1
|
Wang M, Zhang Z, Yang Y, Peng X, Yin H. A targeted MAVS fusion protein for controlled innate immune activation and antitumor therapy. Oncoimmunology 2025; 14:2478850. [PMID: 40085508 PMCID: PMC11913393 DOI: 10.1080/2162402x.2025.2478850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Targeted therapies leveraging the innate immune system are emerging as promising cancer treatments. The mitochondrial antiviral signaling protein (MAVS) plays a crucial role in initiating innate immune responses, but its clinical use is limited by the risk of uncontrolled activation and systemic toxicity. To address this, we developed a novel therapeutic agent, the truncated interferon activation switch (TRIAS), combining MAVS truncates with a tumor antigen-targeting single-chain variable fragment (scFv). This design ensures antigen-dependent, controlled activation. Lentiviral delivery of TRIAS induced significant antitumor responses, including complete tumor regression in some cases. Flow cytometry (FCM) analysis further confirmed that tumor cells were the predominant population expressing the transgene. TRIAS-expressing tumor cells exhibited enhanced antitumor activity, likely due to increased cytokine release and upregulated major histocompatibility complex (MHC) expression, enabling tumor cells to function as antigen-presenting cells. This activated other immune cells, driving adaptive immune responses. Additionally, TRIAS promoted a proinflammatory shift in the tumor microenvironment (TME). In conclusion, TRIAS was validated as an innovative immunotherapeutic agent with MAVS-like immune-activating properties and tightly controlled mechanisms, offering a safer and more effective approach for clinical cancer immunotherapy.
Collapse
Affiliation(s)
- Muhan Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhijie Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - YouYou Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaoyi Peng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Hongping Yin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
2
|
Dong Y, Meng F, Wang J, Wei J, Zhang K, Qin S, Li M, Wang F, Wang B, Liu T, Zhong W, Cao H. Desulfovibrio vulgaris flagellin exacerbates colorectal cancer through activating LRRC19/TRAF6/TAK1 pathway. Gut Microbes 2025; 17:2446376. [PMID: 39718561 DOI: 10.1080/19490976.2024.2446376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/22/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024] Open
Abstract
The initiation and progression of colorectal cancer (CRC) are intimately associated with genetic, environmental and biological factors. Desulfovibrio vulgaris (DSV), a sulfate-reducing bacterium, has been found excessive growth in CRC patients, suggesting a potential role in carcinogenesis. However, the precise mechanisms underlying this association remain incompletely understood. We have found Desulfovibrio was abundant in high-fat diet-induced Apcmin/+ mice, and DSV, a member of Desulfovibrio, triggered colonocyte proliferation of germ-free mice. Furthermore, the level of DSV progressively rose from healthy individuals to CRC patients. Flagella are important accessory structures of bacteria, which can help them colonize and enhance their invasive ability. We found that D. vulgaris flagellin (DVF) drove the proliferation, migration, and invasion of CRC cells and fostered the growth of CRC xenografts. DVF enriched the epithelial-mesenchymal transition (EMT)-associated genes and characterized the facilitation of DVF on EMT. Mechanistically, DVF induced EMT through a functional transmembrane receptor called leucine-rich repeat containing 19 (LRRC19). DVF interacted with LRRC19 to modulate the ubiquitination of tumor necrosis factor receptor-associated factor (TRAF)6, rather than TRAF2. This interaction drove the ubiquitination of pivotal molecule TAK1, further enhancing its autophosphorylation and ultimately contributing to EMT. Collectively, DVF interacts with LRRC19 to activate the TRAF6/TAK1 signaling pathway, thereby promoting the EMT of CRC. These data shed new light on the role of gut microbiota in CRC and establish a potential clinical therapeutic target.
Collapse
Affiliation(s)
- Yue Dong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Fanyi Meng
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingyi Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingge Wei
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Kexin Zhang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Siqi Qin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Mengfan Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Fucheng Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| |
Collapse
|
3
|
Katona BW, Shukla A, Hu W, Nyul T, Dudzik C, Arvanitis A, Clay D, Dungan M, Weber M, Tu V, Hao F, Gan S, Chau L, Buchner AM, Falk GW, Jaffe DL, Ginsberg G, Palmer SN, Zhan X, Patterson AD, Bittinger K, Ni J. Microbiota and metabolite-based prediction tool for colonic polyposis with and without a known genetic driver. Gut Microbes 2025; 17:2474141. [PMID: 40069167 PMCID: PMC11913376 DOI: 10.1080/19490976.2025.2474141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/22/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Despite extensive investigations into the microbiome and metabolome changes associated with colon polyps and colorectal cancer (CRC), the microbiome and metabolome profiles of individuals with colonic polyposis, including those with (Gene-pos) and without (Gene-neg) a known genetic driver, remain comparatively unexplored. Using colon biopsies, polyps, and stool from patients with Gene-pos adenomatous polyposis (N = 9), Gene-neg adenomatous polyposis (N = 18), and serrated polyposis syndrome (SPS, N = 11), we demonstrated through 16S rRNA sequencing that the mucosa-associated microbiota in individuals with colonic polyposis is representative of the microbiota associated with small polyps, and that both Gene-pos and SPS cohorts exhibit differential microbiota populations relative to Gene-neg polyposis cohorts. Furthermore, we used these differential microbiota taxa to perform linear discriminant analysis to differentiate Gene-neg subjects from Gene-pos and from SPS subjects with an accuracy of 89% and 93% respectively. Stool metabolites were quantified via 1H NMR, revealing an increase in alanine in SPS subjects relative to non-polyposis subjects, and Partial Least Squares Discriminant Analysis (PLS-DA) analysis indicated that the proportion of leucine to tyrosine in fecal samples may be predictive of SPS. Use of these microbial and metabolomic signatures may allow for better diagnostric and risk-stratification tools for colonic polyposis patients and their families as well as promote development of microbiome-targeted approaches for polyp prevention.
Collapse
Affiliation(s)
- Bryson W Katona
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ashutosh Shukla
- Division of Digestive & Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weiming Hu
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Thomas Nyul
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Christina Dudzik
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Alex Arvanitis
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel Clay
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michaela Dungan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marina Weber
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Vincent Tu
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fuhua Hao
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA, USA
| | - Shuheng Gan
- Peter O'Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lillian Chau
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anna M Buchner
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gary W Falk
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David L Jaffe
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gregory Ginsberg
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Suzette N Palmer
- Peter O'Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaowei Zhan
- Peter O'Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Josephine Ni
- Division of Digestive & Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
4
|
Lyu M, Zhang T, Bao Z, Li P, Chen M, Quan H, Wang C, Xia L, Li Y, Tang B. In situ forming AIEgen-alginate hydrogel for remodeling tumor microenvironment to boost FLASH immunoradiotherapy. Biomaterials 2025; 320:123281. [PMID: 40138965 DOI: 10.1016/j.biomaterials.2025.123281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
FLASH radiotherapy, which involves the delivery of an ultra-high radiation dose rate exceeding 40 Gy/s, has emerged as a promising tumor ablation strategy. While this approach generally spares normal tissues, the incomplete killing of tumors may sometimes lead to recurrence due to the immunosuppressive tumor microenvironment (TME). Herein, an aggregation-induced-emission luminogen (AIEgen)-alginate hydrogel was used to sensitize colon cancer via photodynamic therapy (PDT). Flower-like calcium carbonate nanoparticles, doped with an AIEgen termed CQu, were designed and applied as a cocktail with sodium alginate. When exposed to the acidic TME, Ca2+ is released from this structure, resulting in sodium alginate termed FA forming a hydrogel in situ within the TME. This hydrogel also captures high concentrations of CQu in the local TME. Under laser irradiation, the CQu can generate sustained reactive oxygen species (ROS) production, thereby facilitating Ca2+ influx and causing mitochondrial damage. Through a single injection of established FA hydrogel, followed by PDT and FLASH radiotherapy, immunogenic tumor cell death was induced which promoted antitumor immunity, thereby protecting against tumor recurrence while realizing abscopal effect. The results highlight the potential to improve the sensitivity of tumor cells to FLASH radiotherapy through sustained ROS production and Ca2+ overload, thereby yielding optimal immunotherapy outcomes.
Collapse
Affiliation(s)
- Meng Lyu
- Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China
| | - Tianfu Zhang
- School of Biomedical Engineering, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhirong Bao
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Pei Li
- Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China
| | - Mingzhu Chen
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei, 430072, China
| | - Hong Quan
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei, 430072, China
| | - Cunchuan Wang
- Clinical Medicine Research Institute, Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China.
| | - Ligang Xia
- Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China.
| | - Yang Li
- Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China.
| | - Benzhong Tang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, (CUHK-Shenzhen), Guangdong, 518172, China
| |
Collapse
|
5
|
Saadh MJ, Allela OQB, Kareem RA, Baldaniya L, Ballal S, Vashishth R, Parmar M, Sameer HN, Hamad AK, Athab ZH, Adil M. Prognostic gene expression profile of colorectal cancer. Gene 2025; 955:149433. [PMID: 40122415 DOI: 10.1016/j.gene.2025.149433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer is a major global health burden, with significant heterogeneity in clinical outcomes among patients. Identifying robust prognostic gene expression signatures can help stratify patients, guide treatment decisions, and improve clinical management. This review provides an overview of current prognostic gene expression profiles in colorectal cancer research. We have synthesized evidence from numerous published studies investigating the association between tumor gene expression patterns and patient survival outcomes. The reviewed literature reveals several promising gene signatures that have demonstrated the ability to predict disease-free survival and overall survival in CRC patients, independent of standard clinicopathological risk factors. These genes are crucial in fundamental biological processes, including cell cycle control, epithelial-mesenchymal transition, and immune regulation. The implementation of prognostic gene expression tests in clinical practice holds great potential for enabling more personalized management strategies for colorectal cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Manisha Parmar
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India.
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq.
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq.
| | | |
Collapse
|
6
|
Akkus E, Karaoğlan BB, Kayaalp M, Turmuş U, Akyol C, Utkan G. Stage-specific characterization of "early-onset colorectal cancer": Localized and synchronous metastatic disease. Int J Cancer 2025; 156:2340-2351. [PMID: 39887374 PMCID: PMC12008821 DOI: 10.1002/ijc.35336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025]
Abstract
Early-onset colorectal cancer (EOCRC) is an alarming entity worldwide. Yet, stage-specific characteristics and prognosis in localized and synchronous metastatic EOCRC are not well-defined. Two cohorts of CRC patients (localized and synchronous metastatic) were evaluated, defining EOCRC as the diagnosis <50 years old. Five hundred sixty-eight patients were included (n = 432 localized, 14.4% [n = 62] EOCRC and n = 136 synchronous metastatic, 20.6% [n = 28] EOCRC). 93.5% of localized and 96.5% of synchronous metastatic EOCRC patients were symptomatic at diagnosis. Among localized patients, female gender (58.1% vs. 40%, p = .008), perineural invasion (41.9% vs. 24.9%, p = .005), folinic acid, 5-fluorouracil, and oxaliplatin chemotherapy (45.2% vs. 25.2%, p = .003), and perioperative chemotherapy cycles (9.21 [± 3.10] vs. 7.98 [± 2.92], p = .006) were higher in EOCRC compared with ≥50-year. Median recurrence-free survival (RFS) and overall survival were not reached in either group (p = .234 and p = .831). Only RAS mutant status was associated with RFS (Hazard ratio: 7.09 [95% confidence interval (CI): 1.87-26.76], p < .001) in EOCRC. Among synchronous metastatic patients, urgent surgery (32.1% vs. 11.1%, p = .014) and local treatments (39.3% vs. 20.4%, p = .037) were more frequent in EOCRC. Median progression-free survival and overall survival in the EOCRC and ≥50 years were 8.07 months (95% CI: 5.03-12.97) vs. 10.03 months (95% CI, 8.40-13.10) (p = .450) and 18.57 months (95% CI, 13.33-43.03) vs. 19.83 months (95% CI, 16.07-27.30) (p = .833), respectively. Synchronous metastatic EOCRC more frequently underwent urgent surgery (32.1% vs. 8%, p = .008) and had RAS mutation (43.5% vs. 16.7%, p = .032) than localized EOCRC. This study suggests that localized and synchronous metastatic EOCRC patients may have different characteristics than average onset, without survival differences. Implementation of stage-specific characteristics into daily practice is necessary for decision-making processes in these young patients.
Collapse
Affiliation(s)
- Erman Akkus
- Department of Medical OncologyAnkara University Faculty of MedicineAnkaraTürkiye
- Ankara University Cancer Research InstituteAnkaraTürkiye
| | - Beliz Bahar Karaoğlan
- Department of Medical OncologyAnkara University Faculty of MedicineAnkaraTürkiye
- Ankara University Cancer Research InstituteAnkaraTürkiye
| | - Mehmet Kayaalp
- Department of Medical OncologyAnkara University Faculty of MedicineAnkaraTürkiye
- Ankara University Cancer Research InstituteAnkaraTürkiye
| | - Utkucan Turmuş
- Department of Internal MedicineAnkara University Faculty of MedicineAnkaraTürkiye
| | - Cihangir Akyol
- Department of SurgeryAnkara University Faculty of MedicineAnkaraTürkiye
| | - Güngör Utkan
- Department of Medical OncologyAnkara University Faculty of MedicineAnkaraTürkiye
- Ankara University Cancer Research InstituteAnkaraTürkiye
| |
Collapse
|
7
|
Sergeev D, Heisser T, Hoffmeister M, Brenner H. Potential for enhancing efficacy of screening colonoscopy by lowering starting ages and extending screening intervals: A modelling study for Germany. Int J Cancer 2025; 156:2303-2310. [PMID: 39751766 PMCID: PMC12008824 DOI: 10.1002/ijc.35322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
Studies aimed to evaluate the expected impact of alternative screening strategies are essential for optimizing colorectal cancer (CRC) screening offers, but such studies are lacking in Germany, where two screening colonoscopies (CS) 10 years apart are offered for men from age 50 and women from age 55. Our aim was to explore whether and to what extent the efficacy of utilizing two CS could be enhanced by alternative starting ages and screening intervals. We modeled the expected numbers of CRC cases, CRC deaths, years of potential life lost (YPLL), and disability-adjusted life years (DALYs) due to CRC in hypothetical cohorts of 100,000 men and women aged 45-85 using COSIMO, a validated Markov-based multi-state simulation model. Modeled strategies included combinations of starting ages (45/50/55/60) and CS (10/15/20 years). For men, CRC deaths could be slightly reduced by extending the interval to 15 years, with a second CS at 65. YPLL and DALYs would be reduced by decreasing starting age to 45 when combined with a 15-year screening interval. For women, use of two CS at ages 50 and 65 would reduce all CRC burden parameters compared to the current earliest-use offer at 55 and 65 years. Our results suggest that lowering the starting age of screening colonoscopy to 45 for men and 50 for women, combined with extending the CS screening interval to 15 years would have the potential to enable significant reductions in years of potential life lost, and disability-adjusted life years compared to current screening offers in Germany.
Collapse
Affiliation(s)
- Dmitry Sergeev
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Medical Faculty Heidelberg, Heidelberg UniversityHeidelbergGermany
| | - Thomas Heisser
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
8
|
Gao X, Zhang G, Wang F, Ruan W, Sun S, Zhang Q, Liu X. Emerging roles of EGFL family members in neoplastic diseases: Molecular mechanisms and targeted therapies. Biochem Pharmacol 2025; 236:116847. [PMID: 40044051 DOI: 10.1016/j.bcp.2025.116847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/09/2025]
Abstract
Epidermal growth factor-like proteins (EGFLs) contain more than a single EGF/EGF-like domain within their protein structure. To date, ten EGFL family members (EGFL1-10) have been characterized across diverse tissues and developmental stages under different conditions. In this review, we conclude that EGFLs are instrumental in regulating biological activities and pathological processes. Under physiological conditions, EGFLs participate in angiogenesis, neurogenesis, osteogenesis, and other processes. Under pathological conditions, EGFLs are linked with different diseases, particularly cancers. Furthermore, we highlight recent advancements in the study of EGFLs in biological conditions and cancers. In addition, the regulatory role and key underlying mechanism of EGFLs in mediating tumorigenesis are discussed. This paper also examines potential antagonists that target EGFL family members in cancer therapeutics. In summary, this comprehensive review elucidates the critical role of EGFLs in neoplastic diseases and highlights their potential as therapeutic targets.
Collapse
Affiliation(s)
- Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Guopeng Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Feitong Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Wenhui Ruan
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China
| | - Shishuo Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Xiangye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China; Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China.
| |
Collapse
|
9
|
Zhong XM, Liu XR. Effect of laxative use and laxative type on colorectal cancer risk: A pooling up analysis and evidence synthesis. Oncol Lett 2025; 29:284. [PMID: 40247990 PMCID: PMC12004035 DOI: 10.3892/ol.2025.15030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide and there is a controversy regarding the influence of laxative use on the incidence of CRC. Therefore, the present study aimed to investigate the effects of laxative use and different subtypes of laxatives on the incidence of CRC. To this aim, a comprehensive search of three databases (PubMed, Embase and the Cochrane Library) was conducted on April 12, 2022, using key words that included 'laxative' and 'CRC', which initially retrieved 305 records. Ultimately, 12 studies involving 415,313 patients met all eligibility criteria and were included in a meta-analysis. Subsequently, patients were categorized into the laxative use and non-laxative use groups. Stata 16.0 software was used for all data analyses. The results indicated that laxative use was not significantly associated with CRC risk [odds ratio (OR), 0.95; 95% confidence interval (CI), 0.75-1.20; P=0.65; I2=94.63%]. In the subgroup analyses, the effects of different laxative types were further examined. Notably, all types of laxatives except for fiber laxatives showed no significant influence on CRC risk (P>0.1). By contrast, fiber laxatives were associated with a reduced risk of CRC (OR, 0.74; 95% CI, 0.59-0.93; P=0.01; I2=32.15%), suggesting a potential protective effect of this medication. In conclusion, the findings of the present study suggest that the use of laxatives does not increase the risk of CRC. Moreover, the use of fiber laxatives may have a protective effect by reducing CRC incidence.
Collapse
Affiliation(s)
- Xue-Mei Zhong
- Department of Endocrinology, School of Clinical Medicine, Chongqing Medical and Pharmaceutical College, Chongqing 401331, P.R. China
| | - Xu-Rui Liu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
10
|
Abdel-Wahab EA, Al-Qaim ZH, Faris Al-Karkhi AT, Fayed AM, Eldmrdash AM, Hussein MA, Abdel-Aziz A, Metwaly AM, Abdelzaher H, Abdelzaher M, ALsherif DA. Phloretin-nanospanlastics for targeting the Akt/PI3K signaling pathways in dimethylhydrazine-induced colon cancer in mice. Int J Pharm X 2025; 9:100311. [PMID: 39811247 PMCID: PMC11732206 DOI: 10.1016/j.ijpx.2024.100311] [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: 05/13/2024] [Revised: 11/07/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
Objectives Colorectal cancer is the third most common cancer worldwide, accounting for approximately 10 % of all cancer cases. It is also the second leading cause of cancer-related deaths globally. Phloretin is a natural compound found in apples and other fruits. It has been studied for its potential health benefits, including antioxidant and anti-inflammatory properties. However, more research is needed to fully understand its impact on cancer prevention or treatment. This article aimed to prepare phloretin-nanospanlastics (Ph-NSLs) to evaluate their effects on dimethylhydrazine (DMH)-induced colon cancer in mice. Methods Morphology, Particle size, zeta potential, UV-vis, entrapment efficiency, polydispersity index, FT-IR spectra, and drug release of phloretin and Ph-NSLs at pH 6.8.were described. Ph-NSLs were also tested for their anti-cancer properties in DMH-induced colon cancer in mice. A 36 mice were divided into 6 groups; Normal control, DMH (20 mg/k.g.b.w.), DMH + Ph-NSLs (25 mg/k.g.b.w.), DMH + Ph-NSLs (50 mg/k.g.b.w.), DMH + 5-FU(20 mg/k.g.b.w.), DMH + Ph-NSLs (50 mg), 5-FU (20 mg). Ph-NSLs were tested for their anticancer properties in DMH-treated mice by evaluating the IC50, viability and inhibitory values of Ph-NSLs against Caco-2. Also, the effect of Ph-NSLs administration on number of surviving mice, number of tumors/mice, average of tumor size, Hb, RBCs, WBCs, C19-9, MDA, GSH, SOD, IL-2, TNF-α, TGF-β1, CEA, and P53 levels in mice treated DMH were estimated. Results The synthesized Ph-NSLs were uniform, spherically shaped, and well dispersed, with a size, entrapment efficiency, and polydispersity index of approximately 114.06 ± 8.35 nm, 78.60 %, and 0.05, respectively. The zeta potential value of Ph-NSLs was measured at -21.5 ± 1.47 mV. Zeta potential reflects the surface charge of nanoparticles and affects their stability and interactions. UV spectra of phloretin and Ph-NSLs showed strong absorption peaks at 225 and 285 nm. These peaks correspond to specific wavelengths where the compound absorbs light. The percentage of Ph- NSLs release was found to be 56.87 ± 2.45 %. IC50 of Ph-NSLs was recorded 15.76 ± 0.42 μg/ml and the viability and inhibitory values of Ph-NSLs against Caco-2 cell lines was resorded 2.39, and 97.61 %, respectively at 100 μg/ml as well as 10.3, and 89.7 %, respectively at 50 μg/ml.Moreover, The combination of 5-FU and Ph-NSLs resulted in a moderate increase in survival and significantly reduces tumor size and number, showing enhanced anticancer efficacy compared to individual treatments as well as attenuated levels of hemoglobin (Hb), red blood cells (RBCs), and white blood cells (WBCs). Reduced plasma cancer antigen 19-9 (CA19-9) levels as well as improved of colon malondialdehyde (MDA), reduced glutathione (GSH), superoxide dismutase (SOD), interleukine-2 (IL-2), tumor necrosis factor-alpha (TNF-α), tumor growth factor-beta1 (TGF-β1), carcinoembryonic antigen (CEA), and tumor protein (P53) levels. Also, Ph-NSLs and 5FU, either alone or together, decreased the expression of the Akt and PI3K genes in the colon. The combination of Ph-NSLs and 5FU showed more pronounced anticancer activity than Ph-NSLs administered individually. Conclusion The combination of 5-FU and Ph-NSLs significantly enhances anticancer efficacy, reducing both the number of tumors and average tumor size more effectively than either treatment alone. This synergistic effect leverages 5-FU's inhibition of DNA synthesis and phloretin's induction of apoptosis and inhibition of cell proliferation, offering a promising approach for improved cancer treatment outcomes.
Collapse
Affiliation(s)
- Ebtsam A. Abdel-Wahab
- Department of Biophysics, Faculty of Applied Health Sciences, October 6 University, Egypt
| | - Zahraa Haleem Al-Qaim
- Anesthesia Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001 Babylon, Iraq
| | | | - Aysam M. Fayed
- Medical Laboratories Techniques Department, AL-Mustaqbal University, 51001 Hillah, Babil, Iraq
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Ahmed M. Eldmrdash
- Department of Medical Labs, Faculty of Applied Medical Sciences Technology, October 6 University, Egypt
| | - Mohammed Abdalla Hussein
- Department of Biotechnology, Faculty of Applied Health Sciences Technology, October 6 University, Egypt
| | - Amal Abdel-Aziz
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Azza M. Metwaly
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Heba.G. Abdelzaher
- Department of Clinical pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - M.A. Abdelzaher
- Environmental Science and Industrial Development Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Diana A. ALsherif
- Technology of Radiology and Medical Imaging Department, Faculty of Applied Health Science Technology, October 6 University, Egypt
| |
Collapse
|
11
|
Li Y, Wang X, Wang X, Qin Z, Li C, Yang J, Cao M. Electrochemical biosensor based on composite of gold nanoparticle/reduced-graphene oxide/graphitic carbon nitride and a caprolactone polymer for highly sensitive detection of CEA. Bioelectrochemistry 2025; 163:108897. [PMID: 39764934 DOI: 10.1016/j.bioelechem.2024.108897] [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: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 02/12/2025]
Abstract
Carcinoembryonic antigen (CEA) is a broad-spectrum biomarker, and its accurate detection and analysis is important for early clinical diagnosis and treatment. This study aimed to develop a highly sensitive and selective sandwich-type immunosensor based on electrochemical impedance spectroscopy (EIS) for the accurate detection of CEA. A novel composite material, gold nanoparticle/reduced-graphene oxide/graphitic carbon nitride (AuNPs/rGO/g-C3N4), was synthesized with excellent electrical conductivity and a large specific surface area to immobilize biological probes. And ab1-CEA-ab2 formed a sandwich structure of 'antibody-antigen-antibody', which ensured the high selectivity of the biosensor. Furthermore, the introduction of caprolactone polymer (DMPA-PCL) significantly amplifies the impedance signal and improves the sensitivity of the analytical method. Scanning electron microscopy, x-ray diffraction, transmission electron microscopy Fourier transform infrared spectroscopy, and ultraviolet-visible spectrophotometry were used to characterise the prepared AuNPs/rGO/g-C3N4 and DMPA-PCL. Under the optimal conditions, the sensor showed good analytical performance for the detection of CEA with a linear range of 100 fg mL-1-100 ng mL-1 and a detection limit of 83.2 fg mL-1. And the sandwich-type immunosensor showed good selectivity and stability for the recognition of CEA in real samples.
Collapse
Affiliation(s)
- Yunpeng Li
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Xia Wang
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China.
| | - Xinling Wang
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China.
| | - Zhe Qin
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Chong Li
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Jing Yang
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Mengmeng Cao
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| |
Collapse
|
12
|
Liu Q, Jia SX, Chi QN, Jin L, Chen XQ, Li J, Qi YK, Du SS. Efficient synthesis, stability-guided optimization and anticancer evaluation of bee venom peptide Melittin. Bioorg Chem 2025; 159:108344. [PMID: 40086188 DOI: 10.1016/j.bioorg.2025.108344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Natural cytotoxic peptides (NCPs) are emerging sources of novel anticancer chemotherapeutics. Especially, Melittin, which is the major component of bee venom and the first-in-class NCP, has been considered as a promising anticancer scaffold. Nevertheless, as a classical linear, cationic, amphipathic, and membrane-lytic peptide, Melittin may be easily degraded by proteases, suffering from poor stability, moderate anticancer durability, and severe hemolysis. In this study, applying the terminal modification and hybridization strategies, ten Melittin-based derivatives were designed, synthesized, and investigated for their anticancer potential. The robust and economic synthetic method, in vitro anticancer efficiency, time-kill kinetics, serum stability, anti-migration activity, hemolysis effects, and anticancer mechanism were explored. As expected, the Melittin-based derivatives exhibited highly potent cytotoxicity against all six tested cancer cell lines. In particular, compared with natural Melittin, the derived peptides LJ-5 containing both N-terminal acetylation and C-terminal hydrazidation, and LJ-6, the methotrexate MTX-GFLG-Melittin conjugate exhibited significantly improved proteolytic stability, more durable anticancer efficiency, higher anti-migration activity, as well as reduced hemolysis effects. Besides, it was further verified that LJ-5 and LJ-6 could efficiently disrupt the integrity of cancer cell membrane, localize to the mitochondria and rapidly reduce the mitochondrial membrane potential of cancer cells. Collectively, the economic synthetic method and stability-guided optimization were conducted on Melittin, affording hydrolysis-resistant LJ-5 and LJ-6 that may serve as anticancer candidates and useful references for further optimizations of cytotoxic peptides.
Collapse
Affiliation(s)
- Qing Liu
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Shi-Xi Jia
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Qiao-Na Chi
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Lan Jin
- National Glycoengineering Research Center and NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong 266237, China
| | - Xin-Qi Chen
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jiamin Li
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yun-Kun Qi
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, #1 Ningde Road, Qingdao 266073, China.
| | - Shan-Shan Du
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, #1 Ningde Road, Qingdao 266073, China.
| |
Collapse
|
13
|
Chang IY, Boo HJ, Hyun JW, Yoon SP. The feasible role of soluble E‑cadherin in spheroidogenesis of HCT116 colorectal cancer cells, a candidate biomarker for liquid biopsy. Oncol Lett 2025; 29:245. [PMID: 40182609 PMCID: PMC11967162 DOI: 10.3892/ol.2025.14991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/21/2025] [Indexed: 04/05/2025] Open
Abstract
Although E-cadherin is known as a tumor suppressor via its effects on cell to cell adhesion, the effects of E-cadherin on malignant transformation have not yet been thoroughly investigated. In the present study, after malignant transformation was induced by spheroid formation in a fetal bovine serum-supplemented environment, the effects of soluble E-cadherin on the spheroidogenesis of colorectal cancer cells were investigated. E-cadherin knock-out (KO) was performed in HCT116 cells, targeting exon 3 of the CDH1 gene. A cell viability assay was performed to determine the proliferation and viability of wild type and CDH1 KO HCT116 cells after treatment with anticancer drugs. Spheroidogenesis was compared with or without exogenous E-cadherin, antibody against the ectodomain of E-cadherin (DECMA-1) and PD98059 treatment. In addition, morphometry, immunocytochemistry and western blotting were performed. Soluble E-cadherin in culture media was measured using an enzyme-linked immunosorbent assay. Firstly, CDH1 KO was confirmed by western blotting. Notably, the proliferation and viability of cells following treatment with 5-fluorouracil, epidermal growth factor receptor inhibitor and src kinase inhibitor were similar between the cell lines. Exogenous E-cadherin or DECMA-1 treatment did not affect spheroidogenesis, although long-term maintenance was slightly disturbed in CDH1 KO spheroids compared with that in wild type spheroids. In addition, E-cadherin was increased in spheroid culture as compared with that in conventional culture. Soluble E-cadherin was increased in a time-dependent manner, particularly in wild type HCT116 cells. PD98059 inhibited ERK activation and enhanced E-cadherin expression in conventional culture without affecting spheroidogenesis. These results suggested that soluble E-cadherin may be considered as a biomarker for colorectal cancer, although exogenous E-cadherin might not have a further role in malignant transformation.
Collapse
Affiliation(s)
- In-Youb Chang
- Department of Anatomy, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Hye-Jin Boo
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jin Won Hyun
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Sang-Pil Yoon
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
- Department of Anatomy, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
14
|
Akkus E, Karaoğlan BB, Akçadağ B, Bahçekapılı B, Akyol C, Utkan G. Combined preoperative and post-adjuvant-chemotherapy carcinoembryonic antigen levels are prognostic for early recurrence and survival in stage III colon cancer. Am J Surg 2025; 243:116256. [PMID: 40015199 DOI: 10.1016/j.amjsurg.2025.116256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/13/2025] [Accepted: 02/07/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND The definitive treatment of stage-III colon cancer is surgery and adjuvant chemotherapy. A combined assessment of pre-operative and post-adjuvant chemotherapy carcinoembryonic antigen (CEA) levels may better prognosticate early recurrence and survival. METHODS A cohort of patients who underwent surgery and adjuvant chemotherapy was assessed. The CEA-Square (CEA2) score was defined as the multiplication of preoperative and post-adjuvant chemotherapy CEA levels and was grouped as "≤25(ng/mL)2" and ">25(ng/mL)2. RESULTS Among the 432 patients,137 were eligible. CEA2 score (>25 vs ≤ 25 (ng/mL)2) was significantly prognostic for early recurrence (34.5 % vs. 14.3 %, log-rank, p < 0.001). In the multivariable analysis, only the CEA2 score remained associated with early recurrence [HR:3.375, (95 % CI:1.488-7.655), p = 0.004]. In a median follow-up of 37.5 months (2.5-101.0), a high CEA2 score [>25 (ng/mL)2] was significantly associated with a worse OS (log-rank, p < 0.001). CONCLUSION CEA2 is a simple, practical score combining prognostic values of preoperative and post-adjuvant chemotherapy CEA levels.
Collapse
Affiliation(s)
- Erman Akkus
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University Cancer Research Institute, Ankara, Türkiye.
| | - Beliz Bahar Karaoğlan
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University Cancer Research Institute, Ankara, Türkiye
| | - Barış Akçadağ
- Ankara University Faculty of Medicine, Department of Internal Medicine, Ankara, Türkiye
| | - Barış Bahçekapılı
- Ankara University Faculty of Medicine, Department of Internal Medicine, Ankara, Türkiye
| | - Cihangir Akyol
- Ankara University Faculty of Medicine, Department of General Surgery, Ankara, Türkiye
| | - Güngör Utkan
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University Cancer Research Institute, Ankara, Türkiye
| |
Collapse
|
15
|
van Stigt BJ, Lansdorp‐Vogelaar I, Spaander MCW, van Vuuren AJ, Dekker E, van Kemenade FJ, Nagtegaal ID, van Leerdam ME, Toes‐Zoutendijk E. Interval cancer risk after the upper age limit of screening has been reached: Informing risk stratification in FIT-based colorectal cancer screening. Int J Cancer 2025; 156:1783-1790. [PMID: 39697047 PMCID: PMC11887016 DOI: 10.1002/ijc.35294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024]
Abstract
Upper age limits are currently fixed for all fecal immunochemical test (FIT)-based colorectal cancer (CRC) screening programs. A risk-stratified upper age limit may be beneficial. Therefore, we assessed differences in interval CRC risk among individuals who had reached the upper age limit of screening (75 years). Individuals with a negative FIT (<47 μg Hb/g feces) in the final round of the Dutch CRC screening program were selected from the national screening database and linked to the national cancer registry to identify CRCs diagnosed within 24 months (interval CRCs). Survival analyses assessed whether sex and last fecal hemoglobin (f-Hb) concentration were associated with interval CRC risk. A multivariable logistic regression assessed whether sex, last f-Hb concentration and screening round were associated with stage distribution (early vs. late). Last f-Hb concentrations were considered detectable when they were >0 μg Hb/g feces. Among 305,761 individuals with a complete follow-up (24 months), 661 were diagnosed with interval CRC (21.6 per 10,000 negative FITs). Individuals with detectable f-Hb (15%) were 5 times more likely to be diagnosed with interval CRC than those without (HR 4.87, 95%CI: 4.19-5.65). Moreover, their cancers were more often detected at a late stage compared to individuals without detectable f-Hb (OR 1.45, 95%CI: 1.06-2.01). Our results show that interval CRC risk among individuals aged ≥75 differs substantially by last f-Hb concentration, indicating a uniform age to stop screening is suboptimal. Future research, taking into account multiple screening rounds and FIT results, should determine the optimal risk-stratified screening strategy.
Collapse
Affiliation(s)
- Brenda J. van Stigt
- Department of Public HealthErasmus MC University Medical CentreRotterdamThe Netherlands
| | | | - Manon C. W. Spaander
- Department of Gastroenterology and HepatologyErasmus MC University Medical CentreRotterdamThe Netherlands
| | - Anneke J. van Vuuren
- Department of Gastroenterology and HepatologyErasmus MC University Medical CentreRotterdamThe Netherlands
| | - Evelien Dekker
- Department of Gastroenterology and HepatologyAmsterdam University Medical CenterAmsterdamThe Netherlands
| | | | - Iris D. Nagtegaal
- Department of PathologyRadboud University Medical CentreNijmegenThe Netherlands
| | - Monique E. van Leerdam
- Department of Gastrointestinal OncologyAntoni van Leeuwenhoek HospitalAmsterdamThe Netherlands
- Department of Gastroenterology and HepatologyLeiden University Medical CentreLeidenThe Netherlands
| | | |
Collapse
|
16
|
Gao H, Zheng S, Liang J, Wang Y, Chen L, Li H, Chen Y, Zhang F, Shi H, Han A. m6A-induced DEAD-box RNA helicase 21 enhances lipid metabolism via 3‑hydroxy-3-methylglutaryl-CoA synthases 1 in colorectal cancer. Transl Oncol 2025; 55:102373. [PMID: 40127603 PMCID: PMC11979938 DOI: 10.1016/j.tranon.2025.102373] [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: 11/06/2024] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Altered lipid metabolism is a well-known hallmark of cancer. However, the underlying mechanisms of altered lipid metabolism in colorectal cancer (CRC) progression requires further investigation. Previously we have revealed that DEAD-box RNA helicase 21 (DDX21) promotes CRC metastasis via liquid-liquid phase separation. In this study, we identify DDX21 as a novel regulator of lipid metabolism in CRC. METHODS In vitro and in vivo assays illustrated the biological role of DDX21 and YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) in CRC lipid metabolism and progression. Bioinformatics analysis, ChIP, meRIP, RIP, RNA stability assay and dual-luciferase reporter assay were applied to explore the underlying molecular mechanisms. The expression levels and prognostic role of YTHDF1/DDX21/HMGCS1 axis in CRC patients were analyzed by immunohistochemical staining and Kaplan-Meier plotter. RESULTS DDX21 enhanced CRC progression via promoting lipid metabolism. Mechanistically, YTHDF1 enhanced DDX21 mRNA stability by recognizing its m6A-modified sites. DDX21 further binded to 3‑hydroxy-3-methylglutaryl-CoA synthase 1 (HMGCS1) promoter region and directly activated HMGCS1 transcription. Moreover, our clinical data showed that a simultaneously high expression of YTHDF1, DDX21 and HMGCS1 predicted an unfavorable overall survival in CRC patients. CONCLUSIONS Our study demonstrates that the YTHDF1/DDX21/HMGCS1 axis promotes CRC progression via regulating lipid metabolism and DDX21 might be a promising target for CRC therapy.
Collapse
Affiliation(s)
- Huabin Gao
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Shuai Zheng
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jiangtao Liang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yuting Wang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Lin Chen
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Hui Li
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yongyu Chen
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Fenfen Zhang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Huijuan Shi
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Anjia Han
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
17
|
Thomas CE, Peters U. Genomic landscape of cancer in racially and ethnically diverse populations. Nat Rev Genet 2025; 26:336-349. [PMID: 39609636 DOI: 10.1038/s41576-024-00796-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 11/30/2024]
Abstract
Cancer incidence and mortality rates can vary widely among different racial and ethnic groups, attributed to a complex interplay of genetic, environmental and social factors. Recently, substantial progress has been made in investigating hereditary genetic risk factors and in characterizing tumour genomes. However, most research has been conducted in individuals of European ancestries and, increasingly, in individuals of Asian ancestries. The study of germline and somatic genetics in cancer across racial and ethnic groups using omics technologies offers opportunities to identify similarities and differences in both heritable traits and the molecular features of cancer genomes. An improved understanding of population-specific cancer genomics, as well as translation of those findings across populations, will help reduce cancer disparities and ensure that personalized medicine and public health approaches are equitable across racial and ethnic groups.
Collapse
Affiliation(s)
- Claire E Thomas
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Epidemiology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
18
|
Chen Y, Huang J, Fan Y, Huang L, Cai X. Understanding the cellular and molecular heterogeneity in colorectal cancer through the use of single-cell RNA sequencing. Transl Oncol 2025; 55:102374. [PMID: 40163910 PMCID: PMC11993189 DOI: 10.1016/j.tranon.2025.102374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
The very prevalent nature, genetic variability, and intricate tumor microenvironment (TUME) of colorectal cancer (COREC) are its defining features. In order to better understand the molecular and cellular make-up of COREC, this work used single-cell RNA sequencing (SRNAS) to isolate and characterize important cell types as well as their interactions within the TUME. Our analysis of 51,204 cells yielded six distinct types: epithelial, fibroblast, endothelial, T&NK, B, and myeloid. C3 B cells were shown to be the most active in immunological regulation, according to chemokine signaling study, which was one of seven clusters of B cells that were thoroughly subtyped. The examination of copy number variation (CONUV) revealed a great deal of genetic variability, especially in epithelial cells. We traced the activity of three key transcription factor clusters (M1, M2, and M3) across all B cell subtypes using transcription factor analysis. We created a predictive model that correctly sorts patients according to survival results by using marker genes from C3 B cells. In addition, the relationship between genetic changes and the immune system was better understood by tumor mutational burden (TUMUB) and immune infiltration studies. Our research sheds light on the genetic complexity and cellular variety of COREC, which in turn opens up new possibilities for targeted treatments and individualized approaches to patient care.
Collapse
Affiliation(s)
| | - Jian Huang
- Wenzhou Central Hospital, Wenzhou, China
| | - Yufang Fan
- Wenzhou Central Hospital, Wenzhou, China
| | | | | |
Collapse
|
19
|
Li ZF, Zhang JN, Tian S, Sun C, Ma Y, Ye ZX. Dual-Time-Point Radiomics for Prognosis Prediction in Colorectal Liver Metastasis Treated with Neoadjuvant Therapy Before Radical Resection: A Two-Center Study. Ann Surg Oncol 2025; 32:3516-3525. [PMID: 39907877 DOI: 10.1245/s10434-025-16941-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Optimal prognostic stratification for colorectal liver metastases (CRLM) patients undergoing surgery with neoadjuvant therapy (NAT) remains elusive. This study aimed to develop and validate dual-time-point radiomic models for CRLM prognosis prediction using pre- and post-NAT imaging features. METHODS Radiomic features were extracted from four MRI sequences in 100 cases of CRLM patients who underwent NAT and radical resection. RAD scores were generated, and clinical/pathologic variables were incorporated into uni- and multivariate Cox regression analyses to construct prognosis models. Time-ROC, time-C index, decision curve analysis (DCA), and calibration curves assessed the predictive performance of Fong score and pre- and post-NAT models for overall survival (OS) and disease-free survival (DFS) in a testing set. RESULTS The final models included four variables for OS and three variables for DFS. The post-NAT models outperformed the pre-NAT models in time-ROC, time-C index, calibration, and DCA analysis, except for the 1-year DFS area under the curve (AUC). The Fong score models underperformed. The post-NAT OS RAD score effectively stratified patients into prognostic subgroups. CONCLUSIONS The radiomic models incorporating pre- and post-NAT MRI features and clinical/pathologic variables effectively stratified CRLM patients prognositically. The post-NAT models demonstrated superior performance.
Collapse
Affiliation(s)
- Zhuo-Fu Li
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China
| | - Jia-Ning Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China
| | | | - Chao Sun
- Department of Radiology, Tianjin Union Medical Center, Tianjin, People's Republic of China
| | - Ying Ma
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhao-Xiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China.
| |
Collapse
|
20
|
Borhani AA, Zhang P, Diergaarde B, Darwiche S, Chuperlovska K, Wang SC, Schoen RE, Su GL. Role of tumor-specific and whole-body imaging biomarkers for prediction of recurrence in patients with stage III colorectal cancer. Abdom Radiol (NY) 2025; 50:1907-1915. [PMID: 39487920 DOI: 10.1007/s00261-024-04656-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Imaging biomarkers are emerging as non-invasive predictors of cancer prognosis and clinical outcome. We assessed tumor-specific ("radiomics") and body composition imaging features ("morphomics") extracted from baseline pre-treatment CT for prediction of recurrence in patients with stage III colorectal cancer (CRC). METHODS Patients with newly diagnosed stage III CRC were enrolled in this prospective observational study. Patients with available preoperative scans were included (N = 101). The tumor, if visible, was manually segmented and first-order radiomics features were extracted with a commercially available software. The morphomics features (reflecting muscle, fat, and bone characteristics) were extracted in a standardized fashion using a proprietary software and the values were adjusted and normalized based on a reference standard. Time to recurrence was the final outcome. Correlation between demographics, clinical features, radiomics, and morphomics features and outcome were assessed using univariate and multivariate tests as well as Kaplan-Meier and log-rank tests. RESULTS Morphomic analysis was performed in all 101 patients. 60 patients had discrete tumors suitable for radiomics analysis. These patients had lower ECOG score (p < 0.05), more muscle mass (p > 0.05), and lower fat density (p > 0.05) compared to the patients in whom radiomics analysis could not be performed. Pathological stage (HR: 2.69; p = 0.03), CEA level after surgery (HR: 1.11 for 1 ng/mL; p < 0.005), bone mineral density (HR: 1.01 for 1 Hounsfield Unit; p < 0.01), and tumor skewness (HR: 0.33 for 1 unit; p < 0.05) had association with recurrence based on both univariate and multivariate analyses. A model using Cox's regression analyses was able to divide the patients into low-, medium-, and high-risk for recurrence. CONCLUSIONS Both radiomics and morphomics features were independently associated with the risk of CRC recurrence and, when combined, each contributed valuable information to explain risk of recurrence. TRIAL REGISTRATION Clinical trial.gov NCT02842203. Patient recruitment occurred between 22/07/2016 and 18/03/2020.
Collapse
Affiliation(s)
- Amir A Borhani
- Northwestern University, Evanston, USA.
- University of Pittsburgh, Pittsburgh, USA.
| | - Peng Zhang
- University of Michigan-Ann Arbor, Ann Arbor, USA
| | | | | | | | | | | | - Grace L Su
- University of Michigan-Ann Arbor, Ann Arbor, USA
| |
Collapse
|
21
|
Zhan Z, Chen B, Lin W, Chen X, Huang R, Yang C, Guo Z. Rising Burden of Colon and Rectum Cancer in China: An Analysis of Trends, Gender Disparities, and Projections to 2030. Ann Surg Oncol 2025; 32:3361-3371. [PMID: 39836276 DOI: 10.1245/s10434-025-16905-w] [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: 08/19/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Colon and rectum cancer (CRC) is a major health burden in China, with notable gender disparities. This study was designed to analyze trends in CRC incidence, prevalence, and mortality from 1990 to 2021 and to project future trends. METHODS Using data from the Global Burden of Disease (GBD) Study 2021, we examined CRC burden in China, including incidence, prevalence, mortality, disability-adjusted life years (DALYs), years lived with disability (YLDs), and years of life lost (YLLs). Joinpoint regression, Bayesian age-period-cohort (BAPC) models, and age-period-cohort (APC) analysis identified trends and projected incidence up to 2030. RESULTS In 2021, CRC incidence was 658,321 cases, disproportionately affecting males, with an age-standardized incidence rate of 42.24 per 100,000 in males and 21.87 per 100,000 in females. The CRC-related deaths reached 275,129, with higher mortality in males (18.95 per 100,000) than females (9.34 per 100,000). The DALYs totaled 6,848,390 and largely impacted males. Joinpoint analysis showed a persistent increase in incidence and prevalence, especially in younger cohorts, whereas mortality declined slightly but began rising again after 2015. The BAPC analysis projected further incidence growth, particularly in males, through 2030. The APC analysis revealed higher CRC risk among younger cohorts, suggesting increasing early-onset CRC linked to lifestyle risk factors, such as smoking, high alcohol consumption, and diets low in fiber and calcium, with a stronger effect on males. CONCLUSIONS The increase of CRC incidence and prevalence in China, particularly among males, underscores the need for targeted prevention and early detection. Future research should address gender disparities and modifiable lifestyle risks through public health interventions.
Collapse
Affiliation(s)
- Zhouwei Zhan
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Bijuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Wei Lin
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Xiamei Chen
- Department of Operation, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Rui Huang
- Digestive Endoscopy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Chunkang Yang
- Department of Gastrointestinal Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| | - Zengqing Guo
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
22
|
Duffy L, Anderson M, Rowe E, Yan M, Abdelhak A, Garcia J, Muller C, Kumar A. Colorectal cancer care equity in underserved communities: Innovative solutions for screening, outreach & capacity in rural Washington. Am J Surg 2025; 243:116246. [PMID: 39986209 DOI: 10.1016/j.amjsurg.2025.116246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) mortality hotspots in rural Washington, notably Yakima and Richland, identified an 8-year earlier median age of death in non-white patients. Post-pandemic data from WA State Department of Health and MultiCare's electronic health records revealed a 40 % decrease in CRC screenings. METHODS During a 2023 CRC prevention summit, barriers and solutions were discussed focusing on rural Hispanic laborers as this population is often seen at partner locations, Yakima Valley Farm Workers Clinic (YVFWC). RESULTS Community outreach-Our team established a presence at Yakima's largest health fair, Fiesta de Salud. We also provided screening resources at Richland's farmers' markets. Planned events-- We will network with local community leaders and health providers to actualize screening (stool-based testing, hands-on endoscopy training). CONCLUSION Persistent CRC mortality disparities in Washington State underscores the need for targeted interventions. Partnering with organizations and engaging in community outreach aims to increase screening rates and enhance education.
Collapse
Affiliation(s)
- Lauren Duffy
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Marley Anderson
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Evelyn Rowe
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Michelle Yan
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Aliya Abdelhak
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Juliana Garcia
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Clemma Muller
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Anjali Kumar
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA.
| |
Collapse
|
23
|
Babaei M, Abrishami A, Iranpour S, Saljooghi AS, Matin MM. Harnessing curcumin in a multifunctional biodegradable metal-organic framework (bio-MOF) for targeted colorectal cancer theranostics. Drug Deliv Transl Res 2025; 15:1719-1738. [PMID: 39302530 DOI: 10.1007/s13346-024-01707-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
Despite significant advancements in managing colorectal cancer (CRC), the issues of efficient diagnosis and targeted therapy remain demanding. To address these challenges and improve treatment outcomes while reducing the cost and side effects, there is a need for more effective theranostic systems that combine diagnostic techniques with therapeutic modalities. This study introduces a pioneering approach for the synthesis of a porous bio-MOF (biodegradable metal-organic framework) using iron as the metal component and curcumin as the pharmaceutical ingredient. Subsequently, the developed drug delivery system was equipped with the anticancer drug doxorubicin (DOX), coated with biocompatible polyethylene glycol (PEG), and targeted with a CRC-specific aptamer (EpCAM). The physicochemical characterization confirmed the successful synthesis of the bio-MOF, demonstrating high encapsulation efficiency and pH-dependent release of DOX. In vitro studies for anticancer activity, cellular uptake, and mechanism of cell death demonstrated that in the case of positive EpCAM HT-29 cells, Apt-PEG-MOF@DOX had enhanced internalization that resulted in massive apoptosis. In vivo studies of the nanoparticles were then conducted in immunocompromised C57BL/6 mice bearing HT-29 tumors. These studies showed that the targeted platform could induce efficient tumor regression with reduced systemic toxicity. The targeted bio-MOF also exhibited MRI imaging properties useful for monitoring tumors. Significantly, the biocompatibility of the introduced bio-MOF was enhanced by pursuing the green synthesis method, which does not engage toxic solvents and strong acids. Overall, this multimodal system acts diversely as a tumor imaging agent and a therapeutic delivery platform suitable for CRC theranostics.
Collapse
Affiliation(s)
- Maryam Babaei
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Abrishami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sonia Iranpour
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Sh Saljooghi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
24
|
Spolverato G, Capelli G, Noel F, Steindler M, Gumbs AA. Pan-immune-inflammation in colon cancer: A prognostic biomarker and the role of tumor location in personalized care. World J Gastrointest Surg 2025; 17:101066. [DOI: 10.4240/wjgs.v17.i4.101066] [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: 09/03/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Despite advances in surgery, chemotherapy, and radiotherapy, the treatment of colorectal cancer (CRC) requires more personalized approaches based on tumor biology and molecular profiling. While some relevant mutations have been associated with differential response to immunotherapy, such as RAS and BRAF mutations limiting response to anti-epithelial growth factor receptor drugs or microsatellite instability predisposing susceptibility to immune checkpoint inhibitors, the role of inflammation in dictating tumor progression and treatment response is still under investigation. Several inflammatory biomarkers have been identified to guide patient prognosis. These include the neutrophil-lymphocyte ratio, Glasgow prognostic score (GPS) and its modified version, lymphocyte-C-reactive protein ratio, and platelet-lymphocyte ratio. However, these markers are not yet included in the standard clinical management of patients with CRC, and further research is needed to evaluate their efficacy in different patient populations. A recent study by Wang et al, published in the World Journal of Gastroenterology, sheds light on the prognostic significance of pan-immune-inflammation value (PIV) in CRC, particularly concerning primary tumor location. Specifically, the authors found that a high PIV was strongly correlated with worse disease-free survival in patients with left-sided colon cancer, whereas no such association was observed in patients with right-sided colon cancer. Integrating tumor location into the prognostic assessment of CRC may improve our ability to more accurately identify high-risk patients and develop personalized treatment plans that are more likely to improve patient outcomes.
Collapse
Affiliation(s)
- Gaya Spolverato
- Department of Surgery, University of Padova, Padua 35122, Italy
| | - Giulia Capelli
- Department of Surgery, ASST Bergamo Est, Bergamo 24068, Lombardy, Italy
| | - Floriane Noel
- Department of Research, Sibylone, Paris 75002, France
| | | | - Andrew Alexander Gumbs
- Department of Surgery, University of Magdeburg, Magdeburg 39130, Saxony-Anhalt, Germany
- Department of Surgery, Service de Chirurgie Digestive Minimale Invasive, Hôpital Antoine Béclère, Assistance Publique-Hôpitaux de Paris, Clamart 92140, France
| |
Collapse
|
25
|
Zhou Y, Gao Y, Peng Y, Cai C, Han Y, Chen Y, Deng G, Ouyang Y, Shen H, Zeng S, Du Y, Xiao Z. QKI-induced circ_0001766 inhibits colorectal cancer progression and rapamycin resistance by miR-1203/PPP1R3C/mTOR/Myc axis. Cell Death Discov 2025; 11:192. [PMID: 40263288 DOI: 10.1038/s41420-025-02478-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/25/2025] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and remains a significant challenge due to high rates of drug resistance and limited therapeutic options. Circular RNAs (circRNAs) are increasingly recognized for their roles in CRC initiation, progression, and drug resistance. However, no circRNA-based therapies have yet entered clinical development, underscoring the need for comprehensive detection and mechanistic studies of circRNAs in CRC. Here, we identified and characterized a circular RNA, circ_0001766 (hsa_circ_0001766), through microarray analysis of CRC tissues. Our results showed that circ_0001766 is downregulated in CRC tissues and closely associated with patient survival and metastasis. Functional experiments demonstrated that circ_0001766 inhibits CRC cell proliferation, migration and invasion both in-vitro and in-vivo. Mechanistically, hypoxia downregulates Quaking (QKI), an RNA-binding protein essential for the biogenesis of circ_0001766 by binding to introns 1 and 3 of PDIA4 pre-mRNA. Reduced QKI expression under hypoxic conditions leads to decreased circ_0001766 levels in CRC. Circ_0001766 acts as a competitive endogenous RNA, sponging miR-1203 to prevent the degradation of PPP1R3C mRNA. Loss of circ_0001766 results in decreased PPP1R3C expression, leading to the activation of mTOR signaling and increased phosphorylation of Myc, which promotes CRC progression and rapamycin resistance. Our study reveals that overexpression of circ_0001766 or PPP1R3C in CRC cells inhibits the mTOR and Myc pathway, thereby resensitizing cells to rapamycin. The combination of circ_0001766 or PPP1R3C with rapamycin markedly inhibits CRC cell proliferation and induces apoptosis by reducing rapamycin-induced Myc phosphorylation. In summary, our study elucidates a critical circ_0001766/miR-1203/PPP1R3C axis that modulates CRC progression and rapamycin resistance. Our findings highlight circ_0001766 as a promising therapeutic target in CRC, providing a new avenue for enhancing the efficacy of existing treatments and overcoming drug resistance.
Collapse
Affiliation(s)
- 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, UT Health Science Center, San Antonio, TX, USA
| | - 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
| | - 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
| | - 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
| | - Yihong Chen
- 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
| | - Gongping Deng
- Department of Emergency, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Yanhong Ouyang
- Department of Emergency, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Hainan, 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
| | - Yangfeng Du
- Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China.
| | - Zemin Xiao
- Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China.
| |
Collapse
|
26
|
Abdel Hamid M, Pammer LM, Oberparleiter S, Günther M, Amann A, Gruber RA, Mair A, Nocera FI, Ormanns S, Zimmer K, Gerner RR, Kocher F, Vorbach SM, Wolf D, Riedl JM, Huemer F, Seeber A. Multidimensional differences of right- and left-sided colorectal cancer and their impact on targeted therapies. NPJ Precis Oncol 2025; 9:116. [PMID: 40263545 DOI: 10.1038/s41698-025-00892-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/31/2025] [Indexed: 04/24/2025] Open
Abstract
Despite advances in metastatic colorectal cancer (mCRC) treatment, long-term survival remains poor, particularly in right-sided colorectal cancer (RCRC), which has a worse prognosis compared to left-sided CRC (LCRC). This disparity is driven by the complex biological diversity of these malignancies. RCRC and LCRC differ not only in clinical presentation and outcomes but also in their underlying molecular and genetic profiles. This article offers a detailed literature review focusing on the distinctions between RCRC and LCRC. We explore key differences across embryology, anatomy, pathology, omics, and the tumor microenvironment (TME), providing insights into how these factors contribute to prognosis and therapeutic responses. Furthermore, we examine the therapeutic implications of these differences, considering whether the conventional classification of CRC into right- and left-sided forms should be refined. Recent molecular findings suggest that this binary classification may overlook critical biological complexities. Therefore, we propose that future approaches should integrate molecular insights to better guide personalized treatments, especially anti-EGFR therapies, and improve patient outcomes.
Collapse
Affiliation(s)
- Marwa Abdel Hamid
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Lorenz M Pammer
- Department of Gastroenterology and Hepatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Silvia Oberparleiter
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Günther
- INNPATH, Institute of Pathology, Tirol Kliniken GmBH, Innsbruck, Austria
| | - Arno Amann
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Rebecca A Gruber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Mair
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Fabienne I Nocera
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Steffen Ormanns
- INNPATH, Institute of Pathology, Tirol Kliniken GmBH, Innsbruck, Austria
| | - Kai Zimmer
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Romana R Gerner
- Department of Medicine III, Hematology and Oncology, University Hospital Rechts der Isar, Technical University of Munich, Munich, Germany
- TUM School of Life Sciences Weihenstephan, ZIEL Institute for Food & Health, 85354, Freising, Germany
| | - Florian Kocher
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Samuel M Vorbach
- Department of Radiation Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dominik Wolf
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Jakob M Riedl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florian Huemer
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Center for Clinical Cancer and Immunology Trials (CCCIT), Paracelsus Medical University, Salzburg, Austria
| | - Andreas Seeber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria.
- Department of Oncology, Hematology and Palliative Care, General Hospital Oberwart, Oberwart, Austria.
| |
Collapse
|
27
|
Qin ZF, Gao XY, Zhu YZ, Ma T, Chen GL, Ma DN, Zhu HZ, Feng LH. Evaluation of LNR and modified N stage systems for prognostic stratification of metastatic lymph nodes in stage III colorectal Cancer. Sci Rep 2025; 15:13735. [PMID: 40258878 PMCID: PMC12012119 DOI: 10.1038/s41598-025-96652-w] [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: 06/10/2024] [Accepted: 03/31/2025] [Indexed: 04/23/2025] Open
Abstract
Lymph node ratio (LNR) and log odds of positive lymph nodes (LOODS) are considered to be potentially more rigorous metastatic lymph node stratification patterns than AJCC N stage due to the integration of the number of examined lymph nodes. Based on the data of colorectal cancer obtained from the Surveillance, Epidemiology, and End Result (SEER) database, comparison of the stratification performance between different prognostic models was validated by statistical methods, includingchi-square test, Cox regression analysis, Kaplan-Meier survival analysis, and Time-dependent Area Under Curve. A prognostic outcome-oriented statistical method based on Log-rank test was performed to determine the cutoff value of LNR. External validation cohort obtained from Zhejiang Cancer Hospital was used to validate the model performance and cutoff value. In the SEER database cohort, the optimal cutoff value for adequate or inadequate lymph node clearance was 14. In performance comparisons, LNR performance was superior to lymph node count alone and similar to LODDS. The optimal cutoff values for LNR were analyzed to be 0.11 (P < 0.001), 0.39 (P < 0.001) and 0.68 (P < 0.001), respectively. The stratification performance of modified N stage phased with the above cutoff value outperforms AJCC N stage. Kaplan-Meier survival analysis of the differences in patient distribution between the two staging systems also demonstrated the excellence of the LNR system. Above results were verified in the external verification cohort. The modified N stage based on LNR has better stratification performance than the AJCC N stage.
Collapse
Affiliation(s)
- Zhao-Fu Qin
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Xin-Yi Gao
- Department of Radiology, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Yan-Zhi Zhu
- School of Pharmacy, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Ting Ma
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Guan-Liang Chen
- Department of General Surgery, Affiliated Hospital of Shaoxing University, Yuecheng District, Zhong Xing Nan Road No. 999, Shaoxing, 312000, China
| | - De-Ning Ma
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
- Postgraduate training base Alliance (Zhejiang Cancer Hospital), Wenzhou Medical University, Hangzhou, Zhejiang Province, China
| | - Hong-Zhou Zhu
- Department of Radiology, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China.
| | - Long-Hai Feng
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
28
|
Bresesti C, Carito E, Notaro M, Giacca G, Breggion S, Kerzel T, Mercado CM, Beretta S, Monti M, Merelli I, Canu T, Naldini L, Squadrito ML. Reprogramming liver metastasis-associated macrophages toward an anti-tumoral phenotype through enforced miR-342 expression. Cell Rep 2025; 44:115592. [PMID: 40253698 DOI: 10.1016/j.celrep.2025.115592] [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: 08/13/2024] [Revised: 01/24/2025] [Accepted: 03/29/2025] [Indexed: 04/22/2025] Open
Abstract
Upon metastatic seeding in the liver, liver macrophages, including Kupffer cells, acquire a transcriptional profile typical of tumor-associated macrophages (TAMs), which support tumor progression. MicroRNAs (miRNAs) fine-tune TAM pro-tumoral functions, making their modulation a promising strategy for macrophage reprogramming into an anti-tumoral phenotype. Here, we analyze the transcriptomic profiles of liver and splenic macrophages, identifying miR-342-3p as a key regulator of liver macrophage function. miR-342-3p is highly active in healthy liver macrophages but significantly downregulated in colorectal cancer liver metastases (CRLMs). Lentiviral vector-engineered liver macrophages enforcing miR-342-3p expression acquire a pro-inflammatory phenotype and reduce CRLM growth. We identify Slc7a11, a cysteine-glutamate antiporter linked to pro-tumoral activity, as a direct miR-342-3p target, which may be at least partially responsible for TAM phenotypic reprogramming. Our findings highlight the potential of in vivo miRNA modulation as a therapeutic strategy for TAM reprogramming, offering an approach to enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Chiara Bresesti
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Eleonora Carito
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Marco Notaro
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giovanna Giacca
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Sara Breggion
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Thomas Kerzel
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Carl Mirko Mercado
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Stefano Beretta
- BioInformatics Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marco Monti
- BioInformatics Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ivan Merelli
- BioInformatics Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Tamara Canu
- Preclinical Imaging Facility, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Luigi Naldini
- Vita-Salute San Raffaele University, 20132 Milan, Italy; Targeted Cancer Gene Therapy Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mario Leonardo Squadrito
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy.
| |
Collapse
|
29
|
Jiang L, Zhuang Z, Tang X, Zhang F, Zhu H, Xu X, Wang D, Zhang L. Diagnostic performance of node-RADS classification for primary lymph node assessment in rectal cancer: a modality benchmarking study. J Cancer Res Clin Oncol 2025; 151:145. [PMID: 40252124 PMCID: PMC12009234 DOI: 10.1007/s00432-025-06196-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/03/2025] [Indexed: 04/21/2025]
Abstract
PURPOSE To evaluate how well the Node Reporting and Data System (Node-RADS) diagnoses lymph node involvement (LNI) in the initial stages of rectal cancer, utilizing contrast-enhanced CT (CE-CT), T2-weighted MRI (T2WI) and contrast-enhanced T1-weighted MRI (T1CE). METHODS This retrospective study included 113 rectal cancer patients who underwent radical surgery without neoadjuvant therapy. Two radiologists independently assessed regional lymph nodes using the highest NODE-RADS classification and histopathology as reference criteria. Diagnostic performance was evaluated using receiver operating characteristic (ROC) curve analysis. Statistical analysis was performed using the McNemar test with Bonferroni correction for multiple comparisons. RESULTS Node-RADS showed improved diagnostic performance over short-axis diameter (SAD) in all modalities (AUC: 0.838 vs. 0.744 for CE-CT, 0.845 vs. 0.747 for T2WI, 0.853 vs. 0.786 for T1CE; all P < 0.05). The sensitivity and specificity of Node-RADS across three modalities ranged from 76.19 - 78.57% and 91.55 - 92.96%, respectively. Pairwise comparisons of sensitivity and specificity among the three modalities showed no significant differences after Bonferroni correction (all McNemar test P = 1.0). There was no significant difference in Node-RADS performance among the three modalities (all P > 0.05). The weighted kappa values were 0.742-0.798. CONCLUSION Node-RADS demonstrated superior diagnostic performance over SAD measurements and similar diagnostic effectiveness in assessing LNI for primary rectal cancer stages across CE-CT, T2WI, and T1CE.
Collapse
Affiliation(s)
- Li Jiang
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Zijian Zhuang
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Xi Tang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Fugang Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Haitao Zhu
- Institute of Radiology and Artificial Intelligence, Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Xuewen Xu
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Dongqing Wang
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China.
- Institute of Radiology and Artificial Intelligence, Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China.
| | - Lirong Zhang
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China.
- Institute of Radiology and Artificial Intelligence, Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China.
| |
Collapse
|
30
|
Zhong H, Li Y, Raza F, Xie J, Rong R, Qiu M, Su J. RBCs as a Bioinspired Drug Delivery System for Co-delivery of Irinotecan and Nanoalumina Enhances Colorectal Cancer Therapy. Mol Pharm 2025. [PMID: 40251121 DOI: 10.1021/acs.molpharmaceut.4c01396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2025]
Abstract
Colorectal cancer (CRC) is a malignant epithelial tumor with high morbidity and mortality. In CRC treatment, irinotecan (CPT-11) as a chemotherapeutic drug is widely applied. However, its half-life is short, leading to large dosages and severe side effects. Red blood cells (RBCs) are biocompatible drug carriers with high capacity, avoiding premature drug degradation and achieving slow drug release. Nanoalumina (AN) is an emerging immune adjuvant that can enhance the immune response. Here, we used RBCs as carriers and absorbed AN to construct AN-CPT-11-RBCs. CPT-11 would induce tumor cell death, releasing much tumor antigen, while AN would activate immune cells to recognize newly released antigens and induce lymphocyte proliferation, enhancing the antitumor effect simultaneously. With loading amounts of 4 mg of CPT-11 and 3 mg of AN per 109 RBCs, AN-CPT-11-RBCs had similar properties to natural RBCs. In vivo, AN-CPT-11-RBCs could circulate for 9 days and stimulate the proliferation of lymphocytes in the spleen and tumor tissue, having a higher tumor growth inhibition rate of 74.01% and a lower frequency of administration. In conclusion, AN-CPT-11-RBCs attain the co-delivery of CPT-11 and AN for the synergistic treatment of CRC.
Collapse
Affiliation(s)
- Hongyu Zhong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yichen Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiyuan Xie
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruonan Rong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
31
|
Tang L, Que H, Wei Y, Yang T, Tong A, Wei X. Replicon RNA vaccines: design, delivery, and immunogenicity in infectious diseases and cancer. J Hematol Oncol 2025; 18:43. [PMID: 40247301 PMCID: PMC12004886 DOI: 10.1186/s13045-025-01694-2] [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: 01/02/2025] [Accepted: 03/23/2025] [Indexed: 04/19/2025] Open
Abstract
Replicon RNA (RepRNA) represents a cutting-edge technology in the field of vaccinology, fundamentally transforming vaccine design and development. This innovative approach facilitates the induction of robust immune responses against a range of infectious diseases and cancers. RepRNA vaccines leverage the inherent capabilities of RNA-dependent RNA polymerase associated with self-replicating repRNA, allowing for extreme replication within host cells. This process enhances antigen production and subsequently stimulates adaptive immunity. Additionally, the generation of double-stranded RNA during RNA replication can activate innate immune responses. Numerous studies have demonstrated that repRNA vaccines elicit potent humoral and cellular immune responses that are broader and more durable than those generated by conventional mRNA vaccines. These significant immune responses have been shown to provide protection in various models for infectious diseases and cancers. This article will explore the design and delivery of RepRNA vaccines, the mechanisms of immune activation, preclinical studies addressing infectious diseases and tumors, and related clinical trials that focus on safety and immunogenicity.
Collapse
Affiliation(s)
- Lirui Tang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Haiying Que
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Ting Yang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, People's Republic of China.
| | - Aiping Tong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
32
|
Zhou H, Zhuang Y, Liang Y, Chen H, Qiu W, Xu H, Zhou H. Curcumin exerts anti-tumor activity in colorectal cancer via gut microbiota-mediated CD8 + T Cell tumor infiltration and ferroptosis. Food Funct 2025. [PMID: 40244948 DOI: 10.1039/d4fo04045g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Colorectal cancer (CRC), as a high-incidence malignancy, continues to present significant challenges in prevention, screening, and treatment. Curcumin (Cur) exhibits notable anti-inflammatory and anticancer properties. Despite its poor solubility in water and low bioavailability, high concentrations of Cur are detected in the gastrointestinal tract after oral administration, suggesting that it may directly interact with the gut microbiota and exert regulatory effects. This study aims to explore the mechanisms by which Cur improves CRC by modulating gut microbiota. Firstly, we evaluated the effect of Cur on CRC cell viability in vitro using the MTT assay, and the results showed a significant inhibitory effect on CRC cell growth. The IC50 values for Cur in CT26 and RKO cells were 23.52 μM, 16.11 μM, and 13.62 μM at 24, 48, and 72 hours, respectively, and 26.3 μM, 16.52 μM, and 14.22 μM at 24, 48, and 72 hours, respectively. Cur induced apoptosis and caused G2 phase cell cycle arrest in tumor cells. Subsequently, we established a CRC mouse model. Oral administration of Cur at 15 mg kg-1 and 30 mg kg-1 inhibited CRC progression, as evidenced by reduced tumor volume, histological analysis, immunohistochemistry, and an increased number of CD8+ T cells infiltrating the tumors. Ferroptosis in tumor cells was also observed. Cur partially restored the gut microbiota of CRC mice, altering the abundance and diversity of the gut microbiota and affecting serum metabolite distribution, with significant increases in the abundance of SCFA-producing microbes such as Lactobacillus and Kineothrix. To verify causality, we designed a fecal microbiota transplantation (FMT) experiment. Compared with CRC mice, the fecal microbiota from Cur-treated mice significantly alleviated CRC symptoms, including slowed tumor growth, enhanced CD8+ T cell tumor infiltration, and induced ferroptosis in tumor cells. Additionally, when gut microbiota was depleted with antibiotics, Cur's antitumor effects disappeared, suggesting that Cur mitigates CRC in a gut microbiota-dependent manner. These findings provide new insights into the mechanisms underlying CRC and propose novel therapeutic interventions, emphasizing the interaction between gut microbiota and immune responses within the tumor immune microenvironment (TIME).
Collapse
Affiliation(s)
- Hongli Zhou
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, 210023, Nanjing, China
| | - Yupei Zhuang
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, 210023, Nanjing, China
| | - Yuwei Liang
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, 210023, Nanjing, China
| | - Haibin Chen
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
| | - Wenli Qiu
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, 210023, Nanjing, China
| | - Huiqin Xu
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
| | - Hongguang Zhou
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, 210023, Nanjing, China
| |
Collapse
|
33
|
Shen P, Wu S, Chen Y, Feng G, Guo X, Chen Y, Wang Z, Shen Y, Wang H, Li K. Yi Gong San inhibits tumor immune escape by sensitizing colorectal cancer stem cells via the NF-κB pathway. Hereditas 2025; 162:64. [PMID: 40247422 PMCID: PMC12004741 DOI: 10.1186/s41065-025-00412-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVE Colorectal cancer (CRC), as a highly prevalent malignant tumor globally, faces the dual challenges of drug resistance of cancer stem cells and immune escape in its treatment. Although the traditional Chinese medicine Yigong San (YGS) shows potential in improving the clinical adverse reactions of CRC, its core active components and mechanism of action remain unclear. Based on network pharmacology screening, this study for the first time discovered that Gomisin B might regulate the progression of CRC through the Toll-like receptor 4/Nuclear Factor-kappa B (TLR4/NF-κB) signaling pathway, and aimed to systematically reveal the molecular mechanisms by which YGS and Gomisin B inhibited the malignant phenotypes and immune escape of CRC cells. METHODS The The Cancer Genome Atlas (TCGA) database was integrated with network pharmacology analysis to screen for the key target of CRC, Gomisin B, and its associated TLR4/NF-κB pathway. Through in vitro CRC stem cell models and mouse xenograft tumor models, techniques such as CCK-8, Transwell, flow cytometry, qPCR/WB were used to evaluate the effects of YGS and Gomisin B on proliferation, migration, invasion, apoptosis, and epithelial-mesenchymal transition (EMT), and to detect the expression of TLR4 and downstream inflammatory factors. RESULTS Both YGS and Gomisin B inhibited the proliferation, migration and invasion of CRC stem cells and tumor tissues. Meanwhile, they promoted apoptosis but reduced the expression of the inflammatory factor TLR4 and proteins associated with the NF-κB pathway, thereby exerting suppressive effects on tumorigenesis and disease progression. YGS might also impede EMT progression through modulation of the NF-κB pathway. CONCLUSION This study for the first time elucidated the dual anti-tumor mechanisms of YGS, which sensitized CRC stem cells and inhibited immune escape by targeting the TLR4/NF-κB pathway through Gomisin B. It provides a pharmacological basis for the modern research of traditional Chinese medicine compound against CRC. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Peng Shen
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Shunli Wu
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Yi Chen
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Guangjing Feng
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Xue Guo
- Infection Control Department, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Yingguo Chen
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Zhigang Wang
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Youfeng Shen
- Department of Laboratory Medicine, Chongqing Precision Medical Industry Technology Research Institute, Chongqing, China
| | - Hongbo Wang
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China.
| | - Ke Li
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China.
| |
Collapse
|
34
|
Zhao Z, He J, Qiu S, Wang L, Huangfu S, Hu Y, Wu Q, Yang Y, Li X, Huang M, Liu S, Guan H, Chen Z, Zhang X, Zhang Y, Ding H, Zhao X, Xiao G, Pan Y, Liu T, Wu Y, Pan J. Targeting PLK1-CBX8-GPX4 axis overcomes BRAF/EGFR inhibitor resistance in BRAFV600E colorectal cancer via ferroptosis. Nat Commun 2025; 16:3605. [PMID: 40240371 PMCID: PMC12003730 DOI: 10.1038/s41467-025-58992-z] [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: 05/01/2024] [Accepted: 04/09/2025] [Indexed: 04/18/2025] Open
Abstract
Metastatic BRAFV600E colorectal cancer (CRC) confers poor prognosis and represents a therapeutic bottleneck. To identify resistance mechanisms of the mitogen-activated protein kinase (MAPK) pathway in BRAFV600E CRC, we perform genome-wide CRISPR-Cas9 screening and discover that targeting glutathione peroxidase 4 (GPX4) overcomes resistance to BRAF inhibitor (BRAFi) combined with or without epidermal growth factor receptor inhibitor (EGFRi) in BRAFV600E CRC. Specifically, BRAFi ± EGFRi upregulates GPX4 expression, which antagonizes therapy-induced ferroptosis. Moreover, polo-like kinase 1 (PLK1) substrate activation promotes PLK1 translocation to the nucleus, activating chromobox protein homolog 8 (CBX8) phosphorylation at Ser265 to drives GPX4 expression. Targeting PLK1 enhances BRAFi ± EGFRi inhibition and triggers ferroptosis in vitro, vivo, organoid, and patient-derived xenograft model. Collectively, we demonstrate a PLK1-CBX8-GPX4 signaling axis that relays the ferroptosis mechanism of therapeutic resistance and propose a clinically actionable strategy to overcome BRAFi ± EGFRi resistance in BRAFV600E CRC.
Collapse
Affiliation(s)
- Zhan Zhao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Jiashuai He
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Shenghui Qiu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou, 510632, P. R. China
| | - Lu Wang
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou, Guangdong, 510632, P. R. China
| | - Shuchen Huangfu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Yangzhi Hu
- The Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, P.R. China
| | - Qing Wu
- Department of Hepatic-biliary-pancreatic Surgery, The Second People's Hospital of Foshan, Foshan, Guangdong, 528000, P. R. China
| | - Yabing Yang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Xiaobo Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Maohua Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Shijin Liu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Hanyang Guan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Zuyang Chen
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Xiangwei Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Yiran Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Hui Ding
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Xiaoxu Zhao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Guandi Xiao
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, P. R. China
| | - Yunlong Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China
| | - Tongzheng Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou, 510632, P. R. China.
| | - Yanping Wu
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, P. R. China.
| | - Jinghua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, P. R. China.
| |
Collapse
|
35
|
Sandhu S, Blandon C, Kumar S. Evaluating Risk Factors for Early-Onset Colorectal Cancer in a Large, Prospective Cohort. Dig Dis Sci 2025:10.1007/s10620-025-09055-2. [PMID: 40234296 DOI: 10.1007/s10620-025-09055-2] [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: 12/11/2024] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Despite the worrisome rise of early-onset colorectal cancer (EOCRC), risk factors have not been definitively established. We use a large, granular database to evaluate risk factors for EOCRC, investigate differences in associations with EOCRC and later-onset CRC (LOCRC), and compare metrics of accelerated aging, a hypothesized driver of EOCRC. METHODS This was a case-control analysis within the UK Biobank. Risk factors for each cancer were identified and compared, including aging measures (chronological age, telomere length, PhenoAge, and homeostatic dysregulation). RESULTS A total of 31,164 persons were matched. We found an increased risk of EOCRC with PRS (OR 1.53; 95% CI 1.19-1.97; p < 0.001). LOCRC was associated with increasing PRS (OR 1.48; 95% CI 1.44 - 1.53; p < 0.001), increasing waist-to-hip ratio (OR 5.81; 95% CI 3.25 - 10.38; p < 0.001), family history of CRC (OR 1.27; 95% CI 1.16 - 1.40; p < 0.001), and history of smoking (OR 1.11; 95% CI 1.03 - 1.19; p = 0.01). Male sex and prior CRC screening were associated with reduced risk of LOCRC. The inclusion of PhenoAge as the measure of aging demonstrated the best model fit for both EOCRC and LOCRC. For each year that PhenoAge exceeded chronological age, the odds of EOCRC increased by 7%, while odds of LOCRC only increased by 1%. CONCLUSIONS Within this study, we find that genetic risk variants are a significant driver of EOCRC risk. Accelerated aging appears to be associated with increased risk of both EOCRC and LOCRC, and measures such as PhenoAge warrant continued study.
Collapse
Affiliation(s)
- Sunny Sandhu
- Division of Digestive Health and Liver Diseases, Department of Medicine, Miller School of Medicine at the University of Miami, 1120 NW 14th St, Locator Code C-240, Miami, FL, 33136, USA
| | - Catherine Blandon
- Division of Digestive Health and Liver Diseases, Department of Medicine, Miller School of Medicine at the University of Miami, 1120 NW 14th St, Locator Code C-240, Miami, FL, 33136, USA
| | - Shria Kumar
- Division of Digestive Health and Liver Diseases, Department of Medicine, Miller School of Medicine at the University of Miami, 1120 NW 14th St, Locator Code C-240, Miami, FL, 33136, USA.
- Sylvester Comprehensive Cancer Center, Miller School of Medicine at the University of Miami, 1120 NW 14th St, Locator Code C-240, Miami, FL, 33136, USA.
| |
Collapse
|
36
|
Sharma A, Zalejski J, Bendre SV, Kavrokova S, Hasdemir HS, Ozgulbas DG, Sun J, Pathmasiri KC, Shi R, Aloulou A, Berkley K, Delisle CF, Wang Y, Weisser E, Buweneka P, Pierre-Jacques D, Mukherjee S, Abbasi DA, Lee D, Wang B, Gevorgyan V, Cologna SM, Tajkhorshid E, Nelson ER, Cho W. Cholesterol-targeting Wnt-β-catenin signaling inhibitors for colorectal cancer. Nat Chem Biol 2025:10.1038/s41589-025-01870-y. [PMID: 40240631 DOI: 10.1038/s41589-025-01870-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/28/2025] [Indexed: 04/18/2025]
Abstract
Most persons with colorectal cancer (CRC) carry adenomatous polyposis coli (APC) truncation leading to aberrant Wnt-β-catenin signaling; however, effective targeted therapy for them is lacking as the mechanism by which APC truncation drives CRC remains elusive. Here, we report that the cholesterol level in the inner leaflet of the plasma membrane (IPM) is elevated in all tested APC-truncated CRC cells, driving Wnt-independent formation of Wnt signalosomes through Dishevelled (Dvl)-cholesterol interaction. Cholesterol-Dvl interaction inhibitors potently blocked β-catenin signaling in APC-truncated CRC cells and suppressed their viability. Because of low IPM cholesterol level and low Dvl expression and dependence, normal cells including primary colon epithelial cells were not sensitive to these inhibitors. In vivo testing with a xenograft mouse model showed that our inhibitors effectively suppressed truncated APC-driven tumors without causing intestinal toxicity. Collectively, these results suggest that the most common type of CRC could be effectively and safely treated by blocking the cholesterol-Dvl-β-catenin signaling axis.
Collapse
Affiliation(s)
- Ashutosh Sharma
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Julian Zalejski
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Shruti Vijay Bendre
- Department of Molecular and Integrative Physiology, Cancer Center at Illinois, Beckman Institute for Advanced Science and Technology, Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Simona Kavrokova
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Hale Siir Hasdemir
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Defne Gorgun Ozgulbas
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jiachen Sun
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | | | - Ruicheng Shi
- Department of Comparative Biosciences, Division of Nutritional Sciences, Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Ahmed Aloulou
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Kyli Berkley
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Charles F Delisle
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Young Wang
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Erin Weisser
- Department of Molecular and Integrative Physiology, Cancer Center at Illinois, Beckman Institute for Advanced Science and Technology, Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Pawanthi Buweneka
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | | | - Sayandeb Mukherjee
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Diana A Abbasi
- Department of Neurogenetics and Translational Neuroscience, Rush University, Chicago, IL, USA
| | - Daesung Lee
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Bo Wang
- Department of Comparative Biosciences, Division of Nutritional Sciences, Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | | | - Emad Tajkhorshid
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, Cancer Center at Illinois, Beckman Institute for Advanced Science and Technology, Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Wonhwa Cho
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
37
|
Yang Q, Zhang H, Luo J, Yu H, Yang X, Wang C. FADS2 inhibits colorectal cancer cell proliferation by regulating ferroptosis through SLC7A11/GPX4. Mol Biol Rep 2025; 52:394. [PMID: 40232565 DOI: 10.1007/s11033-025-10395-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/26/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading factor in cancer mortality globally. Ferroptosis, a regulated cell death described via lipid peroxidation, is crucial in cancer biology. This study explores the link between ferroptosis, FADS2, and CRC, focusing on the prognostic significance and therapeutic potential of targeting FADS2. METHODS The differential expression analysis of the Cancer Genome Atlas-colon adenocarcinoma (TCGA-COAD) and GSE36400 datasets was conducted to determine key ferroptosis-related genes, followed by functional enrichment analysis. Prognosis-related genes were assessed utilizing Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression. Genetic variation analysis and immune analysis were employed to evaluate the clinical significance of FADS2. The impacts of FADS2 knockdown on CRC cell migration, proliferation, invasion, and ferroptosis were evaluated by in vitro cell experiments. RESULTS 64 key ferroptosis-related genes in CRC were highly enriched in pathways such as glutathione metabolism and peroxisome. Eleven prognosis-associated genes were identified, with TP53 showing the highest mutation frequency. High FADS2 expression was linked to poorer prognosis and higher immune cell infiltration. FADS2 knockdown significantly decreased glutathione (GSH) levels, SLC7A11, and GPX4 expression, increased malondialdehyde (MDA) levels, indicating the promotion of ferroptosis. Functional tests revealed knockdown FADS2 repressed CRC cell proliferation, migration, and invasion. SLC7A11 or GPX4 overexpression partially rescued the effects of FADS2 knockdown. Additionally, FADS2 knockdown enhances the chemosensitivity of CRC cells to oxaliplatin. CONCLUSION FADS2 is essential for encouraging CRC cell proliferation and tumor growth by preventing ferroptosis. Targeting FADS2 may enhance ferroptosis and suppress CRC progression, offering a possible course of treatment for CRC patients. The knockdown of FADS2 enhances the chemosensitivity of CRC cells to oxaliplatin, providing valuable insights for future clinical applications.
Collapse
Affiliation(s)
- Qinghui Yang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
| | - Hao Zhang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
| | - Jing Luo
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
| | - Hongmei Yu
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
| | - Xiaodi Yang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China.
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China.
| | - Chen Wang
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, No. 170 Xinsong Road, Minhang District, Shanghai, China.
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer(SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China.
| |
Collapse
|
38
|
Shantha Kumara HMC, Addison P, Yan XH, Sharma AR, Mitra N, Angammana HN, Hedjar Y, Chen YR, Cekic V, Richard WL. Plasma extracellular cold inducible RNA-binding protein levels are elevated for 1 month post-colectomy which may promote metastases. World J Gastrointest Oncol 2025; 17:100678. [PMID: 40235912 PMCID: PMC11995324 DOI: 10.4251/wjgo.v17.i4.100678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/10/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Cold-inducible RNA-binding protein (CIRP) is related to a family of stress-induced RNA-binding proteins. It is primarily found in the nucleus, where it regulates transcription. Under stress, CIRP translocates to the cytoplasm where it modulates translation; a subset is secreted as extracellular CIRP (eCIRP) which is a damage-associated molecular pattern (DAMP) molecule that stimulates the production of inflammatory mediators. Elevated blood eCIRP levels may foster immune tolerance and facilitate tumor growth. Increased CIRP levels have been noted in various malignancies including colorectal cancer (CRC). This study's objective was to determine plasma eCIRP levels before and after minimally invasive colorectal resection (MICR) for CRC. AIM To assess plasma eCIRP levels prior to and following minimally invasive colorectal resection in the context of cancer pathology. METHODS MICR patients from an IRB-approved data/tissue bank for whom plasma samples were available were eligible. Plasma specimens were obtained preoperatively (preop) and at least 3 time's postop [between postoperative day (POD) 1-41]; late samples were grouped into 7-day blocks and were considered separate time points. eCIRP levels were assessed via enzyme-linked immunosorbent assay (pg/mL) and results presented as mean ± SD, analysis with Wilcoxon paired t-test). RESULTS A total of 83 CRC patients who underwent MICR [colon 66%, rectal 34%; laparoscopic-assisted (LA), 70%; hand-assisted laparoscopic (HAL), 30%] were studied. The mean preop eCIRP level was 896.8 ± 757.0 pg/mL. Elevations in mean plasma levels (P = < 0.001) were noted on POD1 (2549 ± 2632 pg/mL, n = 83), POD3 (1871 ± 1362 pg/mL, n = 77), POD7-13 (1788 ± 1403 pg/mL, n = 57), POD14-20 (1473 ± 738.8 pg/mL, n = 30), and POD21-27 (1681 ± 1375 pg/mL, n = 21). No significant differences were noted at POD 28-41. Higher values were noted in the HAL's (vs LA) group, however, there were more rectal cancers in the former. CONCLUSION Elevated plasma eCIRP levels persist for a month post MICR for CRC (change from baseline, 77%-184%); highest values seen on POD1. The initial surge may be due to the acute inflammatory response while later elevations may be related to wound healing and remodeling. The higher levels noted in the HAL's group (with greater IL and more rectal cases) suggest the extent of surgical trauma impacts eCIRP levels. Further investigations are needed.
Collapse
Affiliation(s)
- H M C Shantha Kumara
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY 10028, United States
- Northwell, New Hyde Park, New York, NY 10042, United States
| | - Poppy Addison
- Division of Colon and Rectal Surgery, Department of Surgery, Staten Island University Hospital, Northwell Health, Staten Island, NY 10305, United States
| | - Xiao-Hong Yan
- Department of Pathology and Cell Biology, Columbia University Medical Center, Vanderbilt Clinic, New York, NY 10032, United States
| | - Anuj Raj Sharma
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY 10028, United States
- Northwell, New Hyde Park, New York, NY 10042, United States
| | - Neil Mitra
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY 10028, United States
- Northwell, New Hyde Park, New York, NY 10042, United States
| | - Hansani N Angammana
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY 10028, United States
- Northwell, New Hyde Park, New York, NY 10042, United States
| | - Yanni Hedjar
- Department of Surgery, Brookdale Hospital and Medical Center, Brooklyn, NY 11212, United States
| | - Yi-Ru Chen
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY 10028, United States
- Northwell, New Hyde Park, New York, NY 10042, United States
| | - Vesna Cekic
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY 10028, United States
- Northwell, New Hyde Park, New York, NY 10042, United States
| | - Whelan L Richard
- Division of Colon and Rectal Surgery, Department of Surgery, Lenox Hill Hospital, Northwell Health, New York, NY 10028, United States
- Northwell, New Hyde Park, New York, NY 10042, United States
- Donald and Barbara Zucker School of Medicine, Hofstra/Northwell 500 Hofstra Blvd, Hempstead, NY 11549, United States
| |
Collapse
|
39
|
Li R, Sun X, Yu Z, Li P, Zhao X. Identification of predictors for lymph node metastasis in T2 colorectal cancer: retrospective cohort study from a high-volume hospital. BMC Cancer 2025; 25:700. [PMID: 40234815 PMCID: PMC12001727 DOI: 10.1186/s12885-025-14104-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the most prevalent malignant tumor of the digestive system globally, ranking third in incidence and second in mortality. In previous studies, the rate of lymph node metastasis (LNM) in T2 CRC ranged from 18.0 to 28.0%. We aim to identify T2 CRC patients without LNM and thereby mitigate the complications and potential impact on the quality of life associated with surgery. METHODS In this retrospective study, 787 cases with T2 CRC were selected. The preoperative and postoperative clinicopathological features were retrospectively studied. Univariate analysis and multivariate analysis were performed using binary logistic regression to determine the predictive factor for LNM. Odds ratio (OR) and 95% confidence interval (CI) were conducted. RESULTS 184 (23.4%) patients were diagnosed with LNM, including 144 (78.3%) patients with N1stage and 40 (21.7%) patients with N2 stage. According to univariate analysis and multivariate analysis, poorly differentiated tumors (p = 0.003, OR = 4.405, 95%CI: 1.632-11.893), perineural invasion (p = 0.001, OR = 4.789, 95%CI: 1.958-11.716), and lymphovascular invasion (p = 0.001, OR = 2.779, 95%CI: 1.497-5.159) were independent risk factors of LNM, while male (p = 0.017, OR = 0.652, 95%CI: 0.459-0.926) and elevated preoperative PLR (p = 0.048, OR = 0.996, 95%CI: 0.993-1.000) seemed to be independent protective factors. Larger tumor size did not show significant association with LNM. CONCLUSIONS Approximately three-quarters of T2 CRC patients are likely to avoid unnecessary surgery. Female, poorly differentiated tumors, perineural invasion, and lymphovascular invasion are expected to be used as predictors of LNM in T2 CRC.
Collapse
Affiliation(s)
- Rui Li
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xu Sun
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhiyuan Yu
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, China
| | - Peiyu Li
- Medical School of Chinese PLA, Beijing, China.
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China.
- School of Medicine, Nankai University, Tianjin, China.
| | - Xudong Zhao
- Medical School of Chinese PLA, Beijing, China.
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China.
| |
Collapse
|
40
|
Qi J, Jiang T, Liu B, Hu Q, Chen J, Ma N, Xu Y, Song H, Song J. LINC02167 stabilizes KSR1 mRNA in an m 5C-dependent manner to regulate the ERK/MAPK signaling pathway and promotes colorectal cancer metastasis. J Exp Clin Cancer Res 2025; 44:121. [PMID: 40234937 PMCID: PMC11998267 DOI: 10.1186/s13046-025-03368-w] [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: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Metastasis is a leading cause of colorectal cancer (CRC)-related mortality, yet its molecular mechanisms remain poorly understood. Long noncoding RNAs (lncRNAs) have emerged as critical regulators of CRC metastasis, but their specific roles are not fully elucidated. This study identifies and characterizes a novel lncRNA LINC02167 as a critical regulator of CRC metastasis. METHODS LINC02167 expression was analyzed in CRC tissues via real-time quantitative polymerase chain reaction and fluorescence in situ hybridization. Functional assays evaluated its role in CRC cell migration, invasion, and metastasis in vitro and in vivo. Mechanistic exploration involves a combination of techniques, including RNA sequencing, mass spectrometry, RNA pull-down, RNA immunoprecipitation, chromatin immunoprecipitation, luciferase reporter assays, RNA stability assays, and bioinformatics analysis, to uncover the molecular interactions and pathways regulated by LINC02167. RESULTS LINC02167 is markedly upregulated in CRC tissues and strongly correlates with advanced clinical features and poor prognosis. Functional analyses reveal that LINC02167 enhances CRC cell migration and invasion in vitro and promotes metastasis in vivo. Mechanistically, LINC02167 serves as a molecular scaffold, forming a complex with YBX1 and ILF3 to facilitate YBX1 binding to NSUN2-mediated m5C modification sites on KSR1 mRNA, thereby stabilizing KSR1 mRNA and activating the ERK/MAPK signaling pathway to drive CRC metastasis. Additionally, MYC-driven transcriptional activation leads to the upregulation of LINC02167 in CRC. CONCLUSIONS This study uncovers a novel mechanism through which LINC02167 promotes the ERK/MAPK pathway and CRC metastasis via m5C modification, underscoring its potential as a promising therapeutic target for metastatic CRC treatment.
Collapse
Affiliation(s)
- Junwen Qi
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Bowen Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Qihang Hu
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Junnan Chen
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Ning Ma
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Hu Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| | - Jun Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
41
|
Junwei W, Xin C, Limei G, Fei L, Siyi L, Yao M, Lin H, Xiangchao S, Wei F, Xin Z. Mesenteric benign lymph node enlargement in colorectal cancer: Friend or foe? Transl Oncol 2025; 56:102368. [PMID: 40233503 DOI: 10.1016/j.tranon.2025.102368] [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: 09/24/2024] [Revised: 03/04/2025] [Accepted: 03/16/2025] [Indexed: 04/17/2025] Open
Abstract
INTRODUCTION Benign lymph node enlargement (BLNE) is common in colorectal cancer; however, few studies have investigated its influence on prognosis, clinicopathological features, and pathogenesis. METHODS A cohort study was conducted to analyze the clinicopathologic features and prognosis of colorectal cancer patients, categorized based on the presence or absence of BLNE. Given the correlation between lymph nodes and immune response, immunohistochemistry, transcriptome analysis, and exon sequencing were employed to further investigate the differences in the immune microenvironment of primary tumors. RESULTS Overall, 630 AJCC stage I/II patients were included in the study, with 131 in the BLNE group and 499 in the Non-BLNE (NBLNE) group. Patients in the BLNE group were found to have a significantly better disease-free survival (DFS) (hazard ratio [HR] 0.44, P = 0.016) and overall survival (OS) (HR 0.46, P = 0.011) than those in the NBLNE group. Pathologically, compared with the NBLNE group, the BLNE group had more mature tertiary lymphoid structures (66.7 % vs. 36.5 %, P = 0.002) and higher immunoscores (18.8 % vs. 2.1 %, P = 0.004) in primary tumor tissue. Also, transcriptome analysis showed that, compared with NBLNE, the genes upregulated in BLNE were enriched in immune-related pathways, such as adaptive immune response and immuno-regulatory interactions. Whole-exon sequencing analysis revealed a higher tumor mutation burden (TMB) in the BLNE group [6.03 (5.59, 7.59) vs. 5.33 (4.62, 6.34), P = 0.025]. CONCLUSION BLNE is positively associated with the prognosis of colorectal cancer, possibly because patients with BLNE have a stronger anti-tumor immune response.
Collapse
Affiliation(s)
- Wang Junwei
- Department of General Surgery, Peking University Third Hospital, PR China; Peking university third hospital cancer center, PR China; Beijing Key Laboratory of Collaborative Innovation in Gastrointestinal Oncology, PR China
| | - Chen Xin
- Department of General Surgery, Peking University Third Hospital, PR China; Peking university third hospital cancer center, PR China
| | - Guo Limei
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, PR China
| | - Li Fei
- Department of General Surgery, Peking University Third Hospital, PR China; Peking university third hospital cancer center, PR China
| | - Lu Siyi
- Department of General Surgery, Peking University Third Hospital, PR China; Peking university third hospital cancer center, PR China
| | - Ma Yao
- Department of General Surgery, Peking University Third Hospital, PR China; Peking university third hospital cancer center, PR China
| | - Hsinyi Lin
- Department of General Surgery, Peking University Third Hospital, PR China; Peking university third hospital cancer center, PR China
| | - Shi Xiangchao
- Department of General Surgery, Peking University Third Hospital, PR China; Peking university third hospital cancer center, PR China.
| | - Fu Wei
- Department of General Surgery, Peking University Third Hospital, PR China; Peking university third hospital cancer center, PR China.
| | - Zhou Xin
- Department of General Surgery, Peking University Third Hospital, PR China; Peking university third hospital cancer center, PR China; Beijing Key Laboratory of Collaborative Innovation in Gastrointestinal Oncology, PR China.
| |
Collapse
|
42
|
An L, Chen P, He M, Li H, Zhou Y, Zhao H, Geng C. RBM10 suppresses colorectal cancer invasion by regulating LncRNA SNHG17 alternative splicing. Sci Rep 2025; 15:12761. [PMID: 40229389 PMCID: PMC11997062 DOI: 10.1038/s41598-025-97869-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 04/08/2025] [Indexed: 04/16/2025] Open
Abstract
To explore the role of RBM10 in colorectal cancer (CRC) and the regulatory mechanism of CRC invasion through alternative splicing (AS) of long non-coding RNA (lncRNA) SNHG17 by RBM10. Samples were collected from sixty cases of CRC and their corresponding adjacent normal tissues. Immunohistochemistry and Western blot were performed to analyze the expression of RBM10. A Transwell invasion assay was conducted to evaluate the effect of RBM10 on the invasion of HCT116 cells, and a Western blot was performed to detect the expression of EMT-related proteins. Moreover, CLIP-seq and RIP experiments were performed to explore the interaction between RBM10 and SNHG17. The expression of RBM10 was significantly decreased in CRC tissues and cells compared to the normal adjacent tissues. Overexpression of RBM10 inhibited CRC invasion, while knockdown of RBM10 had the opposite effect. RBM10 was found to interact with SNHG17 and regulate its splice isoform balance. Specifically, the splice variant SNHG17_2 regulated by RBM10 was upregulated in CRC and was positively correlated with CRC invasion. RBM10 inhibits CRC invasion by regulating the alternative splicing of SNHG17, providing new research directions and potential targets for CRC treatment.
Collapse
Affiliation(s)
- Linxia An
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, 150000, China
| | - Peng Chen
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, 150000, China
- Department of Oncological SurgeryChifeng Clinical Medical College, Inner Mongolia Medical University, Chifeng, 024000, China
| | - Miao He
- Department of Pathology, Daqing Oilfield General Hospital, Daqing, 163000, China
| | - Huifeng Li
- Department of Pathology, Daqing Oilfield General Hospital, Daqing, 163000, China
| | - Yingshu Zhou
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163515, China
| | - Hongwei Zhao
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163515, China
| | - Changhui Geng
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163515, China.
| |
Collapse
|
43
|
Abken H. CAR T cell therapies in gastrointestinal cancers: current clinical trials and strategies to overcome challenges. Nat Rev Gastroenterol Hepatol 2025:10.1038/s41575-025-01062-y. [PMID: 40229574 DOI: 10.1038/s41575-025-01062-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/16/2025]
Abstract
Despite multimodal treatment options, most gastrointestinal cancers are still associated with high mortality rates and poor responsiveness to immunotherapy. The unprecedented efficacy of chimeric antigen receptor (CAR)-engineered T cells in the treatment of haematological malignancies raised interest in translating CAR T cell therapies to the treatment of gastrointestinal cancers. Treatment of solid cancers with canonical CAR T cells faces substantial challenges, including the dense architecture of the tumour tissue, the tolerogenic environment with low tumour-intrinsic immunogenicity, the rareness of targetable tumour-selective antigens, the antigenic heterogeneity of cancer cells, and the profound metabolic and immune cell disbalances. This Review provides an overview of CAR T cell trials in the treatment of gastrointestinal cancers, discussing considerations relating to safety, efficacy, potential reasons for failure and options for improving CAR T cells for the future. In addition, lessons regarding how to improve efficacy are drawn from CAR T cells armed with adjuvants that sustain their activation within the hostile environment and activate resident immune cells. As the field is rapidly evolving, current treatment modalities and editing CAR T cell functionalities are being refined towards a potentially more successful CAR T cell therapy for gastrointestinal cancers.
Collapse
Affiliation(s)
- Hinrich Abken
- Leibniz Institute for Immunotherapy, Genetic Immunotherapy Division, Regensburg, Germany.
- Genetic Immunotherapy, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
44
|
Li C, Wei Y, Lei J. Quantitative cancer-immunity cycle modeling for predicting disease progression in advanced metastatic colorectal cancer. NPJ Syst Biol Appl 2025; 11:33. [PMID: 40221414 PMCID: PMC11993626 DOI: 10.1038/s41540-025-00513-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Patients with advanced metastatic colorectal cancer (mCRC) typically exhibit significant interindividual differences in treatment responses and face poor survival outcomes. To systematically analyze the heterogeneous tumor progression and recurrence observed in advanced mCRC patients, we developed a quantitative cancer-immunity cycle (QCIC) model. The QCIC model employs differential equations to capture the biological mechanisms underlying the cancer-immunity cycle and predicts tumor evolution dynamics under various treatment strategies through stochastic computational methods. We introduce the treatment response index (TRI) to quantify disease progression in virtual clinical trials and the death probability function (DPF) to estimate overall survival. Additionally, we investigate the impact of predictive biomarkers on survival prognosis in advanced mCRC patients, identifying tumor-infiltrating CD8+ cytotoxic T lymphocytes (CTLs) as key predictors of disease progression and the tumor-infiltrating CD4+ Th1/Treg ratio as a significant determinant of survival outcomes. This study presents an approach that bridges the gap between diverse clinical data sources and the generation of virtual patient cohorts, providing valuable insights into interindividual treatment variability and survival forecasting in mCRC patients.
Collapse
Affiliation(s)
- Chenghang Li
- School of Mathematical Sciences, Tiangong University, Tianjin, 300387, China
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China.
| | - Jinzhi Lei
- School of Mathematical Sciences, Tiangong University, Tianjin, 300387, China.
- Center for Applied Mathematics, Tiangong University, Tianjin, 300387, China.
| |
Collapse
|
45
|
Zhao M, Jiang Y, Shao T, Tang W. Safety, efficacy, and cost-effectiveness evaluation of systemic treatments for refractory colorectal cancer: a systematic review and modeling study. HEALTH ECONOMICS REVIEW 2025; 15:33. [PMID: 40214895 PMCID: PMC11987259 DOI: 10.1186/s13561-025-00622-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/24/2025] [Indexed: 04/14/2025]
Abstract
OBJECTIVES To conduct pooled estimates and comparative evaluations of safety and efficacy, alongside cost-effectiveness and value-based pricing analyses, for systemic treatments recommended by the National Comprehensive Cancer Network in refractory colorectal cancer. METHODS A comprehensive search for related randomized controlled trials was conducted on PubMed, EMBASE, the Cochrane Library, and ClinicalTrials.gov. Safety was evaluated by aggregating treatment-related adverse events (TRAEs) and performing Bayesian network meta-analysis (NMA) for indirect comparisons. Pooled survival estimates of overall survival (OS) and progression-free survival (PFS) were conducted to assess treatment efficacy. For NMA of OS and PFS, time-variant fractional polynomial models were employed as the primary analysis, with Cox proportional hazards models used for result validation. Economic evaluations were performed using partitioned survival models from the US public sector perspective. Clinical parameters were sourced from meta-analyses; cost parameters included drug treatment, follow-up and administration, end-of-life care, and adverse event management expenses, which were obtained from the Federal Supply Schedule, public databases or published literature. Utility values were sourced from the CORRECT trial. Price simulations were also conducted. Robustness of results was confirmed by sensitivity and scenario analyses RESULTS: We included nine studies comprising 3,978 patients and incorporating six treatments recommended by NCCN, including best supportive care (BSC), regorafenib, regorafenib dose optimization (REDo), trifluridine/tipiracil (TAS-102), TAS-102 with bevacizumab (TAS-BEV), and fruquintinib. Targeted treatments increased serious TRAEs and grade 3 + TRAEs compared to BSC. However, no significant safety differences were found among the targeted therapies. Regarding efficacy, REDo led in median OS, while fruquintinib led in median PFS. NMA indicated that TAS-BEV had the greatest PFS and OS survival benefit, followed by fruquintinib and REDo. Cost-effectiveness analysis favored BSC as the least expensive and the most cost-effective profile. TAS-BEV had the greatest effectiveness, with TAS-102 being the most cost-effective among targeted therapies. For cost-effectiveness against BSC, the price reductions of TAS-102, fruquintinib, REDoS, regorafenib, and TAS-BEV were 39%, 24%, 14%, 8%, and 7%, respectively. CONCLUSIONS Targeted therapies have comparable safety; TAS-BEV is highly effective, TAS-102 is the top cost-effective targeted therapy. Treatment choice should balance individual patient needs with safety, efficacy, and cost.
Collapse
Affiliation(s)
- Mingye Zhao
- Department of Pharmacoeconomics, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
- Center for Pharmacoeconomics and Outcomes Research, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yunlin Jiang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Taihang Shao
- School of Public Health, Faculty of Medicine, The Chinese University of Hong Kong, Ma Liu Shui, China
| | - Wenxi Tang
- Department of Pharmacoeconomics, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China.
- Center for Pharmacoeconomics and Outcomes Research, China Pharmaceutical University, Nanjing, Jiangsu, China.
- Center for Pharmacoeconomics and Outcomes Research, Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
46
|
Li YX, Mu BX, Zhou HJ, Qian J, Zhou JY, Chen M. Development and validation of nomograms for predicting overall survival and cancer-specific survival in unresected colorectal cancer patients undergoing chemotherapy. Sci Rep 2025; 15:12477. [PMID: 40216848 PMCID: PMC11992110 DOI: 10.1038/s41598-025-96526-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
This study aims to develop nomograms for predicting overall survival (OS) and cancer-specific survival (CSS) in colorectal cancer (CRC) patients who did not receive primary site surgery but underwent chemotherapy. We analyzed data from 3,050 patients treated with chemotherapy without primary site surgery from 2010 to 2015, sourced from the Surveillance, Epidemiology, and End Results (SEER) database. The data were randomly divided into training and validation sets. Initial variable selection was performed using the least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analysis was used to identify independent prognostic factors. Two nomograms were subsequently constructed based on these factors. Survival analysis was conducted using Kaplan-Meier plots and the log-rank test. We identified nine significant predictors of OS and CSS: age, marital status, primary site, grade, histology, T stage, M stage, tumor size, and CEA levels. The models for OS and CSS exhibited excellent predictability, with time-dependent area under the receiver operating characteristic curves (AUCs) exceeding 0.7. Calibration curves confirmed the accuracy of these predictions in the training and validation sets. Additionally, decision curve analysis (DCA) indicated that our models provide greater clinical benefit than traditional TNM staging. Notably, survival outcomes varied significantly across risk categories, affirming the models' effective discrimination. For CRC patients who did not receive primary site surgery but underwent chemotherapy, this validated nomogram enables precision prognostication fundamentally shifting the paradigm from population-level TNM estimates to individualized risk-adaptive management.
Collapse
Affiliation(s)
- Yuan-Xiang Li
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Bai-Xiang Mu
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Hua-Jian Zhou
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jun Qian
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jin-Yong Zhou
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| | - Min Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
47
|
Shi S, Jiang T, Liu H, Wu Y, Singh A, Wang Y, Xie J, Li X. Habitat Radiomics Based on MRI for Predicting Metachronous Liver Metastasis in Locally Advanced Rectal Cancer: a Two‑center Study. Acad Radiol 2025:S1076-6332(25)00190-4. [PMID: 40204586 DOI: 10.1016/j.acra.2025.02.046] [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: 01/30/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 04/11/2025]
Abstract
RATIONALE AND OBJECTIVES This study aimed to explore the feasibility of using habitat radiomics based on magnetic resonance imaging (MRI) to predict metachronous liver metastasis (MLM) in locally advanced rectal cancer (LARC) patients. A nomogram was developed by integrating multiple factors to enhance predictive accuracy. METHODS Retrospective data from 385 LARC patients across two centers were gathered. The data from Center 1 were split into a training set of 203 patients and an internal validation set of 87 patients, while Center 2 provided an external test set of 95 patients. K - means clustering was used on T2 - weighted images, and the region of interest was extended at different thicknesses. After feature extraction and selection, four machine - learning algorithms were utilized to build radiomics models. A nomogram was created by combining habitat radiomics, conventional radiomics, and clinical independent predictors. Model performance was evaluated by the AUC, and clinical utility was assessed through calibration curve and DCA. RESULTS Habitat radiomics outperformed other single models in predicting MLM, with AUCs of 0.926, 0.864, and 0.851 in respective sets. The integrated nomogram achieved even higher AUCs of 0.959, 0.925, and 0.889. DCA and calibration curve analysis showed its high net benefit and good calibration. CONCLUSION MRI - based habitat radiomics can effectively predict MLM in LARC patients. The integrated nomogram has optimal predictive performance and improves model accuracy significantly.
Collapse
Affiliation(s)
- Shengming Shi
- Department of Magnetic resonance imaging diagnostic, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang, Harbin 150086, China.
| | - Tao Jiang
- Department of Medical Imaging (MRI), The Fifth Affiliated Hospital of Harbin Medical University, No. 241, Daqing Development Zone Construction Road, Daqing 163316, China.
| | - Han Liu
- Department of Magnetic resonance imaging diagnostic, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang, Harbin 150086, China.
| | - Yupeng Wu
- Department of Magnetic resonance imaging diagnostic, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang, Harbin 150086, China.
| | - Apekshya Singh
- Department of Magnetic resonance imaging diagnostic, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang, Harbin 150086, China.
| | - Yuhang Wang
- Department of Magnetic resonance imaging diagnostic, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang, Harbin 150086, China.
| | - Jiayi Xie
- Department of Magnetic resonance imaging diagnostic, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang, Harbin 150086, China.
| | - Xiaofu Li
- Department of Magnetic resonance imaging diagnostic, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang, Harbin 150086, China.
| |
Collapse
|
48
|
Huang Y, Cheng N, Zhi Y, Qiao D, Wang Y, Ma M, Ge C, Tao W, Liu W. Phillyrin from Forsythia suspensa suppresses the proliferation, angiogenesis, and metastasis of colorectal cancer via targeting CD147. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119759. [PMID: 40210179 DOI: 10.1016/j.jep.2025.119759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/26/2025] [Accepted: 04/05/2025] [Indexed: 04/12/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Forsythia suspensa (Thunb.) Vahl, a traditional Chinese herbal medicine, is widely used in clinical practice. Phillyrin (PHN), a major bioactive component of Forsythia suspensa, exhibits significant anti-inflammatory, neuroprotective, and antibacterial properties, offering potential for colorectal cancer (CRC) prevention and treatment. AIM OF THE STUDY This study aimed to clarify the effects of PHN on CRC progression, focusing on epithelial-mesenchymal transition (EMT) and angiogenesis, to elucidate the underlying molecular mechanisms involving CD147. MATERIALS AND METHODS In vitro, cell viability and colony formation were conducted to detect the inhibition of PHN on CRC cells. Wound healing and Transwell assays were used to detect the migration and invasion. PCR, Western blot and ELISA were performed to clarify the relevant molecular levels. Overexpression plasmids were constructed to regulate the target molecule for mechanism research. In vivo, subcutaneous xenograft and lung metastasis models evaluated PHN's anti-cancer effects including histological and immunohistochemical (IHC) analysis. RESULTS PHN inhibited CRC cell proliferation, migration, and invasion in vitro, downregulating CD147 expression, while CD147 overexpression reversed the effects of PHN. In vivo, PHN significantly suppressed tumor growth and lung metastasis, reducing VEGFA, N-Cadherin, Snail1, and MMP-9 expression, and increasing E-Cadherin levels. CONCLUSION These findings indicated that PHN suppressed the proliferation and metastasis of CRC via regulating CD147-mediated EMT and angiogenesis. PHN may be a promising therapeutic agent for CRC treatment in clinic, and CD147 may be a potential target for drug development.
Collapse
Affiliation(s)
- Yuzhen Huang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Nan Cheng
- Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Yingru Zhi
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Dan Qiao
- Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Yan Wang
- Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Mengqing Ma
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Chun Ge
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Weiwei Tao
- Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
| | - Wanli Liu
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
49
|
Chen E, Chen L, Zhang W. Robotic-assisted colorectal surgery in colorectal cancer management: a narrative review of clinical efficacy and multidisciplinary integration. Front Oncol 2025; 15:1502014. [PMID: 40260300 PMCID: PMC12009946 DOI: 10.3389/fonc.2025.1502014] [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: 09/26/2024] [Accepted: 03/20/2025] [Indexed: 04/23/2025] Open
Abstract
Colorectal cancer (CRC) remains a formidable global health challenge, ranking among the most prevalent malignancies and a principal contributor to cancer-associated mortality. While traditional open surgery has historically been the cornerstone of CRC treatment, the advent of minimally invasive techniques, particularly robotic-assisted colorectal surgery (RACS), has garnered significant momentum owing to technological advancements in the field. Robotic platforms, exemplified by the da Vinci Surgical System, offer superior three-dimensional visualization, enhanced dexterity, and heightened precision, yielding improved perioperative outcomes, particularly in anatomically intricate regions such as the pelvis. This review provides a critical appraisal of the current landscape of RACS, emphasizing its superiority over conventional open and laparoscopic approaches. The increased control and precision afforded by robotic surgery have been shown to optimize outcomes in complex procedures such as total mesorectal excision, with evidence indicating reduced intraoperative blood loss, shortened hospital stays, and improved functional recovery. Nonetheless, challenges persist, including absence of haptic feedback, prohibitive costs, and steep learning curve associated with robotic systems. Despite these limitations, RACS has demonstrated considerable promise in sphincter-preserving and function-preserving procedures, ultimately enhancing postoperative quality of life. Beyond the surgical field, this review also investigates the integration of robotic surgery within multidisciplinary treatment strategies for CRC, particularly in the context of locally advanced rectal cancer. The combination of robotic techniques with total neoadjuvant therapy and immunotherapy-especially in tumors characterized by mismatch repair deficiency or high microsatellite instability has shown notable clinical efficacy. Furthermore, emerging personalized therapeutic approaches, including immunotherapies and targeted chemotherapeutic agents, emphasize the transformative potential of RACS in delivering superior oncologic outcomes. Looking towards the future, innovations in robotic platforms, including intraoperative imaging, artificial intelligence, and augmented reality, herald new possibilities for further enhancing the precision and efficacy of colorectal surgeries. The standardization of RACS protocols, alongside ongoing training and robust clinical research, will be critical to fully realizing the benefits of these advancements across diverse clinical settings. By incorporating cutting-edge technologies and personalized treatment methods, robotic-assisted surgery is prepared to become a cornerstone in future of CRC management, with the potential to significantly improve both survival outcomes and patient quality of life.
Collapse
Affiliation(s)
- Engeng Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | | | | |
Collapse
|
50
|
Su A, Lee H, Tran M, Dela Cruz RC, Sathe A, Bai X, Wichmann I, Pflieger L, Moulton B, Barker T, Haslem D, Jones D, Nadauld L, Nguyen Q, Ji HP, Rhodes T. The single-cell spatial landscape of stage III colorectal cancers. NPJ Precis Oncol 2025; 9:101. [PMID: 40189697 PMCID: PMC11973205 DOI: 10.1038/s41698-025-00853-5] [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: 11/13/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
We conducted a spatial analysis of stage III colorectal adenocarcinomas using Hyperion Imaging Mass Cytometry, examining 52 tumors to assess the tumor microenvironment at the single-cell level. This approach identified 10 distinct cell phenotypes in the tumor microenvironment, including stromal and immune cells, with a subset showing a proliferative phenotype. By focusing on spatial neighborhood interactions and tissue niches, particularly regions with tumor-infiltrating lymphocytes, we investigated how cellular organization relates to clinicopathological and molecular features such as microsatellite instability (MSI) and recurrence. We determined that microsatellite stable (MSS) colorectal cancers had an increased risk of recurrence if they had the following features: 1) a low level of stromal tumor-infiltrating lymphocytes, and 2) low interactions between CD4 + T cells and stromal cells. Our results point to the utility of spatial single-cell interaction analysis in defining novel features of the tumor immune microenvironments and providing useful clinical cell-related spatial biomarkers.
Collapse
Affiliation(s)
- Andrew Su
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - HoJoon Lee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Minh Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | | | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xiangqi Bai
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ignacio Wichmann
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Division of Obstetrics and Gynecology, Department of Obstetrics, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | | | - Bryce Moulton
- Intermountain Healthcare, Saint George, UT, 84770, USA
| | - Tyler Barker
- Intermountain Healthcare, Saint George, UT, 84770, USA
| | | | - David Jones
- Intermountain Healthcare, Saint George, UT, 84770, USA
| | | | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | | |
Collapse
|