were classified into a low-risk group and a high-risk group based on the average risk score of this signature. In addition, the low-risk group experienced longer overall survival (OS) than the high-risk group. The area under the ROC curve (AUC) validated the sensitivity and specificity of the signature. The signature was identified as an individual element and was closely related to the progression of CRC. Finally, two glycolysis-associated lncRNAs, namely, TNFRSF10A-AS1 and ZKSCAN2-DT, were further clinically verified to effectively predict the prognosis of CRC patients. CONCLUSION Glycolysis-associated lncRNAs may be employed as prognostic and therapeutic biomarkers for CRC.
Collapse
Affiliation(s)
- Gaoceng Zhu
- Department of Clinical Laboratory Medicine, Affiliated Mental Health Center of Nantong University, Nantong Fourth People’s HospitalNantong 226005, Jiangsu, China
| | - Yutian Kang
- Department of Clinical Laboratory Diagnostics, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Mei Luo
- Department of Clinical Laboratory Diagnostics, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Linling Ju
- Nantong Institute of Liver Diseases, Nantong Third People’s Hospital Affiliated Nantong Hospital 3 of Nantong UniversityNantong 226006, Jiangsu, China
| | - Yajun Sun
- Department of Clinical Laboratory Medicine, Affiliated Mental Health Center of Nantong University, Nantong Fourth People’s HospitalNantong 226005, Jiangsu, China
| | - Lin Chen
- Department of Clinical Laboratory Diagnostics, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
- Nantong Institute of Liver Diseases, Nantong Third People’s Hospital Affiliated Nantong Hospital 3 of Nantong UniversityNantong 226006, Jiangsu, China
| |
Collapse
|
129
|
Li X, Wu M, Wu Y, Xin Y, Gao L, Elsabahy M, Wang X, Zhang J, Qu X, Gao H. Multifunctional nanodrug for simultaneously combating chemoresistance and immunosuppression in Fusobacterium nucleatum-associated colorectal cancer. Acta Biomater 2025; 195:406-420. [PMID: 39922512 DOI: 10.1016/j.actbio.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Fusobacterium nucleatum (Fn) infection in colorectal cancer (CRC) induces chemoresistance and creates an immunosuppressive tumor microenvironment, compromising the efficacy of conventional chemotherapy. To address these challenges, a multifunctional MPLO@HA nanodrug was developed by conjugating metformin (Met), oxaliplatin (OxPt), and lauric acid (LA) onto oligomethyleneimine, subsequently complexed with hyaluronic acid (HA). The MPLO@HA nanodrug is designed to target Fn-infected CRC, offering multiple mechanisms for enhanced therapeutic outcomes. The nanodrug features a multi-stimuli responsive structure that enables precise and controlled release at the tumor site, responsive to pH, glutathione, and hyaluronidase levels. The enhanced positive charge of self-assembled nanodrug combined with Met effectively eradicates both extracellular and intracellular Fn, overcoming Fn-induced chemoresistance. Furthermore, incorporating Met improves the efficacy of chemotherapy by sensitizing CRC cells to treatment. The immunomodulatory properties of the MPLO@HA nanodrug promote immunogenic cell death, repolarize macrophages from the M2 to the M1 phenotype, and reduce the levels of regulatory T cells and myeloid-derived suppressor cells. By integrating antimicrobial, chemotherapeutic, and immunomodulatory capabilities, the MPLO@HA nanodrug offers a promising and comprehensive approach to combating Fn-induced chemoresistance and immunosuppression in CRC. This strategy could also provide a foundation for developing treatments for other cancers associated with bacterial infections. STATEMENT OF SIGNIFICANCE: Fusobacterium nucleatum (Fn) infection in colorectal cancer (CRC) induces chemoresistance and creates an immunosuppressive tumor microenvironment, severely compromising treatment efficacy. Current therapies face challenges in addressing these issues due to the complex interactions between bacterial infection and tumor development. Our study introduces a multifunctional nanodrug, MPLO@HA, which integrates metformin, oxaliplatin, lauric acid, and hyaluronic acid into a multi-responsive nanodrug system. This nanodrug simultaneously combats bacterial infection, chemoresistance, and immunosuppression in Fn-associated CRC. MPLO@HA demonstrates synergistic effects by eradicating both extracellular and intracellular Fn, enhancing chemosensitivity, and modulating the tumor immune microenvironment. This comprehensive approach offers a promising strategy to overcome Fn-induced treatment barriers, potentially improving outcomes for patients with Fn-infected CRC and opening new avenues in bacteria-associated cancer therapy.
Collapse
Affiliation(s)
- Xiaohui Li
- State Key Laboratory of Separation Membranes and Membrane Processes& Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China
| | - Mengdi Wu
- State Key Laboratory of Separation Membranes and Membrane Processes& Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China
| | - Yu Wu
- State Key Laboratory of Separation Membranes and Membrane Processes& Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China
| | - Youtao Xin
- State Key Laboratory of Separation Membranes and Membrane Processes& Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China
| | - Linran Gao
- State Key Laboratory of Separation Membranes and Membrane Processes& Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China
| | - Mahmoud Elsabahy
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, 11829, Egypt
| | - Xuan Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Jimin Zhang
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300401, China
| | - Xiongwei Qu
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300401, China
| | - Hui Gao
- State Key Laboratory of Separation Membranes and Membrane Processes& Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE) & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China.
| |
Collapse
|
130
|
Lu YJ, Chen CH, Lin EK. In-hospital outcomes of percutaneous ablative therapy for colorectal cancer liver metastasis in patients with and without frailty: nationwide inpatient sample analysis 2005-2020. Am J Cancer Res 2025; 15:1280-1290. [PMID: 40226451 PMCID: PMC11982708 DOI: 10.62347/mqxg6358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/07/2025] [Indexed: 04/15/2025] Open
Abstract
Percutaneous ablative therapies are widely used to treat colorectal liver metastases (CRLM), particularly in patients who are not candidates for surgical resection. Frailty has been associated with poor outcomes in colorectal cancer (CRC) and liver resections. This study aimed to evaluate the clinical impact of frailty on short-term outcomes in patients undergoing percutaneous ablative therapies for CRLM. This population-based, retrospective study used data from the US Nationwide Inpatient Sample database (2005-2020). Adults aged ≥ 50 years diagnosed with CRLM who underwent percutaneous ablative therapies were included. Frailty was confirmed using the Hospital Frailty Risk Score (HFRS). Associations between frailty and in-hospital mortality, length of hospital stay (LOS), non-home discharge, total hospital charges, and postoperative complications were evaluated using univariate and multivariable regression analyses. A total of 670 patients (mean age: 66.3 years) were included, of whom 23% were categorized as frail (HFRS ≥ 5). Multivariable analysis showed that frail patients had significantly increased risks of complications (adjusted odds ratio [aOR] = 4.80, 95% confidence interval [CI]: 3.04-7.59), longer LOS (adjusted Beta [aBeta] = 1.69 days, 95% CI: 1.68-1.70), and higher total hospital charges (aBeta = $22.04 thousand, 95% CI: $21.92-$22.16). Complications with the highest risks in frail patients included, sepsis/shock (aOR = 17.39), surgical site infection (aOR = 3.55), respiratory failure/mechanical ventilation (aOR = 4.43), acute kidney injury (aOR = 9.37), and bleeding (aOR = 4.79). In conclusion, in adults aged ≥ 50 years undergoing percutaneous ablative therapies for CRLM, frailty independently predicted worse short-term outcomes, including higher complication rates, longer LOS, and increased hospital charges. The absence of detailed tumor characteristics and specific types of ablative therapy performed underscores the need for further research.
Collapse
Affiliation(s)
- Yen-Jung Lu
- Division of Colorectal Surgery, Department of Surgery, Wanfang Hospital, Taipei Medical UniversityTaipei, Taiwan
- Graduate Institute of Clinical Medicine, School of Medicine, College of Medicine, Taipei Medical UniversityTaipei, Taiwan
| | - Chien-Hsin Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical UniversityTaipei, Taiwan
| | - En-Kwang Lin
- Division of Colorectal Surgery, Department of Surgery, Wanfang Hospital, Taipei Medical UniversityTaipei, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung UniversityTaipei, Taiwan
| |
Collapse
|
131
|
Liu S, Hu L, Zhang X. Factors associated with lymph node metastasis and survival in T2 colon cancer. BMC Gastroenterol 2025; 25:175. [PMID: 40087580 PMCID: PMC11909863 DOI: 10.1186/s12876-025-03748-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/28/2025] [Indexed: 03/17/2025] Open
Abstract
PURPOSE This study aimed to explore the clinical factors associated with lymph node metastasis (LNM) and survival in T2 colon cancer. METHOD Patients with T2 colon cancer and receiving radical surgery from 2017 to 2021 in our hospital were retrospectively enrolled. Patients were divided into two groups according to the LN status (LNM, non-LNM). The demographic, radiological, pathological, and survival data were collected and analyzed. Logistic regression was used to find the factors associated with LNM, and cox regression was adopted to identify factors contributing to poor survival. All the data analysis was performed by SPSS 22.0 and R. RESULTS A total of 150 patients were included in this study, among them thirty were with LNM (20%). The LNM group had significantly higher incidence of lymph-vascular invasion (LVI) and perineural invasion. Besides, positive LNs had more proportion of irregular margin (P < 0.001) and heterogeneity (P < 0.001) than the negative ones. The multivariate analysis indicated that LVI and heterogeneity of LN were independent risk factors of LNM in T2 colon cancer. The disease-free survival (DFS) was 80% and 93.3% in the LNM and non-LNM group (P = 0.02), respectively. Besides, the overall survival (OS) was 92.9% and 95% in the LNM and non-LNM group (P = 0.103), respectively. The results indicated that elevated CA199 value and LNM were independent risk factors contributing to poorer OS and DFS. CONCLUSION The current data indicated LVI and LN heterogeneity were independent risk factors of LNM in T2 colon cancer. More extended surgery should be considered when these factors were detected.
Collapse
Affiliation(s)
- Shaojun Liu
- Department of Colorectal Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Lei Hu
- Department of Colorectal Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Xubing Zhang
- Department of Colorectal Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
132
|
Haveric A, Balogun F, Gilliland J, Narang B, Gany F. Fecal Immunochemical Tests (FIT) and Focus Groups: Tailoring Bilingual Cancer Screening Education. J Community Health 2025:10.1007/s10900-025-01454-w. [PMID: 40082309 DOI: 10.1007/s10900-025-01454-w] [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] [Accepted: 02/16/2025] [Indexed: 03/16/2025]
Abstract
Virtual focus groups were conducted in English and in French to gather community feedback on colorectal cancer screening education materials, designed for use among immigrant and non-immigrant Black/African-American men in New York City. Participants were recruited from community health fair events, religious and cultural organizations, and an existing Community Advisory Board. Six total focus groups were conducted, four in English and two in French, with a total of 25 participants, until data saturation. Focus groups were audio recorded, transcribed, two were translated from French into English, and analyzed in NVivo software to develop and consolidate themes in participant responses. Focus groups revealed key knowledge gaps about colorectal cancer screening protocols and cancer risk heritability. Discussing racial health disparities may cause offense to some readers. Participants were interested to see root causes for racial health disparities discussed in health promotion material. Virtual focus groups can be used to gather community member feedback on health promotion materials. Real-time simultaneous language interpreting can be used if facilitator and audience do not speak the same language. Health educators should be mindful that the recommended screening age of 45 years for colorectal cancer is not generally known, and that individuals may believe that cancer is contagious. Readers may be offended by references to health disparities. Health educators should test health promotion materials with target audiences to ensure appropriateness and cultural sensitivity.
Collapse
Affiliation(s)
- Arman Haveric
- The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
- Immigrant Health and Cancer Disparities Service, Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fiyinfolu Balogun
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering, New York, NY, USA
| | - Jaime Gilliland
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bharat Narang
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francesca Gany
- Immigrant Health and Cancer Disparities Service, Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
- , 633 3rd Avenue, 4th Floor, New York, NY, 10017, USA.
| |
Collapse
|
133
|
Zhang H, Han Y, Wu C, Wang S, Chen M, Xu Q, Wei H, Zhou X, Wang G. m6A-modified LINC02418 induces transcriptional and post-transcriptional modification of CTNNB1 via interacting with YBX1 and IGF2BP1 in colorectal cancer. Cell Death Discov 2025; 11:101. [PMID: 40082414 PMCID: PMC11906587 DOI: 10.1038/s41420-025-02365-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 03/16/2025] Open
Abstract
Colorectal cancer (CRC) represents a significant menace to human health, but its molecular pathogenesis remains unclear. Herein, we explored the functional role of LINC02418 in CRC progression. The function of LINC02418 in CRC was determined through vitro and in vivo experiments. The molecular mechanism of LINC02418 in CRC was explored by quantitative real-time PCR (qPCR) analyses, western blot, luciferase reporter assay, methylated RNA immunoprecipitation (MeRIP) assay, RNA pull-down, RNA immunoprecipitation (RIP) assay and chromatin immunoprecipitation (ChIP) assay. The results revealed that LINC02418 expression was upregulated in CRC tissues and the high expression of LINC02418 was related to unfavorable survival of CRC patients. Besides, knockdown of LINC02418 expression resulted in the inhibition of proliferation and metastasis of CRC cells in vitro and in vivo. Mechanistically, we found METTL3-mediated m6A modification induced the aberrant expression of LINC02418 in CRC. LINC02418 could interact with YBX1 and enhance YBX1 DNA-binding ability to the CTNNB1 promoter, resulting in transcriptional activation of CTNNB1. In the post-transcriptional stage, LINC02418 could also enhance CTNNB1 stability by promoting the interaction between IGF2BP1 protein and CTNNB1 mRNA. What is more, LINC02418 expression could be transcriptionally enhanced by YBX1 protein. Collectively, this study unveils a novel oncogenic mechanism for LINC02418 in CRC and the LINC02418 might be a novel therapeutic target in CRC treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Ye Han
- In-Patient Ultrasound Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Chengwei Wu
- In-Patient Ultrasound Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Siying Wang
- In-Patient Ultrasound Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Mingquan Chen
- In-Patient Ultrasound Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Qian Xu
- In-Patient Ultrasound Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Hong Wei
- In-Patient Ultrasound Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Xianli Zhou
- In-Patient Ultrasound Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China.
| | - Guiyu Wang
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China.
| |
Collapse
|
134
|
Liao YS, Chiu HY, Huang FH, Chang YH, Huang YM, Wei PL, Wang W, Hung CS, Tung HH. Prehabilitation Interventions in Patients Undergoing Colorectal Cancer Surgery: A Systematic Review and Meta-Analysis. J Am Geriatr Soc 2025. [PMID: 40079672 DOI: 10.1111/jgs.19425] [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: 08/12/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Surgical resection is the primary treatment modality for colorectal cancer. Prehabilitation is about enhancing the patient's physiological capacity preoperatively to reduce the risk of treatment-related complications. Clear definitions of the modality, content, and duration of prehabilitation, including its components such as nutrition, exercise, and psychological support, are lacking. Some review articles have proposed that a multimodal approach may yield the best overall outcomes, but the clinical efficacy of such an approach requires further exploration. OBJECTIVE This study consisted of a systematic review and meta-analysis to investigate the effectiveness of multimodal prehabilitation programs for patients undergoing colorectal surgery. METHODS We searched PubMed, Embase, CINAHL, and the Cochrane Library from inception to August 5, 2023, without language or publication period restrictions. The included studies were randomized controlled trials, prospective studies, or retrospective studies that examined the effectiveness of multimodal prehabilitation programs for patients undergoing colorectal surgery. A random-effects model was used for data analysis. RESULTS This study included 14 articles that analyzed data from 2314 patients who underwent colorectal cancer surgery. In comparisons against a control group, multimodal prehabilitation significantly reduced the length of hospital stay ([mean difference; MD] = -2.47 days, 95% confidence interval [CI] [-3.56, -1.39]), postoperative complication rate (odds ratio; [OR] = 0.74, 95% CI [0.59, 0.94]), and time to the first passage of flatus (MD = -0.43 days, 95% CI [-0.66, -0.20]). CONCLUSION Multimodal prehabilitation interventions before colorectal cancer surgery reduce hospital stay lengths, lower complication rates, and promote bowel recovery, particularly in older populations.
Collapse
Affiliation(s)
- Yi-Shu Liao
- College of Nursing, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Nursing, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hsiao-Yean Chiu
- Department of Nursing, Taipei Medical University Hospital, Taipei, Taiwan
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan
| | - Fu-Huan Huang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, Division of Pediatric Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yu-Han Chang
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan
| | - Yu-Min Huang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, Division of Gastrointestinal Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Po-Li Wei
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, Division of Colorectal Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Weu Wang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, Division of Gastrointestinal Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chin-Sheng Hung
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, Division of Breast Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Heng-Hsin Tung
- College of Nursing, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
135
|
Liu QL, Zhou H, Wang Z, Chen Y. Exploring the role of gut microbiota in colorectal liver metastasis through the gut-liver axis. Front Cell Dev Biol 2025; 13:1563184. [PMID: 40181829 PMCID: PMC11965903 DOI: 10.3389/fcell.2025.1563184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Colorectal liver metastasis (CRLM) represents a major therapeutic challenge in colorectal cancer (CRC), with complex interactions between the gut microbiota and the liver tumor microenvironment (TME) playing a crucial role in disease progression via the gut-liver axis. The gut barrier serves as a gatekeeper, regulating microbial translocation, which influences liver colonization and metastasis. Through the gut-liver axis, the microbiota actively shapes the TME, where specific microbial species and their metabolites exert dual roles in immune modulation. The immunologically "cold" nature of the liver, combined with the influence of the gut microbiota on liver immunity, complicates effective immunotherapy. However, microbiota-targeted interventions present promising strategies to enhance immunotherapy outcomes by modulating the gut-liver axis. Overall, this review highlights the emerging evidence on the role of the gut microbiota in CRLM and provides insights into the molecular mechanisms driving the dynamic interactions within the gut-liver axis.
Collapse
Affiliation(s)
- Qiu-Luo Liu
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Institute of Digestive Surgery, Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Huijie Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Health Management Center, General Practice Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ziqiang Wang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Chen
- Department of Gastrointestinal Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
136
|
Peng Q, Shen Y, Xu Y, Feng Z, Xu Y, Wang Y, Zou L, Zhu Y, Shen Y. Association of metabolic dysregulation with treatment response in rectal cancer patients undergoing chemoradiotherapy. BMC Med Genomics 2025; 18:48. [PMID: 40075399 PMCID: PMC11905535 DOI: 10.1186/s12920-025-02114-7] [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: 11/08/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND This study aimed to explore the metabolic changes during neoadjuvant chemoradiotherapy (NCRT) in patients with locally advanced rectal cancer (LARC) by serum metabolomics analysis, and to provide new biomarkers for individualized treatment and efficacy prediction. METHODS Serum samples from 20 patients with LARC before, during and after NCRT were collected for metabolomic analysis. The metabolites in the serum samples were analyzed qualitatively and quantitatively using gas chromatography-mass spectrometry (GC-MS). Meanwhile, the differences in metabolic profiles at different time points were compared and significantly changed metabolites were screened. RESULTS The metabolic profiles of patients were significantly altered at different time points of NCRT. Through metabolomic analysis, we identified metabolites that were significantly altered during NCRT and revealed alterations in the associated metabolic pathways. The predictive power of pre-radiotherapy isocitric acid and pro-radiotherapy 3-hydroxy-3-(4'-hydroxy-3'-methoxyphenyl) propionic acid in distinguishing patients sensitive and non-sensitive to NCRT was markedly high, with AUC values of 0.875 and 0.75, respectively. Additional analysis indicated that a combined panel of serum metabolites yielded even higher AUC values, thereby enhancing the accuracy of predicting the efficacy of neoadjuvant NCRT. CONCLUSION This study revealed metabolic changes and corresponding alterations in metabolic pathways during NCRT in patients with LARC by serum metabolomic analysis. The metabolic disorders may be associated with poor outcomes in patients treated with NCRT for rectal cancer, providing new biomarkers for individualized treatment and prognostic assessment. Further studies and validation will help to gain insight into the mechanism of these metabolic changes and provide more basis for clinical application.
Collapse
Affiliation(s)
- Qiliang Peng
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu, 215004, China
- Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Yi Shen
- Department of Radiation Oncology, Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Yingying Xu
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu, 215004, China
- Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Zhengyang Feng
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yao Xu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yong Wang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu, 215004, China
- Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Li Zou
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu, 215004, China
- Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Yaqun Zhu
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu, 215004, China.
- Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.
| | - Yuntian Shen
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu, 215004, China.
- Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.
| |
Collapse
|
137
|
Zhang P, Feng S, Liu F, Han S, Fan T, Chen H, Dong X, Wang X, Qin Y, Chen Y, Jiang Y. Cascaded Strand Displacement Amplification and CRISPR/Cas12a Aptasensor Utilizing MoS 2 Nanoflowers for Colorectal Cancer Biomarker Porphyromonas gingivalis Detection. Anal Chem 2025; 97:4932-4944. [PMID: 40016920 DOI: 10.1021/acs.analchem.4c05014] [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: 03/01/2025]
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer globally, both in terms of diagnoses and cancer-related mortality. Increasing evidence suggests that an imbalance in intestinal flora can contribute to the progression of CRC, and fecal microbiota may serve as potential biomarkers for its screening and diagnosis. Notably, Porphyromonas gingivalis has been identified in the malignant tissues and feces of CRC patients, establishing it as a significant biomarker for early screening, diagnosis, and prognostic assessment of CRC. Current methods for detecting P. gingivalis face numerous challenges, including high costs, complex procedures, and lengthy implementation times. Therefore, developing rapid, highly specific, and sensitive detection methods for P. gingivalis is of great importance. In this study, we utilized the whole-bacterium systematic evolution of ligands by exponential enrichment method to identify highly specific and high-affinity aptamers targeting P. gingivalis through 15 selection cycles. Subsequently, we developed an aptasensor driven by MoS2 nanoflowers, which integrates strand displacement amplification and CRISPR/Cas12a double amplification for sensitive detection of P. gingivalis, achieving a limit of detection of 10 CFU/mL. Using this aptasensor, we evaluated the abundance of P. gingivalis in clinical fecal samples and observed significantly higher levels in the feces of CRC patients compared to healthy individuals, corroborating the results obtained from quantitative polymerase chain reaction. In summary, we developed a highly specific and sensitive aptasensor for the first time, representing a promising new approach for the identification of P. gingivalis, with significant potential for CRC screening and diagnosis.
Collapse
Affiliation(s)
- Peiyi Zhang
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, P. R. China
| | - Shanshan Feng
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, P. R. China
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, P. R. China
| | - Feng Liu
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, P. R. China
| | - Sanyang Han
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, P. R. China
| | - Tingting Fan
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518000, P. R. China
| | - Hui Chen
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, P. R. China
| | - Xiangyan Dong
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, P. R. China
| | - Xiaopeng Wang
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, P. R. China
| | - Ying Qin
- Department of Gastrointestinal Surgery, Shenzhen Second People's Hospital, Shenzhen 518035, P. R. China
| | - Yan Chen
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, P. R. China
| | - Yuyang Jiang
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, P. R. China
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, P. R. China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
138
|
Meng N, Wang Z, Peng Y, Wang X, Yue W, Wang L, Ma W. Analysis of the predictive postoperative recurrence performance of three lymph node staging systems in patients with colon cancer. Front Oncol 2025; 15:1545082. [PMID: 40134603 PMCID: PMC11932909 DOI: 10.3389/fonc.2025.1545082] [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: 12/14/2024] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Background Colon cancer remains a major cause of cancer-related deaths worldwide, with recurrence post-surgery, posing a significant challenge. Accurate lymph node (LN) staging is critical for prognosis and treatment decisions, but traditional systems, such as the AJCC TNM, often fail to predict recurrence. This study compares the prognostic performance of three LN staging systems Lymph Node Ratio (LNR), Log Odds of Metastatic Lymph Nodes (LODDS), and pN in colon cancer. Methods We retrospectively analyzed data from 812 colon cancer patients who underwent radical surgery at two tertiary hospitals (2010-2019). LNR, LODDS, and pN were calculated, and their ability to predict postoperative recurrence was assessed using C-index, AIC, BIC, and ROC curves. Machine learning models (LASSO, Random Forest, XGBoost) identified the most predictive staging system. A nomogram was developed integrating the best staging system with clinical factors to predict postoperative recurrence. Results The study identified LNR as the most predictive staging system for colon cancer. The nomogram based on LNR, along with other variables such as T stage and tumor grade, demonstrated superior predictive performance compared to individual staging systems. In the training cohort, the nomogram achieved an AUC of 0.791 at 1 year, 0.815 at 3 years, and 0.789 at 5 years. The C-index for the nomogram was 0.788, higher than that of LNR (C-index = 0.694) and tumor stage (C-index = 0.665). The nomogram successfully stratified patients into high- and low-risk groups, with higher risk scores correlating with poorer survival outcomes. The validation cohort confirmed the robustness of the model, showing that patients with lower risk scores had better prognoses. Conclusions LNR is an effective predictor of recurrence and prognosis in colon cancer. The nomogram developed from LNR and other clinical factors offers superior prognostication and can aid in personalized treatment strategies.
Collapse
Affiliation(s)
- Ning Meng
- Department of General Surgery, Shijiazhuang People’s Hospital, Shijiazhuang, Hebei, China
| | - Zhiqiang Wang
- Department of General Surgery, Shijiazhuang People’s Hospital, Shijiazhuang, Hebei, China
| | - Yaqi Peng
- Basic College, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoyan Wang
- Department of General Surgery, Shijiazhuang People’s Hospital, Shijiazhuang, Hebei, China
| | - Wenju Yue
- Department of General Surgery, Shijiazhuang People’s Hospital, Shijiazhuang, Hebei, China
| | - Le Wang
- Department of General Surgery, Shijiazhuang People’s Hospital, Shijiazhuang, Hebei, China
| | - Wenqian Ma
- Department of Endoscopy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
139
|
Heng Y, Huang M, Xu J, Wu X, Huang N, Cao Y, Qin L. Prognostic value of tumor deposits and positive lymph nodes in colorectal cancer surgery: improved staging for long-term prognosis. BMC Gastroenterol 2025; 25:154. [PMID: 40069628 PMCID: PMC11899793 DOI: 10.1186/s12876-025-03713-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND To evaluate the prognostic value of the presence and number of tumor deposits (TDs) and the combination of TDs and number of positive lymph nodes (PLNs) in patients undergoing colorectal cancer (CRC) surgery, and to modify N staging. METHOD The clinical data of 1470 patients with stage I-IV CRC who underwent surgery in Wuhan Union Hospital from February 2014 to May 2018 were collected. The optimal cutoff value for TD + PLNs was obtained using X-tile software, and patients were regrouped accordingly. Cox univariate and multivariate analysis were used to screen the factors affecting the prognosis of patients. The receiver operating characteristic (ROC) curve and the area under the curve (AUC) were used to evaluate the predictive ability of independent prognostic factors for overall survival (OS) and disease-free survival (DFS) of patients. RESULT The presence of TD was associated with poor OS (HR = 2.478, 95%CI: 1.794-3.422, P<0.001) and DFS (HR = 2.516, 95%Cl: 1.874-3.377, P<0.001). Combined with TD and PLNs, a total of 128 of 395 N1 patients were reclassified re-staged as N2(TD + PLNs ≥ 3), which had a worse prognosis than those diagnosed with N1. Compared with Tumor Node Metastasis stage and TD number, the multivariate model constructed using independent prognostic factors showed better predictive power for OS (AUC:0.769 vs. 0.681 vs. 0.650) and DFS (AUC:0.757 vs. 0.702 vs. 0.650). CONCLUSION TD significantly affects the long-term prognosis of CRC patients. Combining TD and PLNs to redefine the tumor staging of CRC patients can improve the accuracy of long-term prognosis of surgical patients.
Collapse
Affiliation(s)
- Yixin Heng
- Department of General Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, 832008, P. R. China
| | - Mudan Huang
- Department of Radiation Oncology, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, 5 18000, China
| | - Jiaxin Xu
- Department of General Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, 832008, P. R. China
| | - Xiaoyu Wu
- Department of General Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, 832008, P. R. China
| | - Ning Huang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Yinghao Cao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No. 1277, Wuhan, Hubei Province, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Le Qin
- Department of General Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, 832008, P. R. China.
| |
Collapse
|
140
|
Xu J, Cai Z, Pang Z, Chen J, Zhu K, Wang D, Tu J. Smilax glabra Flavonoids Inhibit AMPK Activation and Induce Ferroptosis in Obesity-Associated Colorectal Cancer. Int J Mol Sci 2025; 26:2476. [PMID: 40141120 PMCID: PMC11942472 DOI: 10.3390/ijms26062476] [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: 12/26/2024] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Smilax glabra flavonoids (SGF), the active components of Smilax glabra Roxb., have been demonstrated to exhibit antioxidant activity and metabolic benefits in obesity, leading us to further explore their antitumor effects in obesity-related colorectal cancer (CRC). This study investigated the antiproliferative effects of SGF on obesity-related CRC by using a murine colon adenocarcinoma MC38 cell line. The underlying mechanisms were further explored via RNA-Seq and bioinformatics analysis in combination with experimental validation. SGF was proven to possess cytotoxic effects against MC38 cells, indicated by the inhibition of proliferation and migration, especially in an adipocyte-rich environment. In line with this, SGF exhibited much stronger antiproliferative effects on MC38-transplanted tumors in obese mice. Transcriptomics analysis showed that the cytotoxic effects of SGF might be related to the AMPK pathway and ferroptosis. On this basis, SGF was confirmed to induce ferroptosis and dictate ferroptosis sensitivity in a high-fat context mimicked by a two-step conditioned medium (CM) transfer experiment or a Transwell coculture system. The results of Western blotting validated that SGF suppressed the phosphorylation of AMPK, accompanied by alterations in the biomarkers of ferroptosis. These results demonstrate that SGF exerts in vitro and in vivo antiproliferative effects in obesity-associated CRC through inhibiting AMPK activation, thereby driving ferroptosis.
Collapse
Affiliation(s)
- Jianqin Xu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
| | - Zhaowei Cai
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ziyao Pang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiayan Chen
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Keyan Zhu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
| | - Dejun Wang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jue Tu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.X.); (Z.C.); (Z.P.); (J.C.); (K.Z.)
| |
Collapse
|
141
|
Guo Y, Shang S, Liang L, Liu E. ZNF385A was identified as a novel colorectal cancer-related functional gene by analysis of the interaction and immune characteristics of oxidative stress and the inflammatory response. Discov Oncol 2025; 16:290. [PMID: 40064736 PMCID: PMC11893970 DOI: 10.1007/s12672-025-02024-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Recently, oxidative stress and inflammatory responses have been shown to directly impact tumor growth and the tumor microenvironment (TME). However, more research is necessary to fully understand the relationship between oxidative stress and inflammatory responses and colorectal cancer (CRC). METHODS The FindCluster algorithm was used to extract CRC Single-cell RNA sequencing (scRNA-seq) data and identify tumor cell groupings. From the MSigDB database, genes associated with oxidative stress and the inflammatory response were taken. We identified molecular subtypes and built a predictive risk model with the LASSO-Cox method using the ConsensusClusterPlus software suite. We incorporated the prognostic risk model and other clinicopathological parameters into a column-line chart. Finally, we used Quantitative Polymerase Chain Reaction (qPCR) and immunohistochemistry to check the expression of the unreported hub model genes. Cell proliferation was assessed using EDU and colony formation assays. Reactive Oxygen Species (ROS) tests were used to quantitatively determine the ROS content in CRC cells. The ability of CRC cells to invade and migrate was examined using transwell experiments. The regulatory functions of hub model genes were discovered in vivo using a xenograft model tumor assay. RESULTS Oxidative stress and inflammatory response factors in monocytic/macrophages of CRC were significantly upregulated, and their oxidative stress and inflammatory response functions were significantly higher than those of other cell subgroups, as indicated by the enrichment score. These factors showed significant synergistic overexpression and enrichment in this cell population. We constructed a prognostic risk model consisting of seven signatures. The good and stable prognostic evaluation efficacy of the model was confirmed, and risk scores were determined to be independent prognostic factors for CRC. We explored the relationship between the risk score model and malignant progression of tumor cells, tumor immune microenvironment, genomic variation, chemotherapy resistance, and immune response. Further qPCR and immunohistochemistry analysis showed that the expression of ZNF385A was high in CRC tissues. The functional experiment results indicated that interfering with the expression of ZNF385A could suppress the proliferation, ROS, migration and invasion of SW620 cells in vitro and the growth of xenograft tumors in vivo. CONCLUSION In this study, we investigated the critical expression patterns of oxidative stress- and inflammatory response-related genes in CRC, which may contribute to the prognosis and immunotherapy of CRC. Additionally, we discovered ZNF385A to be a novel oncogene in CRC. These findings imply that this model may be applied to assess prognostic risk and identify potential therapeutic targets for CRC patients.
Collapse
Affiliation(s)
- Yaqi Guo
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Shipeng Shang
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Leilei Liang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Enrui Liu
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China.
| |
Collapse
|
142
|
Zushuai M, Yanrong J, Chengdu Z, Xu Z, Qianshan D. Network pharmacological approach combined with weighted gene co-expression network analysis identifies CDKN2A as the keg target of Changweiqing against colorectal cancer. Hereditas 2025; 162:33. [PMID: 40065477 PMCID: PMC11892207 DOI: 10.1186/s41065-025-00405-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Changweiqing (CWQ) is a Chinese herbal formula for the treatment of the gastrointestinal tract diseases, but its role in the treatment of colorectal cancer (CRC) has not been clarified. This study aimed to explore the molecular mechanism of CWQ in CRC treatment through bioinformatics analysis and network pharmacology. METHODS Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform and SwissTargetPrediction database were used to collect the bioactive components of CWQ. The databases including DisgeNET, GeneCards, MalaCards, Online Mendelian Inheritance in Man and Comparative Toxicogenomics were used to obtain CRC-related targets. The Cancer Genome Atlas - colon adenocarcinoma dataset was used to obtain prognosis-related genes in CRC based on weighted gene co-expression network analysis (WGCNA). A protein-protein interaction network was constructed to screen core targets, with STRING database and Cytoscape software. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed using the Database for Annotation, Visualization and Integrated Discovery database. Molecular docking was performed with AutoDock Vina software. Core targets were further analyzed using Gene Expression Profiling Interactive Analysis platform, Human Protein Atlas database, University of ALabama at Birmingham CANcer data analysis Portal (UALCAN) and GeneMANIA database. In vitro experiments were further performed to investigate the effects of quercetin, one of the main components of CWQ, on CRC cells. RESULTS 6356, 1901 and 2980 CRC-related genes were obtained from differential expression analysis, WGCNA and open access databases, respectively. CWQ contained a total of 70 bioactive ingredients, of which 64 ingredients had a total of 836 therapeutic targets. Functional enrichment analysis indicated that CWQ may be involved in regulating pathways in cancer, MAPK signaling pathway and AGE-RAGE signaling pathway, and further analysis identified 14 core targets of CWQ. These core targets were significantly correlated with cell cycle, p53 signaling pathway, FoxO signaling pathway and pathways in cancer. Among these core targets, cyclin-dependent kinase inhibitor 2 A (CDKN2A) expression was closely associated with shorter overall survival and clinical stage of CRC patients. The main bioactive ingredients of CWQ targeting CDKN2A were quercetin, luteolin, kaempferol, isorhamnetin, 7-O-methylisomucronulatol and 7-Methoxy-2-methyl isoflavone. Additionally, quercetin caused G0/G1 phase arrest and inhibited the viability of CRC cells. CONCLUSION The active ingredients of CWQ may play an anti-CRC role through multi-targets and multi-pathways, regulating the cell cycle and cell viability of CRC cells.
Collapse
Affiliation(s)
- Ma Zushuai
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Zhongxiang, Hubei, 431900, China
- Department of Gastroenterology, Zhongxiang People's Hospital, Zhongxiang, Hubei, 431900, China
| | - Ji Yanrong
- Department of Gastroenterology, Zhongxiang People's Hospital, Zhongxiang, Hubei, 431900, China
| | - Zhao Chengdu
- Department of Gastroenterology, Zhongxiang People's Hospital, Zhongxiang, Hubei, 431900, China
| | - Zhu Xu
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Ding Qianshan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Zhongxiang, Hubei, 431900, China.
| |
Collapse
|
143
|
Zhao W, Wu Y, Wang Y, Li T, Liu Q, Hou Z. Exosomal miR-92a-3p modulates M2 macrophage polarization in colorectal cancer: implications for tumor migration and angiogenesis. Med Oncol 2025; 42:96. [PMID: 40059261 DOI: 10.1007/s12032-025-02635-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/13/2025] [Indexed: 03/29/2025]
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignant neoplasms globally. Its development and metastasis are closely associated with the polarization of macrophages within the tumor microenvironment (TME). In particular, the polarization of M2-type macrophages has been demonstrated to be related to the promotion of tumor growth, migration, and angiogenesis. This study aims to investigate the role of miR-92a-3p in colon cancer-derived exosomes in regulating M2-type macrophage polarization by targeting EID2B and to elucidate the impact of this process on tumor migration and angiogenesis. MicroRNAs that were differentially expressed in plasma exosomes from CRC patients were initially identified through a search of the GEO database. The results were then verified by RT-qPCR using miR-92a-3p. The uptake of exosomes was observed via laser confocal microscopy, and the impact of miR-92a-3p on the polarization of exosomes and macrophages was examined through the use of RT-qPCR and WB. A bioinformatics analysis and a dual-luciferase reporter assay were employed to identify the downstream target of miR-92a-3p and to investigate its effect on the MAPK/ERK pathway. miR-92a-3p was upregulated in plasma exosomes of colon cancer patients and exhibited a positive correlation with lymph node metastasis. The results demonstrated that miR-92a-3p was capable of promoting M0 macrophage polarization toward the M2 phenotype, and of enhancing the migratory and invasive capacities of CRC cells, as well as their angiogenic potential in vitro. Bioinformatic analysis and experimental validation demonstrated that miR-92a-3p targeted EID2B and that this target gene was negatively correlated with M2-type macrophage polarization. The results demonstrated that miR-92a-3p promotes macrophage M2 polarization by activating the MAPK/ERK pathway. miR-92a-3p activates the MAPK/ERK pathway and induces macrophage M2 polarization by targeting EID2B, thereby promoting migration and angiogenesis in CRC. These findings offer new potential targets for the treatment of colon cancer.
Collapse
Affiliation(s)
- Wei Zhao
- School of Basic Medical Sciences, Chengde Medical University, Chengde, China
| | - Yudan Wu
- Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yixiao Wang
- School of Basic Medical Sciences, Chengde Medical University, Chengde, China
| | - Tongyi Li
- School of Basic Medical Sciences, Chengde Medical University, Chengde, China
| | - Qiuyan Liu
- Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Zhiping Hou
- School of Basic Medical Sciences, Chengde Medical University, Chengde, China.
| |
Collapse
|
144
|
Li QY, Yan XY, Guan Z, Sun RJ, Lu QY, Li XT, Zhang XY, Sun YS. A method of matching nodes between MRI and pathology with MRI-based 3D node map in rectal cancer. Abdom Radiol (NY) 2025:10.1007/s00261-025-04826-x. [PMID: 40056208 DOI: 10.1007/s00261-025-04826-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/20/2025] [Accepted: 01/26/2025] [Indexed: 03/10/2025]
Abstract
PURPOSE To propose a node-by-node matching method between MRI and pathology with 3D node maps based on preoperative MRI for rectal cancer patients to improve the yet unsatisfactory diagnostic performance of nodal status in rectal cancer. METHODS This methodological study prospectively enrolled consecutive participants with rectal cancer who underwent preoperative MRI and radical surgery from December 2021 to August 2023. All nodes with short-axis diameters of ≥ 3 mm within the mesorectum were regarded as target nodes and were localized in three directions based on the positional relationship on MRI and drawn on a node map with the primary tumor as the main reference, which was used as a template for node-by-node matching with pathological evaluation. Patient and nodal-level analyses were performed to investigate factors affecting the matching accuracy. RESULTS 545 participants were included, of whom 253 received direct surgery and 292 received surgery after neoadjuvant therapy (NAT). In participants who underwent direct surgery, 1782 target nodes were identified on MRI, of which 1302 nodes (73%) achieved matching with pathology, with 1018 benign and 284 metastatic. In participants who underwent surgery after NAT, 1277 target nodes were identified and 918 nodes (72%) achieved matching, of which 689 were benign and 229 were metastatic. Advanced disease and proximity to primary tumor resulted in matching difficulties. CONCLUSION An easy-to-use and reliable method of node-by-node matching between MRI and pathology with 3D node map based on preoperative MRI was constructed for rectal cancer, which provided reliable node-based ground-truth labels for further radiological studies.
Collapse
Affiliation(s)
- Qing-Yang Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/ Beijing), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xin-Yue Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/ Beijing), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhen Guan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/ Beijing), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Rui-Jia Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/ Beijing), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qiao-Yuan Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/ Beijing), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiao-Ting Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/ Beijing), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiao-Yan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/ Beijing), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Ying-Shi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/ Beijing), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
145
|
Edfelt E, Shahrivar M, Holmsten K, Radkiewicz C. Rising incidence trends of synchronous prostate and rectal cancers: a population-based study. Acta Oncol 2025; 64:374-379. [PMID: 40052250 PMCID: PMC11905149 DOI: 10.2340/1651-226x.2025.42592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/24/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND There is a lack of comprehensive reports on time trends in synchronous prostate and rectal cancers. To address this, we conducted the largest cohort study to date to assess these trends in a population-based setting. METHODS We included all adult (ages 18-99) men with incident prostate cancer in the Swedish Cancer Register in 1993-2019. Age-standardized incidence rates (ASIRs) of prostate cancer per 100,000 male population per year were calculated and compared to the ASIR of synchronous (± 6 months from rectal cancer diagnosis) prostate cancer. Age-adjusted synchronous-to-general incidence rate ratios (IRRs) were predicted using Poisson regression. As a sensitivity analysis to assess the effect of incidental findings due to the anatomical proximity, we investigated synchronous prostate and non-sigmoid colon cancers. RESULTS Among 238,252 prostate cancer cases, 594 were synchronous with rectal cancer. The incidence of synchronous prostate cancer increased over the study period, with mean ASIR rising from 418/100,000 (1993-2001) to 788/100,000 (year 2011-2019). The synchronous-to-general IRR increased from 1.92 (95% confidence interval (CI) 1.60-2.31) to 2.61 (95% CI 2.32-2.95) over the same periods. Prostate cancer was also more commonly diagnosed in conjunction with non-sigmoid colon cancer than in the overall male population, but no time trend was observed. INTERPRETATION The incidence of synchronous prostate and rectal cancers has increased over the past 20 years in Sweden, with no signs of plateauing. Future studies are warranted to explore factors contributing to prostate cancer overdiagnosis and to optimize clinical management strategies for this complex patient group.
Collapse
Affiliation(s)
- Elias Edfelt
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Mehrnoosh Shahrivar
- Colorectal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Surgery and Oncology, Capio Sankt Görans Hospital, Stockholm, Sweden.
| | - Karin Holmsten
- Department of Surgery and Oncology, Capio Sankt Görans Hospital, Stockholm, Sweden; Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Radkiewicz
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Surgery and Oncology, Capio Sankt Görans Hospital, Stockholm, Sweden
| |
Collapse
|
146
|
Wu D, Song QY, Dai BS, Li J, Wang XX, Liu JY, Xie TY. Colorectal cancer early screening: Dilemmas and solutions. World J Gastroenterol 2025; 31:98760. [DOI: 10.3748/wjg.v31.i9.98760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 02/18/2025] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignancy worldwide, posing a significant public health concern. Mounting evidence has confirmed that timely early screening facilitates the detection of incipient CRC, thereby enhancing patient prognosis. Obviously, non-participation of asymptomatic individuals in screening programs hampers early diagnosis and may adversely affect long-term outcomes for CRC patients. In this letter, we provide a comprehensive overview of the current status of early screening practices, while also thoroughly examine the dilemmas and potential solutions associated with early screening for CRC. In response to these issues, we proffer a set of recommendations directed at governmental authorities and the general public, which focus on augmenting financial investment, establishing standardized screening protocols, advancing technological capabilities, and bolstering public awareness campaigns. The importance of collaborative efforts from various stakeholders cannot be overstated in the quest to enhance early detection rates and alleviate the societal burden of CRC.
Collapse
Affiliation(s)
- Di Wu
- Department of General Surgery, First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Qi-Ying Song
- Department of General Surgery, First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Bai-Shu Dai
- Department of General Surgery, First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Jie Li
- Department of General Surgery, First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Xin-Xin Wang
- Department of General Surgery, First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Jia-Yu Liu
- Department of Neurosurgery, First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Tian-Yu Xie
- Department of General Surgery, First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
147
|
Wang J, Zhu L, Li Y, Ding M, Wang X, Xiong B, Chen H, Chang L, Chen W, Han B, Lu J, Shi Q. Multi-omics analysis reveals Jianpi formula-derived bioactive peptide-YG-22 potentially inhibited colorectal cancer via regulating epigenetic reprogram and signal pathway regulation. Front Genet 2025; 16:1560172. [PMID: 40110036 PMCID: PMC11919836 DOI: 10.3389/fgene.2025.1560172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Colorectal cancer (CRC) is a prevalent malignancy worldwide, often treated with chemotherapy despite its limitations, including adverse effects and resistance. The traditional Chinese medicine (TCM) Jianpi formula has been demonstrated to improve efficacy of chemotherapy, however the underlying mechanisms still need to be explored. In this study, we aim to screen bioactive peptides derived from the blood of CRC patients through peptidomics and explore the molecular mechanisms of the candidate peptides in the inhibition of CRC using multi-omics analysis. Methods In this study, we recruited 10 patients with CRC who had received either adjuvant chemotherapy or adjuvant chemotherapy combined with the traditional Chinese medicine Jianpi formula after surgery. We collected plasma samples at 2 cycles of adjuvant therapy and performed peptidomic analysis on these samples. The differentially bioactive peptides were screened using a model of HCT116 cells in vitro. To investigate the molecular mechanism underlying YG-22's inhibition of the colorectal cancer cell line HCT116, we performed a multi-omics analysis, including transcriptome, metabolome, chromatin accessibility, H3K4Me3 histone methylation, and NF-κB binding site analyses. Results Differential peptides were identified in plasma samples from patients treated with adjuvant chemotherapy combined with the Jianpi formula. Among these peptides, YG-22 exhibited the strongest cytotoxic effect on HCT116 cells, reducing cell viability in a dose- and time-dependent manner. Transcriptome analysis highlighted that YG-22 treatment in CRC modulates key pathways associated with lysosome-mediated degradation and apoptosis. Metabolomic profiling further indicated disruptions in tumor-supportive metabolic pathways. Chromatin accessibility and histone modification analyses suggested that YG-22 induces epigenetic reprogramming. Additionally, treatment with YG-22 resulted in significant changes in NF-κB binding and pathway activation. Conclusions This study demonstrates that combining chemotherapy with TCM Jianpi formula enriches the molecular landscape and generates bioactive peptides with strong antitumor activity. Furthermore, this study also lays the foundation for further development of peptide-based therapies and highlights the value of combining traditional and modern therapeutic strategies for CRC management.
Collapse
Affiliation(s)
- Jun Wang
- Department of Oncology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of General Surgery, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lijuan Zhu
- Department of Anorectal, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mingming Ding
- Department of Oncology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiyu Wang
- Department of Oncology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bo Xiong
- Department of Clinical Pharmacy, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongyu Chen
- Department of Oncology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lisheng Chang
- Department of Oncology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenli Chen
- Department of Oncology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bo Han
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Lu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Shi
- Department of Oncology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
148
|
Zhu J, Xu B, Wu Z, Yu Z, Ji S, Lian J, Lu H. Integrative analysis of semaphorins family genes in colorectal cancer: implications for prognosis and immunotherapy. Front Immunol 2025; 16:1536545. [PMID: 40103807 PMCID: PMC11913869 DOI: 10.3389/fimmu.2025.1536545] [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: 11/29/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Background Semaphorins (SEMAs), originally identified as axon guidance factors, have been found to play crucial roles in tumor growth, invasiveness, neoangiogenesis, and the modulation of immune responses. However, the prognostic value of SEMA-related genes in colorectal cancer (CRC) remains unclear. Methods We applied a novel machine learning framework that incorporated 10 machine learning algorithms and their 101 combinations to construct a SEMAs-related score (SRS). Multi-omics analysis was performed, including single-cell RNA sequencing (scRNA-seq), and spatial transcriptome (ST) to gain a more comprehensive understanding of the SRS. A series of cell experiments were conducted to prove the impact of key genes on CRC biological behavior. Result A consensus SRS was finally constructed based on a 101-combination machine learning computational framework, demonstrating outstanding performance in predicting overall survival. Moreover, distinct biological functions, mutation burden, immune cell infiltration, and immunotherapy response were observed between the high- and low-SRS groups. scRNA-seq and ST demonstrated unique cellular heterogeneity in CRC. We observed that SRS-high and SRS-low malignant epithelial cells exhibit different biological characteristics. High SRS malignant epithelial cells interact with myeloid and endothelial cells via SPP1 and COL4A2-ITGAV-ITGB8 pathways, respectively. Low SRS cells engage with myeloid and endothelial cells through MIF and JAG1-NOTCH4 pathways. Additionally, knocking down SEMA4C significantly inhibits the proliferation and invasion of CRC cells, while promoting apoptosis in vitro. Conclusion SRS could serve as an effective tool to predict survival and identify potential patients benefiting from immunotherapy in CRC. It also reveals tumor heterogeneity and provides valuable biological insights in CRC.
Collapse
Affiliation(s)
- Jiahao Zhu
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Benjie Xu
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Zhixing Wu
- Department of Computer Science, University of Liverpool, Liverpool, United Kingdom
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, China
| | - Zhiwei Yu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Shengjun Ji
- Department of Radiotherapy and Oncology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jie Lian
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Haibo Lu
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
149
|
Yao L, Gu C, Ge R, Zhang X, Meng X, Wang L, Peng D, Li G. Acetylated Dendrobium huoshanense polysaccharide: a novel inducer of apoptosis in colon cancer cells via Fas-FasL pathway activation and metabolic reprogramming. Front Oncol 2025; 15:1529868. [PMID: 40104499 PMCID: PMC11913854 DOI: 10.3389/fonc.2025.1529868] [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: 11/18/2024] [Accepted: 02/12/2025] [Indexed: 03/20/2025] Open
Abstract
Introduction Not all polysaccharides function as antitumor drugs, nor do they universally possess the same advantages regarding safety and biocompatibility. Those polysaccharides that are effective antitumor agents typically demonstrate superior safety profiles and biocompatibility compared to synthetic anticancer drugs, which can exhibit high toxicity and harmful side effects. Dendrobium huoshanense polysaccharide (DHP) has been recognized for its potential bioactive properties, particularly in anti-tumor treatment. This study investigates the effects of DHP on the proliferation and apoptosis of HCT116 colon cancer cells. Methods DHP was extracted according to previously published experimental methods. The inhibitory effects of DHP were evaluated using IEC6, Caco-2, and HCT116 cell lines, with changes in cell morphology observed via transmission electron microscopy. After establishing the conditions for DHP administration, flow cytometry was employed to assess its effects on apoptosis, reactive oxygen species (ROS), and mitochondrial membrane potential of HCT116 cells. Additionally, immunoprecipitation, quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, and biomarker detection were utilized to investigate the mechanisms underlying DHP's inhibition of HCT116 cells and its impact on metabolic reprogramming. Results In the present study, we observed that DHP treatment at 600 μg/ml for 24 h reduced HCT116 cell viability to 54.87%. In contrast, the inhibitory effect of DHP on the viability of IEC6 and Caco-2 cells was relatively mild. The specific mechanism involves DHP activating the mitochondrial apoptotic pathway leading to the downregulation of key metabolic intermediates and enzymes such as uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) and ST6Gal-I. By inhibiting ST6Gal-I activity, DHP activates the Fas/FasL signaling pathway. Additionally, DHP-induced ROS production effectively triggers apoptosis in HCT116 cells. Conclusion Our study demonstrates that DHP effectively inhibits the proliferation and induces apoptosis in HCT116 colon cancer cells through the activation of the Fas-FasL signaling pathway and metabolic reprogramming. The selective inhibitory effect of DHP on HCT116 cells, the activation of both death receptor and mitochondrial apoptotic pathways, and the modulation of metabolic reprogramming provide novel insights into the potential therapeutic strategies for colon cancer.
Collapse
Affiliation(s)
- Liang Yao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
| | - Chen Gu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
| | - Ruipeng Ge
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
| | - Xiaoqian Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
| | - Xinqian Meng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
| | - Lei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory for Research and Development of Research and Development of Chinese Medicine, Hefei, China
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory for Research and Development of Research and Development of Chinese Medicine, Hefei, China
| | - Guozhuan Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
| |
Collapse
|
150
|
Daca-Alvarez M, Perea J, Corchete L, Spinelli A, Foppa C, de Miranda NFCC, Nielsen M, Palles C, Curley HM, Marti-Gallostra M, Verdaguer M, Vivas A, Lorenzo S, Latchford A, Faiz O, Monahan K, Pawa N, Szczepkowski M, Ziółkowski B, Tarnowski W, Uryszek M, Makkai-Popa ST, Azagra JS, Llach J, Moreria L, Pellise M, Holowatyj AN, González-Sarmiento R, Balaguer F. Regional patterns of early-onset colorectal cancer from the GEOCODE (Global Early-Onset COlorectal Cancer DatabasE)-European consortium: retrospective cohort study. BJS Open 2025; 9:zraf024. [PMID: 40103402 PMCID: PMC11920508 DOI: 10.1093/bjsopen/zraf024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND The incidence of early-onset colorectal cancer is increasing, but in Europe this growth shows a heterogeneous pattern in different countries and regions. METHODS Patients from six countries who participated in the Global Early-Onset COlorectal Cancer DatabasE (GEOCODE)-Europe group were included. The inclusion criteria were patients with colorectal adenocarcinoma diagnosed between 18 and 49 years of age, between January 2010 and December 2017, with at least 3 years of follow-up. Patients with inherited colorectal cancer syndromes were excluded. RESULTS A total of 851 patients were included with almost equal sex distribution, most were diagnosed at age 39 years or older and 42% of patients were overweight or obese. Diagnoses were predominantly at later stages (62.5% stage III-IV) and tumours were predominantly located in the distal colon (76.9% left colon and rectum). Comparative analysis between countries demonstrated that the UK had a younger age at diagnosis and the Italian cohort had a higher prevalence of being overweight or obese. Patients from Luxembourg had more advanced stage diagnoses and those from The Netherlands had more polyps. Patients from the UK had a greater family history of colorectal cancer. Comparison of Mediterranean versus non-Mediterranean countries showed significant differences in the age at diagnosis and body mass index. The prevalence of early-onset colorectal cancer over the age of 40 years in Mediterranean versus non-Mediterranean countries was 71.4% versus 62.1% (P = 0.002), and early-onset colorectal cancer was diagnosed at a more advanced stage in Mediterranean countries versus non-Mediterranean countries (65.3% versus 54.7%; P = 0.033). Family history of colorectal cancer in a first-degree relative was more common in non-Mediterranean versus Mediterranean countries (19.1% versus 11.4%; P < 0.001). CONCLUSION This study highlights significant geographical disparities in the clinical, pathological and familial features of early-onset colorectal cancer across European countries.
Collapse
Affiliation(s)
- Maria Daca-Alvarez
- Department of Gastroenterology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - José Perea
- Molecular Medicine Unit-Department of Medicine, Biomedical Research Institute of Salamanca (IBSAL), Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-SACYL-CSIC, Salamanca, Spain
- Surgical Department, Vithas Arturo Soria Hospital, Fundación Vithas, Grupo Hospitales Vithas, Madrid, Spain
| | - Luis Corchete
- Molecular Medicine Unit-Department of Medicine, Biomedical Research Institute of Salamanca (IBSAL), Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-SACYL-CSIC, Salamanca, Spain
| | - Antonino Spinelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Colon and Rectal Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Caterina Foppa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Colon and Rectal Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Noel F C C de Miranda
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Maartje Nielsen
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Claire Palles
- Department of Cancer and Genomic Sciences, Birmingham City University, Birmingham, West Midlands, UK
| | - Helen M Curley
- Department of Cancer and Genomic Sciences, Birmingham City University, Birmingham, West Midlands, UK
| | - Marc Marti-Gallostra
- Department of Surgery, Hospital Universitari Vall d'Hebron, Barcelona, Catalunya, Spain
| | - Mireia Verdaguer
- Department of Surgery, Hospital Universitari Vall d'Hebron, Barcelona, Catalunya, Spain
| | - Alfredo Vivas
- Department of Surgery, Hospital Universitario 12 de Octubre, Madrid, Comunidad de Madrid, Spain
| | - Sofia Lorenzo
- Department of Surgery, Hospital Universitario 12 de Octubre, Madrid, Comunidad de Madrid, Spain
| | - Andrew Latchford
- Department of Gastroenterology, London Northwest Healthcare NHS Trust, Harrow, London, UK
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Omar Faiz
- Department of Gastroenterology, London Northwest Healthcare NHS Trust, Harrow, London, UK
| | - Kevin Monahan
- Department of Surgery and Cancer, Imperial College, London, UK
- Department of Surgery, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Nikhil Pawa
- Department of Surgery, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Marek Szczepkowski
- Clinical Department of Colorectal, General and Oncological Surgery, Centre of Postgraduate Medical Education in Warsaw, Bielanski Hospital in Warsaw, Warsaw, Poland
| | - Bartosz Ziółkowski
- Clinical Department of Colorectal, General and Oncological Surgery, Centre of Postgraduate Medical Education in Warsaw, Bielanski Hospital in Warsaw, Warsaw, Poland
| | - Wieslaw Tarnowski
- Department of Surgery, Samodzielny Publiczny Szpital Kliniczny im prof Witolda Orrowskiego, Warszawa, Poland
| | - Mariusz Uryszek
- Department of Surgery, Samodzielny Publiczny Szpital Kliniczny im prof Witolda Orrowskiego, Warszawa, Poland
| | | | - Juan S Azagra
- Department of Surgery, Centre Hospitalier de Luxembourg Eich, Luxembourg, Luxembourg
| | - Joan Llach
- Department of Gastroenterology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Leticia Moreria
- Department of Gastroenterology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
- Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain
| | - Maria Pellise
- Department of Gastroenterology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
- Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain
| | - Andreana N Holowatyj
- Department of Medicine, Vanderbilt University Medical Centre, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt-Ingram Cancer Centre, Nashville, Tennessee, USA
| | - Rogelio González-Sarmiento
- Molecular Medicine Unit-Department of Medicine, Biomedical Research Institute of Salamanca (IBSAL), Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-SACYL-CSIC, Salamanca, Spain
| | - Francesc Balaguer
- Department of Gastroenterology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
- Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain
| |
Collapse
|