1
|
Roy S, Deka D, Kondaveeti SB, Ayyadurai P, Siripragada S, Philip N, Pathak S, Duttaroy AK, Banerjee A. An overview of potential of natural compounds to regulate epigenetic modifications in colorectal cancer: a recent update. Epigenetics 2025; 20:2491316. [PMID: 40239010 PMCID: PMC12005453 DOI: 10.1080/15592294.2025.2491316] [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: 12/30/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Colorectal cancer (CRC) remains an alarming global health concern despite advancements in treatment modalities over recent decades. Among the various factors contributing to CRC, this review emphasizes the critical role of epigenetic mechanisms in its pathogenesis and progression. This review also describes the potential role of natural compounds in altering the epigenetic landscape, focused mainly on DNA methylation, histone modification, and non-coding RNAs. Publications from the previous five years were searched and retrieved using well-known search engines and databases like PubMed, Google Scholar, and ScienceDirect. Keywords like CRC/colorectal cancer, CAC/Colitis associated CRC, inflammasomes, epigenetic modulation, genistein, curcumin, quercetin, resveratrol, anthocyanins, sulforaphane, and epigallocatechin-3-gallate were used in various combinations during the search. These natural compounds predominantly affect pathways such as Wnt/β-catenin, NF-κB, and PI3K/AKT to suppress CRC cell proliferation and oxidative stress and enhance anti-inflammation and apoptosis. However, their clinical use is restricted due to their low bioavailability. However, multiple methods exist to overcome challenges like this, including but not limited to structural modifications, nanoparticle encapsulations, bio-enhancers, and novel advanced delivery systems. These methods improve their potential as supportive therapies that target CRC progression epigenetically with fewer side effects. Current research focuses on enhancing epigenetic targeting to control CRC progression while minimizing side effects, emphasizing improved specificity, bioavailability, and efficacy as standalone or synergistic therapies.
Collapse
Affiliation(s)
- Susmita Roy
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Dikshita Deka
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Suresh Babu Kondaveeti
- Department of Biochemistry, Symbiosis Medical College for Women, Symbiosis International (Deemed University), Pune, India
| | - Pavithra Ayyadurai
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Sravani Siripragada
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Neha Philip
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Surajit Pathak
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Antara Banerjee
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| |
Collapse
|
2
|
Kowalski S, Wityk P, Raczak-Gutknecht J, Olszewska A, Żmijewski M, Kocić I. The imidazoline I 2 receptor agonist 2-BFI enhances cytotoxic activity of hydroxychloroquine by modulating oxidative stress, energy-related metabolism and autophagic influx in human colorectal adenocarcinoma cell lines. Eur J Pharmacol 2025; 996:177590. [PMID: 40185322 DOI: 10.1016/j.ejphar.2025.177590] [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: 01/18/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Recently, interest in imidazoline receptors (IRs) has been increasing. Over the years, a growing number of studies have highlighted the therapeutic potential of ligands targeting these receptors, however, the potential role of imidazoline I2 receptor agonists in cancer treatment has not been thoroughly investigated. Colorectal cancer (CRC) is among the most prevalent and lethal forms of cancer worldwide. The complexity of CRC necessitates individualized approaches. One promising area of research within CRC therapy is the regulation of autophagy. Recent studies have explored the relationship between autophagy and cancer progression, revealing that autophagy modulation could be a potential strategy for CRC treatment. However, the mechanisms underlying autophagy regulation remain poorly understood. This study aimed to evaluate the effect of the imidazoline I2 receptor agonist, namely 2-(2-benzofuranyl)-2-imidazoline hydrochloride (2-BFI), on colorectal cancer cells, HT-29 and HCT-116 cell lines, particularly its impact when co-incubated with the autophagy inhibitor, hydroxychloroquine (HCQ). The results showed that 2-BFI synergistically increased the cytotoxic effect of HCQ by inducing oxidative stress and apoptosis. Furthermore, our investigation indicated impairment autophagic influx in colon cancer cells treated by 2-BFI. The comprehensive metabolomic analysis revealed significant alterations in key metabolic pathways including MAO activity, oxidative stress responses, energy-related metabolites and amino acids metabolism. Altogether, these findings demonstrate potential a new therapeutic strategy based on autophagy regulation and the selective induction of oxidative stress in colorectal cancer cells. Moreover, this study provides a foundation for further investigation into the therapeutic potential of imidazoline receptor agonists in cancer therapy.
Collapse
Affiliation(s)
- Szymon Kowalski
- Department of Pharmacology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland.
| | - Paweł Wityk
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland; Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdańsk, Gdansk, Poland; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joanna Raczak-Gutknecht
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdańsk, Gdansk, Poland
| | - Anna Olszewska
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Michał Żmijewski
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Ivan Kocić
- Department of Pharmacology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
3
|
Gholipour F, Entezar M, Amini M, Vandghanooni S, Baradaran B, Eskandani M, Mokhtarzadeh AA. In vitro effects of crocin on the possible anticancer properties of Lactococcus lactis against colorectal adenocarcinoma cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5729-5741. [PMID: 39607550 DOI: 10.1007/s00210-024-03636-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Probiotics have been suggested to contribute to cancer prevention through various mechanisms. Additionally, recent studies have established a connection between diet, microbiota, and overall health. In this respect, the current study aims to understand the impact of crocin on possible anti-cancer and antibacterial effects of Lactococcus lactis (L. lactis) in colorectal cancer cells and pathogenic bacteria. The study involved collecting cell-free supernatants (CFSs) from untreated bacteria as a control group and bacteria treated with crocin, and then examining their ability to prevent the growth of HCT-116 colon cancer cells. It was demonstrated that L. lactis, when treated with crocin, can effectively combat against various types of pathogenic bacteria and can survive in acidic conditions. Both CFS and cro-CFS exhibited a dose-dependent inhibition of HCT-116 cell growth but crocin-treated bacteria showed more significant effects. The half-maximal inhibitory concentration (IC50) for cell growth inhibition was 97.41 µL/mL in CSF group and 72.07 µL/mL in cro-CFS group. The results of flow cytometry tests confirmed the MTT assay findings, showing that cro-CFS group had a significantly higher rate of apoptosis compared to CFS of control group. The results obtained from qPCR also showed that the Caspase 9 and BAX genes were upregulated, and the BCL-2 expression level was reduced in cells treated with cro-CFS compared to the CFS group. Overall, these findings suggest that crocin may alter the composition of CFS from probiotics that are present in the gut, potentially impacting their ability to combat cancer.
Collapse
Affiliation(s)
- Faranak Gholipour
- Department of Biological Science, Faculty of Basic Science, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Mahdi Entezar
- Department of Biological Science, Faculty of Basic Science, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology (RCPN), Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | |
Collapse
|
4
|
Kabeli RG, Boursi B, Zilberberg A, Efroni S. Leveraging machine learning for integrative analysis of T-cell receptor repertoires in colorectal cancer: Insights into MAIT cell dynamics and risk assessment. Transl Oncol 2025; 55:102358. [PMID: 40088748 PMCID: PMC11957502 DOI: 10.1016/j.tranon.2025.102358] [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: 08/23/2024] [Revised: 01/30/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
This study investigates the T-cell receptor (TCR) repertoires in colorectal cancer (CRC) patients by analyzing three distinct datasets: one bulk sequencing dataset of 205 patients with various tumor stages, all newly diagnosed at Sheba Medical Center between 2017 and 2022, with minimal recruitment in 2014 and 2016, and two (public) single-cell sequencing datasets of 10 and 12 patients. Despite the significant variability in the TCR repertoire and the low likelihood of sequence overlap, our analysis reveals an interesting set of TCR sequences across these data. Notably, we observe elevated presence of mucosal-associated invariant T (MAIT) cells in both metastatic and non-metastatic patients. Furthermore, we identify nine identical TCR alpha and TCR beta pairs that appear in both single-cell datasets, with 13 out of 18 sequences from these sequences also appearing in the bulk data. Clinical risk analysis over the bulk dataset, using a subset of these unique sequences, demonstrates a correlation between TCR repertoire disease stage and risk. These findings enhance our understanding of the TCR landscape in CRC and underscore the potential of TCR sequences as biomarkers for disease outcome.
Collapse
Affiliation(s)
- Romi Goldner Kabeli
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Ben Boursi
- Department of Oncology, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel; Faculty of Medical & Health Sciences, Tel-Aviv University, Tel-Aviv, Israel; Center for Clinical Epidemiology and Biostatistics, Perlman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Alona Zilberberg
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Sol Efroni
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
5
|
Nopour R. Optimizing prediction of metastasis among colorectal cancer patients using machine learning technology. BMC Gastroenterol 2025; 25:272. [PMID: 40251500 PMCID: PMC12007332 DOI: 10.1186/s12876-025-03841-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/02/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND AND AIM Colorectal cancer is among the most prevalent and deadliest cancers. Early prediction of metastasis in patients with colorectal cancer is crucial in preventing it from the advanced stages and enhancing the prognosis among these patients. So far, previous studies have been conducted to predict metastasis in colorectal cancer patients using clinical data. The current research attempts to leverage a combination of demographic, lifestyle, nutritional, and clinical factors, such as diagnostic and therapeutical factors, to construct an ML model with more predictive insights and generalizability than previous ones. MATERIALS AND METHODS In this retrospective study, we used 1156 CRC patients referred to the Masoud internal clinic in Tehran City from January 2017 to December 2023. The chosen machine learning algorithms, including LightGBM, XG-Boost, random forest, artificial neural network, support vector machine, decision tree, K-Nearest Neighbor and logistic regression, were utilized to establish prediction models for predicting metastasis among colorectal cancer patients. We also assessed features based on the best-performing model to improve clinical usability. To show the generalizability of the established prediction model for predicting CRC metastasis, we leveraged the data of 115 CRC patients from Imam Khomeini Hospital in Sari City. We assessed the predictive ability of LightGBM as the best-performing model based on external data. RESULTS The LightGBM model with a PPV of 97.32%, NPV of 84.67%, sensitivity of 83.14%, specificity of 93.14%, accuracy of 88.14%, F1-score of 87.51%, and an AU-ROC of 0.9 [Formula: see text]0.01 obtained satisfactory performance for prediction purposes on this topic. Factors including the history of IBD, family history of CRC, number of lymph nodes involved, fruit intake, and tumor size were considered as more strengthful predictors for metastasis in colorectal cancer and clinical usability. The external validation cohort showed a PPV of 0.8, NPV of 0.85, sensitivity of 0.78, specificity of 0.86, accuracy of 0.834, F1-score of 0.795, and AU-ROC of 0.77[Formula: see text]0.03, demonstrating satisfactory generalizability when leveraging external data from other clinical settings. CONCLUSION The current empirical results indicated that LighGBM has predictive competency that can be leveraged by physicians in clinical environments for early prediction of metastasis and enhanced prognosis in patients with colorectal cancer. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Raoof Nopour
- Department of Health Information Management, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Han HJ, Han J, Choi Y, Hwang GJ, Kim S, Ryoo IJ, Kim BY, Soung NK. A novel tubulin inhibitor, No.07, shows anti-cancer and anti-metastatic effects in colon cancer and tumoroids. Life Sci 2025:123644. [PMID: 40252756 DOI: 10.1016/j.lfs.2025.123644] [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: 01/02/2025] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 04/21/2025]
Abstract
Colorectal cancer is a highly metastatic disease and the second leading cause of cancer-related death worldwide. Despite the use of various treatment strategies, including chemotherapy and targeted therapy, challenges such as toxicity, drug resistance, and poor response indicate the critical need for new therapeutic agents. Microtubule target agents are one of the major treatment options for chemotherapy in various cancer patients. However, most of these agents are substrates of the MDR1 protein, which leads to the development of multidrug resistance, significantly limiting their effectiveness. Therefore, the development of new drugs is being actively pursued. In this study, we synthesized a novel compound, No.07, which demonstrates significant anti-cancer activity in 3D spheroid models, patient-derived colon cancer organoid models, and mice xenograft models. No.07 directly binds to tubulin dimers, interfering with microtubule polymerization and thereby disrupting tubulin dynamics, ultimately inducing mitotic arrest. Furthermore, No.07 increases mitochondria reactive oxygen species level, leading to the inactivation of the RAF-MEK-ERK signaling cascade, which consequently inhibits metastasis. Notably, Swiss ADME predictions suggest that No.07 is not a substrate of MDR1 and can cross the blood-brain barrier, unlike other microtubule target agents that are limited by MDR1-mediated drug resistance and poor brain penetration. Additionally, experiments using multidrug-resistant cell lines confirmed that No.07 effectively overcomes multidrug resistance, providing a significant improvement over traditionally used chemotherapy agents. In conclusion, No.07 has the potential to address the limitations of existing treatments as a novel therapeutic option.
Collapse
Affiliation(s)
- Ho Jin Han
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Junyeol Han
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Yerim Choi
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Gwi-Ja Hwang
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Sumin Kim
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - In-Ja Ryoo
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Bo Yeon Kim
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Nak-Kyun Soung
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
7
|
Masdor NA, Hod R, Syed Soffian SS, Mohammed Nawi A. Qualitative insights into ecobiosocial factors influencing colorectal cancer risk in Malaysia. Health Psychol Behav Med 2025; 13:2493143. [PMID: 40256261 PMCID: PMC12006936 DOI: 10.1080/21642850.2025.2493143] [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: 09/12/2024] [Accepted: 04/05/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction Colorectal cancer (CRC) is a growing public health concern in Malaysia influenced by a complex interplay of ecological, biological, and social (EBS) factors. Despite its increasing incidence, limited research has explored how these factors interact to shape CRC risk in the Malaysian context, especially from the perspectives of affected individuals. This study explores Malaysians' perceptions and experiences regarding CRC risk within the EBS framework. Methods A qualitative case study approach involved in-depth interviews with twelve Malaysians aged 35-75 who had undergone colonoscopy at a university hospital. All interviews were recorded and transcribed. Data were collected until saturation was achieved. The transcripts were coded and analysed using ATLAS.ti software. The data were analysed using thematic analysis. Results Findings revealed key themes related to ecological factors in the physical activity environment, which included the sub-themes of type, facilitators, barriers to physical activity, and food sources. The biological factors theme revealed that a family history of CRC influences experience and perception. The subthemes of social factors were sociocultural customs, misconceptions, food preparation methods, CRC-related foods, and food affordability. Conclusion The findings highlighted the multifactorial nature of CRC risk. Understanding the aspects of EBS supports the development of targeted public health interventions to address modifiable CRC risk factors and promote prevention and early CRC detection in the Malaysian context.
Collapse
Affiliation(s)
- Noor Azreen Masdor
- Department of Public Health Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Rozita Hod
- Department of Public Health Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Azmawati Mohammed Nawi
- Department of Public Health Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Szewczyk A, Baczyńska D, Choromańska A, Łapińska Z, Chwiłkowska A, Saczko J, Kulbacka J. Advancing cancer therapy: Mechanisms, efficacy, and limitations of calcium electroporation. Biochim Biophys Acta Rev Cancer 2025; 1880:189319. [PMID: 40222421 DOI: 10.1016/j.bbcan.2025.189319] [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: 11/20/2024] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Calcium electroporation, an innovative technique, uses high-voltage pulses to introduce calcium ions into cells, leading to cell death and tumor growth inhibition. This review explores the potential of calcium electroporation as a promising therapeutic approach in cancer treatment. We provide an in-depth analysis of the underlying mechanisms by which calcium ions function within cells and how their introduction through electroporation can effectively induce cell death in cancer cells. Furthermore, we present a comprehensive overview of the current literature, covering both preclinical and clinical studies, to highlight the safety and efficacy of calcium electroporation in various cancer types, including melanoma, head and neck cancer, and breast cancer. We also discuss the distinct advantages of calcium electroporation over traditional cancer therapies, such as its specific targeting of cancer cells while sparing healthy cells. However, we also address the challenges and limitations associated with this technique, underscoring the need for further research. By providing a comprehensive examination of calcium electroporation, this review aims to contribute to understanding this emerging field and encourage further investigation into its potential as a novel cancer treatment strategy.
Collapse
Affiliation(s)
- Anna Szewczyk
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Poland; Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania.
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Poland
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Poland
| | - Zofia Łapińska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Poland
| | - Agnieszka Chwiłkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Poland
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Poland; Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| |
Collapse
|
9
|
Ungvari Z, Fekete M, Varga P, Lehoczki A, Munkácsy G, Fekete JT, Bianchini G, Ocana A, Buda A, Ungvari A, Győrffy B. Association between red and processed meat consumption and colorectal cancer risk: a comprehensive meta-analysis of prospective studies. GeroScience 2025:10.1007/s11357-025-01646-1. [PMID: 40210826 DOI: 10.1007/s11357-025-01646-1] [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: 02/11/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Increasing evidence suggests that red and processed meat consumption may elevate the risk of colorectal cancer (CRC), yet the magnitude and consistency of this association remain debated. This meta-analysis aims to quantify the relationship between red and processed meat intake and the risk of CRC, colon cancer, and rectal cancer using the most comprehensive set of prospective studies to date. We conducted a comprehensive search in PubMed, Web of Science, Cochrane Library, Embase, and Google Scholar databases from 1990 to November 2024, to identify relevant prospective studies examining red, processed, and total meat consumption in relation to colorectal, colon, and rectal cancer risk. Hazard ratios (HR) and 95% confidence intervals (CI) were extracted for each study and pooled using a random-effects model to account for variability among studies. Statistical evaluation was executed using the online platform MetaAnalysisOnline.com. A total of 60 prospective studies were included. Red meat consumption was associated with a significantly increased risk of colon cancer (HR = 1.22, 95% CI 1.15-1.30), colorectal cancer (HR = 1.15, 95% CI 1.10-1.21), and rectal cancer (HR = 1.22, 95% CI 1.07-1.39). Processed meat consumption showed similar associations with increased risk for colon cancer (HR = 1.13, 95% CI 1.07-1.20), colorectal cancer (HR = 1.21, 95% CI 1.14-1.28), and rectal cancer (HR = 1.17, 95% CI 1.05-1.30). Total meat consumption also correlated with an elevated risk of colon cancer (HR = 1.22, 95% CI 1.11-1.35), colorectal cancer (HR = 1.17, 95% CI 1.12-1.22), and rectal cancer (HR = 1.28, 95% CI 1.10-1.48). This meta-analysis provides robust evidence that high consumption of red and processed meats is significantly associated with an increased risk of colorectal, colon, and rectal cancers. These findings reinforce current dietary recommendations advocating for the limitation of red and processed meat intake as part of cancer prevention strategies.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College, Health Sciences Division/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Mónika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Jozsef Fodor Center for Prevention and Healthy Aging, Semmelweis University, Budapest, Hungary
| | - Péter Varga
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Jozsef Fodor Center for Prevention and Healthy Aging, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Division, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Jozsef Fodor Center for Prevention and Healthy Aging, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Division, Budapest, Hungary
| | - Gyöngyi Munkácsy
- Department of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, H- 1117, Budapest, Hungary
| | - János Tibor Fekete
- Department of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, H- 1117, Budapest, Hungary
| | - Giampaolo Bianchini
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alberto Ocana
- Experimental Therapeutics in Cancer Unit, Instituto de Investigación Sanitaria San Carlos (IdISSC), and CIBERONC, Madrid, Spain
- INTHEOS-CEU-START Laboratory, Facultad de Medicina, Universidad CEU San Pablo, 28668 Boadilla del Monte, Madrid, Spain
| | - Annamaria Buda
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Jozsef Fodor Center for Prevention and Healthy Aging, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Division, Budapest, Hungary
| | - Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
- Jozsef Fodor Center for Prevention and Healthy Aging, Semmelweis University, Budapest, Hungary.
| | - Balázs Győrffy
- Jozsef Fodor Center for Prevention and Healthy Aging, Semmelweis University, Budapest, Hungary
- Department of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, H- 1117, Budapest, Hungary
- Department of Biophysics, Medical School, University of Pecs, H- 7624, Pecs, Hungary
| |
Collapse
|
10
|
Cui Y, Mei J, Zhao S, Zhu B, Lu J, Li H, Bai B, Sun W, Jin W, Zhu X, Rao S, Yi Y. Identification of a PANoptosis-related long noncoding rna risk signature for prognosis and immunology in colon adenocarcinoma. BMC Cancer 2025; 25:662. [PMID: 40211224 PMCID: PMC11987197 DOI: 10.1186/s12885-025-14021-2] [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: 06/06/2024] [Accepted: 03/26/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND PANoptosis, a complex programmed cell death (PCD) pathway that includes apoptosis, pyroptosis and necroptosis, is significantly involved in the progression of cancers. Long noncoding RNAs (lncRNAs) play crucial roles in PCD. However, the predictive value of PANoptosis-related lncRNAs (PRlncRNAs) for colon adenocarcinoma (COAD) has not been established. METHODS Gene expression data and clinical characteristics of patients with COAD were obtained from The Cancer Genome Atlas database. Differential expression analysis and Pearson correlation analysis were used to identify PRlncRNAs. In addition to least absolute shrinkage and selection operator, univariate and multivariate Cox regression analyses were employed to obtain PRlncRNAs for constructing a risk signature. Patients with COAD in the training set, testing set and entire set were stratified into high- and low-risk groups for further comparison of survival prognosis, using the median risk score as the cut-off point. Time-dependent receiver operating characteristic curves, a nomogram and multivariate Cox regression analysis were conducted to validate the risk signature in the testing set and the entire set. In addition, critical pathways, immune infiltration cells, immune checkpoint-related genes, Tumor Immune Dysfunction and Exclusion (TIDE) scores and antitumour drugs were compared between the two risk groups in the entire set. Correlations between ferroptosis, cuproptosis, disulfidptosis and the PRlncRNA risk score were evaluated. Finally, a competitive endogenous RNA (ceRNA) network was established, and enrichment analysis of the predicted mRNAs was performed using Gene Ontology (GO) analysis. The Kaplan-Meier plotter database was used as an external database to confirm the accuracy of the risk signature in predicting patient prognosis. Additionally, small interfering RNA (siRNA), a cell counting kit- 8 assay, a cell colony formation assay, quantitative polymerase chain reaction (qPCR) and an apoptosis assay were further employed to investigate the roles of AP003555.1 in colon cancer. RESULTS A risk signature comprising four PRlncRNAs (LINC01133, FOXD3-AS1, AP001066.1, and AP003555.1) was developed to predict the prognosis of patients with COAD. Kaplan‒Meier curves demonstrated significant differences in prognosis between the high- and low-risk groups across the three sets. Multivariate Cox regression analysis confirmed that the risk signature was an independent prognostic factor across the three sets. A nomogram, receiver operating characteristic curves and calibration curves indicated strong confidence in the risk signature. Using the CIBERSORT algorithm and gene set enrichment analysis, variations in infiltrating immune cells and immune processes were observed between the two risk groups. Furthermore, TIDE algorithm suggested that the high-risk group exhibited a lower risk of immunotherapy escape and better immunotherapy outcomes than the low-risk group. Distinct responses to various antitumour drugs were observed between the two risk groups. Additionally, we constructed a ceRNA network based on PRlncRNAs, and GO enrichment analysis of the predicted mRNAs revealed different functions. In addition, the results of the Kaplan‒Meier plotter database revealed that patients who exhibited high levels of LINC01133 and FOXD3-AS1 experienced significantly shorter overall survival than those with low levels of these lncRNAs. Specifically, in terms of functionality, AP003555.1 was found to be highly expressed in colon cancer tissue and promoted viability and proliferation while suppressing the apoptosis of colon cancer cells. CONCLUSION We identified a novel risk signature consisting of four PRlncRNAs, which is an independent prognostic indicator for patients with COAD. This PRlncRNA risk signature is potentially relevant for immunotherapy and could serve as a therapeutic target for COAD.
Collapse
Affiliation(s)
- Yuekai Cui
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Mei
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shengsheng Zhao
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bingzi Zhu
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianhua Lu
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongzheng Li
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Binglong Bai
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weijian Sun
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenyu Jin
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xueqiong Zhu
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China.
- Wenzhou Medical University, Wenzhou, China.
| | - Shangrui Rao
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Yongdong Yi
- Second Affiliated Hospital & Yuying Childrens' & Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
11
|
Alkhaldy AA. Knowledge, attitudes, and practices toward colorectal cancer lifestyle risk factors among adults in Saudi Arabia. Front Nutr 2025; 12:1507563. [PMID: 40264557 PMCID: PMC12011586 DOI: 10.3389/fnut.2025.1507563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Background Despite an apparent increase in early-onset colorectal cancer (CRC) in Saudi Arabia, with the majority of patients being diagnosed at an advanced disease stage, no previous assessment of the knowledge, attitudes, and practices (KAP) toward its dietary and lifestyle-related risk factors has been reported. The aim of this study was to investigate the KAP levels with respect to these risk factors for CRC and examine possible associations between the studied variables among the Saudi population. Methods This cross-sectional study involved 1,040 participants aged 18 years or older. Data were collected by convenience sampling via a self-administered online questionnaire in Saudi Arabia between June and December 2023. Results A majority of participants (77.8%) displayed low knowledge about the dietary and lifestyle-related risk factors for CRC, while only 22.2% possessed high knowledge. Similarly, 78.6% of participants exhibited negative attitudes toward these risk factors, with just 21.4% having positive attitudes. Furthermore, 75.0% of participants reported engaging in poor practices, leaving only 25.0% demonstrating good practices related to CRC risk factors. Conclusions The findings of this study indicate insufficient KAP levels toward dietary and lifestyle-related risk factors for CRC in Saudi Arabia, highlighting the urgent need for nationwide initiatives and programs to promote improved knowledge, attitudes, and practices and reduce the effect of the risk factors contributing to CRC.
Collapse
Affiliation(s)
- Areej Ali Alkhaldy
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
12
|
Wang M, Liu K, Bao W, Hang B, Chen X, Zhu X, Li G, Liu L, Xiang H, Hu H, Lu Y, Song Z, Chen J, Wang Y. Gut microbiota protect against colorectal tumorigenesis through lncRNA Snhg9. Dev Cell 2025; 60:1008-1017.e7. [PMID: 39755115 DOI: 10.1016/j.devcel.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 09/30/2024] [Accepted: 12/05/2024] [Indexed: 01/06/2025]
Abstract
The intestinal microbiota is a key environmental factor in the development of colorectal cancer (CRC). Here, we report that, in the context of mild colonic inflammation, the microbiota protects against colorectal tumorigenesis in mice. This protection is achieved by microbial suppression of the long non-coding RNA (lncRNA) Snhg9. Snhg9 promotes tumor growth through inhibition of the tumor suppressor p53. Snhg9 suppresses p53 activity by dissociating the p53 deacetylase sirtuin 1 (SIRT1) from the cell cycle and apoptosis regulator 2 (CCAR2). Consequently, the depletion of the microbiota by antibiotics causes upregulation of Snhg9 and accelerates CRC progression. Moreover, Snhg9 is functionally conserved. Human SNHG9 promotes tumor growth via the same mechanism as mouse Snhg9, despite their low sequence similarity.
Collapse
Affiliation(s)
- Meng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Kailin Liu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Wu Bao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Bingqing Hang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Xianjiong Chen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Xinyi Zhu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Guifang Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China
| | - Lihong Liu
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Haoyi Xiang
- Department of Colorectal Surgery and Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Hai Hu
- Breast Cancer Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Yanhui Lu
- School of Nursing, Peking University, Beijing 100191, China
| | - Zhangfa Song
- Department of Colorectal Surgery and Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China.
| | - Jiaxin Chen
- Department of Breast Surgery and Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China.
| | - Yuhao Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310029, Zhejiang, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310029, Zhejiang, China.
| |
Collapse
|
13
|
Fukumori C, Ken Kawassaki R, Daré RG, Lopes LB. Polymer-lipid hybrid microcarriers for oral codelivery of paclitaxel and tributyrin: development, optimization, and cytotoxicity in cells and spheroids of colorectal cancer. Int J Pharm 2025; 676:125549. [PMID: 40189171 DOI: 10.1016/j.ijpharm.2025.125549] [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: 02/04/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/19/2025]
Abstract
Colorectal cancer (CRC) is the third most frequent cancer worldwide. Despite advances in treatment, conventional chemotherapy suffers from severe side effects and limited drug selectivity, highlighting the importance of alternative therapies. In this study, a polymer-lipid hybrid microcarrier was developed for oral co-administration of paclitaxel (PTX) and tributyrin (TB) as a novel approach for CRC therapy. The microcarrier was designed with a pH-sensitive polymeric shell that encapsulates drug-loaded nanostructured lipid carriers (NLC); shell dissolution at intestinal pH enables localized release of the NLC. The methodological approach employed an emulsion of vegetable oil and NLC as a template for polymer deposition. Multiple parameters were optimized, including polymers ratios, NLC dilution, acid concentration, and sonication time. Spherical hybrid particles with smooth surface and mean size of 1000 nm were obtained; PTX encapsulation efficiency was 99.9 ± 0.2 %, with a production yield of 97.2 ± 0.08 %. Drug release followed the Korsmeyer-Peppas kinetic model. Cytotoxic evaluation in human colorectal adenocarcinoma HCT-116 monolayers showed that PTX encapsulation increased cytotoxicity, lowering IC50 to 83.7 nM compared to 199.5 nM for free PTX. The addition of TB further improved cytotoxicity, reducing the IC50 to 60.8 nM. A similar potentiation cytotoxicity was observed in spheroids. The microcarrier induced reductions in colony formation, alterations in cell cytoskeleton, and led to a significant reduction in P-glycoprotein expression compared to its free form, suggesting its potential to help to overcome drug resistance. These results point to the promising applicability of the hybrid microcarrier as an innovative delivery system for oral administration of cytotoxic agents.
Collapse
Affiliation(s)
- Claudio Fukumori
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Rodrigo Ken Kawassaki
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Regina G Daré
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luciana B Lopes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
14
|
Lu YN. Blocking lncRNA NOP14-AS1 overcomes 5-Fu resistance of colon cancer cells by modulating miR-30a-5p-LDHA-glucose metabolism pathway. Discov Oncol 2025; 16:458. [PMID: 40180667 PMCID: PMC11968611 DOI: 10.1007/s12672-025-02156-4] [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: 12/14/2024] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
Colorectal cancer (CRC) is a malignant digestive tumor associated with high mortality rate. Currently, 5-Fu therapy is a frequently used treatment approach for CRC. Yet, acquirement of 5-Fu resistance ultimately leads to therapeutic failure in CRC patients. LncRNA NOP14-AS1 was upregulated in cancers, but its biological functions and mechanisms in 5-Fu resistant colorectal cancer remain elusive. We discovered that NOP14-AS1 was high-expressed in colorectal tumors and cancer cells. Silencing NOP14-AS1 sensitized CRC cells to 5-Fu. By creating a 5-Fu resistant CRC cell line (HT-29 5-Fu R) and observed that expression of NOP14-AS1 was remarkedly elevated in 5-Fu resistant cells compared to parental cells. Additionally, we found that miRNA-30a-5p was a target of NOP14-AS1 and directly affected its function. miR-30a-5p overexpression sensitized CRC cells to 5-Fu treatment and targeted the glycolysis key enzyme, LDHA. Rescue experiments showed that restoring LDHA in CRC cells which were overexpressing miR-30c-5p successfully overridden 5-Fu resistance. Importantly, restoring miR-30a-5p in NOP14-AS1-overexpressing cells effectively restored 5-Fu sensitivity in HT-29 5-Fu R cells by targeting the LDHA-mediated glucose metabolism. In summary, our results revealed that lncRNA NOP14-AS1 promotes 5-Fu resistance by mediating the miR-30a-5p-LDHA axis.
Collapse
Affiliation(s)
- Ya-Nan Lu
- School of Medicine, Xuchang University, No.2111 Xinghua Road, Weidu District, Xuchang, 461000, Henan, China.
| |
Collapse
|
15
|
Wang H, Wang KN, Hua J, Tang Y, Chen Y, Zhou GQ, Li S. Dynamic spectrum-driven hierarchical learning network for polyp segmentation. Med Image Anal 2025; 101:103449. [PMID: 39847953 DOI: 10.1016/j.media.2024.103449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/06/2024] [Accepted: 12/26/2024] [Indexed: 01/25/2025]
Abstract
Accurate automatic polyp segmentation in colonoscopy is crucial for the prompt prevention of colorectal cancer. However, the heterogeneous nature of polyps and differences in lighting and visibility conditions present significant challenges in achieving reliable and consistent segmentation across different cases. Therefore, this study proposes a novel dynamic spectrum-driven hierarchical learning model (DSHNet), the first to specifically leverage image frequency domain information to explore region-level salience differences among and within polyps for precise segmentation. A novel spectral decoupler is advanced to separate low-frequency and high-frequency components, leveraging their distinct characteristics to guide the model in learning valuable frequency features without bias through automatic masking. The low-frequency driven region-level saliency modeling then generates dynamic convolution kernels with individual frequency-aware features, which regulate region-level saliency modeling together with the supervision of the hierarchy of labels, thus enabling adaptation to polyp heterogeneous and illumination variation simultaneously. Meanwhile, the high-frequency attention module is designed to preserve the detailed information at the skip connections, which complements the focus on spatial features at various stages. Experimental results demonstrate that the proposed method outperforms other state-of-the-art polyp segmentation techniques, achieving robust and superior results on five diverse datasets. Codes are available at https://github.com/gardnerzhou/DSHNet.
Collapse
Affiliation(s)
- Haolin Wang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China; Jiangsu Key Laboratory of Biomaterials and Devices, Southeast University, Nanjing, China
| | - Kai-Ni Wang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China; Jiangsu Key Laboratory of Biomaterials and Devices, Southeast University, Nanjing, China
| | - Jie Hua
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Tang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China; Jiangsu Key Laboratory of Biomaterials and Devices, Southeast University, Nanjing, China
| | - Yang Chen
- Laboratory of Image Science and Technology, Southeast University, Nanjing, China; Key Laboratory of Computer Network and Information Integration, Southeast University, Nanjing, China
| | - Guang-Quan Zhou
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China; Jiangsu Key Laboratory of Biomaterials and Devices, Southeast University, Nanjing, China.
| | - Shuo Li
- Department of Computer and Data Science and Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| |
Collapse
|
16
|
Zheng B, Wang H, Zhai S, Li J, Lu K. Mitochondria-targeted photothermal-chemodynamic therapy enhances checkpoint blockade immunotherapy on colon cancer. Mater Today Bio 2025; 31:101542. [PMID: 40018055 PMCID: PMC11867542 DOI: 10.1016/j.mtbio.2025.101542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/28/2025] [Accepted: 02/01/2025] [Indexed: 03/01/2025] Open
Abstract
Immunotherapy has emerged as a hotspot for cancer treatment. However, the response rate of monotherapy remains relatively low in clinical settings. Photothermal therapy (PTT), which employs light energy to ablate tumors, can also activate tumor-specific immune responses. This effect has been attributed in several studies to the release of damage-associated molecular patterns (DAMPs) triggered by mitochondrial injury. We propose that mitochondria-targeted PTT may better synergize with immunotherapy. Herein, we constructed a multifunctional nanoplatform that enables mitochondria-targeted photothermal-chemodynamic combination therapy by conjugating indocyanine green-thiol (ICG-SH) and mercaptoethyl-triphenylphosphonium (TPP-SH) onto polyvinyl pyrrolidone (PVP)-coated gold-copper nanoparticles (AIT). Upon near-infrared light (NIR) irradiation, AIT ablates cancer cells and amplifies the effect of chemodynamic therapy (CDT), thereby inducing apoptosis in the tumor. The combination of CDT and PTT promotes immunogenic cell death, which could synergize with checkpoint blockade immunotherapy. In a bilateral mouse colon cancer model, we observed complete eradication of light-irradiated primary tumors and significant inhibition of distant untreated tumors in the group treated with AIT plus anti-PD-1 (αPD-1). We found a significant increase in serum levels of pro-inflammatory factors, including interleukin-6 (IL-6), interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α), following PTT/CDT/immunotherapy treatment, suggesting effective activation of the immune response. The enhanced immunogenicity caused by AIT with αPD-1 treatment resulted in efficient antigen presentation, as indicated by the increased infiltration of dendritic cells (DCs) into the tumor-draining lymph nodes (LNs). We also observed enhanced infiltration of CD8+ T cells in distant tumors in the AIT with αPD-1 group compared to αPD-1 alone. Hence, mitochondria-targeting represents an effective strategy to potentiate the combination of photothermal, chemodynamic, and immune checkpoint blockade therapies for the treatment of metastatic cancer.
Collapse
Affiliation(s)
- Benchao Zheng
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, PR China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, PR China
| | - Hongbo Wang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, PR China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, PR China
| | - Shiyi Zhai
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, PR China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, PR China
| | - Jiangsheng Li
- Key Laboratory of Carcinogenesis and Translational Research of Ministry of Education, Key Laboratory for Research and Evaluation of Radiopharmaceuticals of National Medical Products Administration, Department of Nuclear Medicine, Peking University Cancer Hospital, Beijing, 100142, PR China
| | - Kuangda Lu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, PR China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, PR China
| |
Collapse
|
17
|
Zou J, Chen J, Deng L, Xu B, Yu T, Wang J, He C. Mechanistic insights into SENP1 and OCT4 interaction in promoting drug resistance and stem cell features in colon cancer. Am J Physiol Cell Physiol 2025; 328:C1260-C1278. [PMID: 40063360 DOI: 10.1152/ajpcell.00817.2024] [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: 11/04/2024] [Revised: 11/18/2024] [Accepted: 02/17/2025] [Indexed: 04/01/2025]
Abstract
This study explores the molecular mechanism by which sentrin/SUMO-specific protease 1 (SENP1) promotes cisplatin (Cis) resistance and tumor stem cell characteristics in colon adenocarcinoma (COAD) through deSUMOylation-mediated modification of octamer-binding transcription factor 4 (OCT4). By analyzing single-cell and transcriptome sequencing datasets, we identified key genes and regulatory pathways in both resistant and sensitive COAD cells. Malignant cells were isolated and evaluated for stemness using the infercnv package, and differential genes between Cis-resistant and -sensitive groups were identified. Machine learning algorithms highlighted essential genes, and databases predicted interaction sites between OCT4 and SENP1. In vitro experiments using enriched HCT116 stem cells revealed that SENP1 and OCT4 expression significantly elevated CD44 and CD133 levels, enhancing stemness. Functional assays showed that SENP1's deSUMOylation of OCT4 intensified Cis resistance, migration, and invasion in cisplatin-resistant cell line 116 (Cis-116) cells. In vivo, SENP1 knockdown reduced tumor growth and stem cell markers, whereas OCT4 overexpression escalated tumor metastasis and structural damage. These findings demonstrate that SENP1's modulation of OCT4 is central to COAD's resistance and stem cell properties, offering a novel target for COAD therapy.NEW & NOTEWORTHY This study uncovers the critical role of SENP1 in regulating OCT4 through deSUMOylation, driving Cis resistance and tumor stemness in COAD. Targeting this pathway may provide novel therapeutic strategies for COAD management.
Collapse
Affiliation(s)
- Jun Zou
- Department of Abdominal Oncology Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Jing Chen
- Department of nursing, Nanchang Medical College, Nanchang, People's Republic of China
| | - Lei Deng
- Department of Medical Oncology, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Bangran Xu
- Department of Abdominal Oncology Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Jun Wang
- General Surgery Department of the Trauma Center, Nanchang First Hospital, Nanchang, People's Republic of China
| | - Chongwu He
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| |
Collapse
|
18
|
Moodley Y, Brink W, van Wyk J, Kader S, Wexner SD, Neugut AI, Kiran RP. Risk Model for Predicting Gaps in Surgical Oncology Care Among Patients With Stage I-III Rectal Cancer From KwaZulu-Natal, South Africa. JCO Glob Oncol 2025; 11:e2400480. [PMID: 40249891 DOI: 10.1200/go-24-00480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 02/14/2025] [Accepted: 03/20/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE Gaps in surgical oncology care (GISOC), including delayed or nonreceipt of surgery, are detrimental to cancer control. This research sought to develop a risk model for predicting GISOC in South African rectal cancer (RC) patients with localized disease. METHODS This retrospective cohort study analyzed data from an existing colorectal cancer patient registry. GISOC was defined as surgery received >62 days after diagnosis with stage I-III RC or nonreceipt of surgery for stage I-III RC. Patient demographics, comorbidity, disease staging, and neoadjuvant therapy receipt were included as covariates in the analysis. A supervised logistic regression machine learning algorithm was used to train and test an appropriate risk model, which was translated into a nomogram. Receiver operating characteristic curve analyses and AUC assessments were used to establish the nomogram's performance. RESULTS The analysis included 490 patients (training data set = 245, testing data set = 245). Overall, there were 242 patients who experienced GISOC (49.4%), of whom 33 (13.6%) did not receive surgery and 209 (86.4%) had a delay in receiving surgery. The trained risk model consisted of patient race (Indian, odds ratio [OR] = 0.24; White, OR = 0.23; v Black), comorbidity (OR = 2.29 v no comorbidity), and neoadjuvant therapy receipt (OR = 18.40 v nonreceipt). AUCs for the risk model were >0.800. CONCLUSION An accurate, setting-specific risk model and nomogram was developed for predicting GISOC in patients with RC. The nomogram can be implemented without the use of technology to identify patients at high risk for GISOC, who can then be targeted with risk-reduction interventions. The impact of the nomogram on existing surgical unit workflows requires further investigation.
Collapse
Affiliation(s)
- Yoshan Moodley
- Gastrointestinal Cancer Research Group, Department of Surgery, University of KwaZulu-Natal, Durban, South Africa
- Division of Health Systems and Public Health, Stellenbosch University, Cape Town, South Africa
| | - Willie Brink
- Division of Applied Mathematics, Stellenbosch University, Cape Town, South Africa
| | - Jacqueline van Wyk
- Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Health Sciences Education, University of Cape Town, Cape Town, South Africa
| | - Shakeel Kader
- Gastrointestinal Cancer Research Group, Department of Surgery, University of KwaZulu-Natal, Durban, South Africa
| | - Steven D Wexner
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL
| | - Alfred I Neugut
- Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY
| | - Ravi P Kiran
- Department of Surgery, Columbia University, New York, NY
| |
Collapse
|
19
|
Haq MU, Pritchard DM, Myint AS, Javed MA, Duckworth CA, Than NW, Bonnett LJ, Hughes DM. Clinical Prediction Models for Contact X-Ray Brachytherapy in Managing Rectal Cancers: A Scoping Review. Cancer Med 2025; 14:e70697. [PMID: 40178039 PMCID: PMC11966560 DOI: 10.1002/cam4.70697] [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: 05/27/2024] [Revised: 12/13/2024] [Accepted: 02/05/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Currently, there are no clinically predictive models that can prognosticate the response of rectal cancers to Contact X-ray brachytherapy (CXB). This review aims to critically evaluate existing models that have attempted to predict the response of rectal cancer to external beam radiotherapy, with the objective of laying the foundation for the development of a CXB-specific prediction model. METHODS A random-effects meta-analysis was employed to calculate pooled estimates of the discriminative ability of published models. Using the Prediction Model Risk Of Bias Assessment Tool (PROBAST), each model was evaluated for its risk of bias and applicability. Additionally, the frequency of commonly utilised predictive factors was documented. RESULTS Twelve papers discussed fifteen models based on pre-treatment factors. Models predicting response based on the Tumour regression grade (TRG) classified responders as patients who achieved a complete response or near complete response and achieved a pooled AUC of 0.82 (95% CI 0.74-0.89). Models that predicted pathologic complete response (pCR) had a pooled AUC of 0.76 (95% CI 0.71-0.82). The most utilised predictive parameters were age, tumour grade and T stage. However, these models were prone to significant risk of bias and had limited applicability to the general population. CONCLUSIONS Although the existing models were statistically robust, they lacked broad applicability. This was primarily due to a lack of external validation, which limits their clinical utility. A future CXB-specific model should prioritise dedicated data collection based on pre-calculated sample size and include the predictive factors identified in this review.
Collapse
Affiliation(s)
- Muneeb Ul Haq
- Institute of Systems, Molecular and Integrative BiologyThe University of LiverpoolLiverpoolUK
- The Clatterbridge Cancer Centre NHS Foundation TrustLiverpoolUK
| | - D. Mark Pritchard
- Institute of Systems, Molecular and Integrative BiologyThe University of LiverpoolLiverpoolUK
- Liverpool University Hospitals NHS Foundation TrustLiverpoolUK
| | | | - Muhammad Ahsan Javed
- Liverpool University Hospitals NHS Foundation TrustLiverpoolUK
- Institute of Life Course and Medical SciencesThe University of LiverpoolLiverpoolUK
| | - Carrie A. Duckworth
- Institute of Systems, Molecular and Integrative BiologyThe University of LiverpoolLiverpoolUK
| | - Ngu Wah Than
- Institute of Systems, Molecular and Integrative BiologyThe University of LiverpoolLiverpoolUK
- The Clatterbridge Cancer Centre NHS Foundation TrustLiverpoolUK
| | - Laura J. Bonnett
- Department of Health Data Science, Institute of Population HealthThe University of LiverpoolLiverpoolUK
| | - David M. Hughes
- Department of Health Data Science, Institute of Population HealthThe University of LiverpoolLiverpoolUK
| |
Collapse
|
20
|
An Y, Gong J, Xiao A. Development and validation of nomograms for predicting the prognosis of colorectal cancer patients. Transl Cancer Res 2025; 14:1651-1663. [PMID: 40224992 PMCID: PMC11985181 DOI: 10.21037/tcr-24-1924] [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: 10/10/2024] [Accepted: 01/22/2025] [Indexed: 04/15/2025]
Abstract
Background Accurate prognosis prediction is essential in colorectal cancer (CRC) for guiding treatment decisions, yet the traditional tumor-node-metastasis (TNM) staging system often lacks precision. This study aimed to develop improved prognostic tools for CRC patients. Methods Prognostic nomogram models were developed using data from 2,435 CRC patients who underwent curative resection. Parameters were selected via least absolute shrinkage and selection operator (LASSO) regression to include overall survival (OS) and disease-free survival (DFS) nomograms. The performance of these nomograms was evaluated against the TNM staging system using ROC analysis, calibration curves, and decision curve analysis (DCA). Results Critical prognostic factors identified included tumor invasion depth, distant metastasis, tumordifferentiation grade, extranodal tumor deposits (ENTD), R1 resection, and log odds of positive lymph nodes (LODDS). The OS nomogram demonstrated area under the curve (AUC) values of 0.786, 0.776, and 0.803 for predicting 1-, 3-, and 5-year survival, respectively, compared to 0.768, 0.750, and 0.782 for TNM staging. The DFS nomogram predicted 1-, 3-, and 5-year DFS with AUCs of 0.764, 0.777, and 0.789, respectively, compared to 0.762, 0.761, and 0.770 for TNM staging. Calibration plots indicated strong predictive capabilities, and DCA confirmed greater net benefits over TNM staging. Conclusions Our developed prognostic nomogram models offer enhanced accuracy over traditional TNM staging in predicting CRC prognosis. Integrating these models into clinical practice can potentially improve personalized treatment strategies for postoperative CRC patients, enhancing overall clinical outcomes.
Collapse
Affiliation(s)
- Yingqi An
- Department of Gastrointestinal Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianping Gong
- Department of Gastrointestinal Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aitang Xiao
- Department of Gastrointestinal Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Naldi L, Peri A, Fibbi B. Apelin/APJ: Another Player in the Cancer Biology Network. Int J Mol Sci 2025; 26:2986. [PMID: 40243599 PMCID: PMC11988549 DOI: 10.3390/ijms26072986] [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: 12/30/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
The apelinergic system exerts multiple biological activities in human pathologies, including cancer. Overactivation of apelin/APJ, which has been detected in many malignant tumors, and the strong correlation with progression-free and overall survival, suggested the role of an oncogene for the apelin gene. Emerging evidence sheds new light on the effects of apelin on cellular functions and homeostasis in cancer cells and supports a direct role for this pathway on different hallmarks of cancer: "sustaining proliferative signaling", "resisting cell death", "activating invasion and metastasis", "inducing/accessing vasculature", "reprogramming cellular metabolism", "avoiding immune destruction" and "tumor-promoting inflammation", and "enabling replicative immortality". This article reviews the currently available literature on the intracellular processes regulated by apelin/APJ, focusing on those pathways correlated with tumor development and progression. Furthermore, the association between the activity of the apelinergic axis and the resistance of cancer cells to oncologic treatments (chemotherapy, immunotherapy, radiation) suggests apelin/APJ as a possible target to potentiate traditional therapies, as well as to develop diagnostic and prognostic applications. This issue will be also covered in the review.
Collapse
Affiliation(s)
- Laura Naldi
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Alessandro Peri
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Benedetta Fibbi
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| |
Collapse
|
22
|
Wang Y, Zhang X, Zhang Y, Shi F, Du S, Zhang Z, Zhao C, Luo S, Wang P, Wu D, She J. Sprayed hyaluronic acid based multidrug composite hydrogel for postoperative colorectal cancer ultra-efficient long-lasting multi-stage immuno-chemo synergistic therapy. MATERIALS HORIZONS 2025. [PMID: 40129385 DOI: 10.1039/d5mh00108k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
To enhance the therapeutic efficacy of postoperative colorectal cancer treatment and prevent peritoneal metastasis, we propose a strategy utilizing the photothermal-induced ultra-efficient and long-lasting multi-stage immuno-chemo synergistic therapy. To implement this strategy, oxaliplatin (OXA), curcumin (Cur), and Mn2+ were coordinated to form infinite coordination polymer nanoparticles (OXA-Mn(II)-Cur ICP NPs). These nanoparticles are encapsulated with polydopamine (PDA) to create OXA-Mn(II)-Cur ICP@PDA NPs, which are subsequently embedded in a sprayable hyaluronic acid-based hydrogel. The resulting ICP@PDA NPs@composite hydrogel exhibits strong tissue adhesion and segmented pH-responsive drug release properties. Notably, the hydrogel can sustainably release drugs for over 20 days in vivo, maximizing local drug concentration while minimizing systemic toxic side effects. Each component of the composite hydrogel serves multiple functions, and its application to postoperative tumor sites enables long-term, dual-pathway, multi-stage immune activation. This immune response synergizes with chemotherapy to achieve a highly effective therapeutic outcome. In vivo experiments demonstrated that the composite hydrogel effectively eliminates residual tumors, ensuring a 100% survival rate without recurrence for 80 days in treated mice. Furthermore, it inhibits peritoneal metastasis and completely eradicates intraperitoneal tumors within 20 days. The ICP@PDA NPs@composite hydrogel represents a promising therapeutic platform for postoperative colorectal cancer treatment and metastasis prevention.
Collapse
Affiliation(s)
- Ya Wang
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, P. R. China
| | - Xiaojiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, P. R. China.
| | - Yujie Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, P. R. China.
| | - Feiyu Shi
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, P. R. China.
| | - Siyuan Du
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, P. R. China
| | - Zhe Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, P. R. China.
| | - Chenyu Zhao
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.
| | - Siyuan Luo
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.
| | - Pengqian Wang
- Department of Chemical Engineering, School of Water and Environment, Chang'an University, 710064 Xi'an, P. R. China.
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, P. R. China.
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, P. R. China
| |
Collapse
|
23
|
Nourbakhsh ST, Mirzaei SA, Mohamadhashem F, Naghizadeh MM, Razavi AN, Mansoori Y, Daraei A, Mohamadhashem F. Pathological expression of mitochondrial genome-derived circRNA SCAR/mc-COX2 and its ceRNA network in colorectal cancer: implications for clinical significance. BMC Cancer 2025; 25:466. [PMID: 40082804 PMCID: PMC11907809 DOI: 10.1186/s12885-025-13886-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Mitochondrial-encoded circular RNAs (mecciRNAs) are a newly discovered class of mitochondrial-encoded non-coding RNAs (mt-ncRNAs) that play important biological roles in the cell. This study aimed to examine the expression profile of SCAR/mc-COX2 (has_circ_0089762) in colorectal cancer (CRC) and its relationship with clinicopathological variables. Furthermore, to better understand SCAR/mc-COX2's functional role in CRC, we constructed a competing endogenous RNA (ceRNA) network. METHODS Quantitative real-time PCR (qRT-PCR) was employed to analyze the expression levels of SCAR/mc-COX2 in 40 pairs of CRC samples, consisting of 40 tumor samples and 40 adjacent non-tumoral samples from patients. The ceRNA regulatory network was constructed using online bioinformatics tools. Furthermore, the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene Ontology (GO) enrichment analysis were conducted using the Enrichr database. RESULTS The results demonstrated a significant decrease in SCAR/mc-COX2 expression in tumor tissues compared to adjacent non-tumoral tissues (p-value<0.05). In another finding, a significant relationship was observed between pathological T staging and the expression status of SCAR/mc-COX2 (p-value=0.02). Additionally, the Receiver Operating Characteristic (ROC) curve analysis revealed that SCAR/mc-COX2 had an area under the curve (AUC) of 0.77, with 80% sensitivity and 75% specificity. Finally, a ceRNA regulatory network including SCAR/mc-COX2, 5 miRNA, and 9 mRNAs was found. CONCLUSION These findings suggest that SCAR/mc-COX2 may act as a tumor suppressor in CRC, and its dysregulation could play a crucial role in the pathophysiology of this cancer. The significant association with pathological T staging and its robust diagnostic performance (AUC = 0.77, sensitivity = 80%, specificity = 75%) highlight its potential as a novel biomarker for CRC detection and prognosis. Further functional studies are required to elucidate its precise role in CRC tumorigenesis and clinical applicability.
Collapse
Affiliation(s)
- Seyed Taha Nourbakhsh
- Department of Medical Genetics, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Mohamadhashem
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amir Nader Razavi
- Iran National Tumor Bank, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | - Faezeh Mohamadhashem
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
24
|
Altan Z, Johnson R. Targeting Wnt signalling through LINC02418: insights from CRISPR screens. Gut 2025; 74:513-515. [PMID: 39978931 DOI: 10.1136/gutjnl-2024-334266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/31/2025] [Indexed: 02/22/2025]
Affiliation(s)
- Zekiye Altan
- School of Biology and Environmental Science, University College Dublin, Dublin, Leinster, Ireland
- School of Medicine, University College Dublin, Dublin, Leinster, Ireland
| | - Rory Johnson
- School of Biology and Environmental Science, University College Dublin, Dublin, Leinster, Ireland
- University College Dublin Conway Institute of Biomolecular and Biomedical Research, Dublin, Leinster, Ireland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Medical Oncology, University of Bern, Bern, Switzerland
| |
Collapse
|
25
|
Ofluoğlu CB, Mülküt F, Aydın İC, Karahan M. Early Detection and Age-Comparative Analysis of Colorectal Cancer Screening: Insights from the Turkish Population. Curr Oncol 2025; 32:153. [PMID: 40136357 PMCID: PMC11941067 DOI: 10.3390/curroncol32030153] [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: 01/27/2025] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND This study aimed to evaluate the diagnostic yield of colonoscopy in asymptomatic individuals aged 45-49 years compared with those aged 50-54 years in a Turkish population, providing insights into region-specific screening strategies. METHODS This retrospective multicenter study was conducted across three tertiary endoscopy units in Turkey. Screening colonoscopy data from 3943 asymptomatic individuals aged 45-54 years between 2018 and 2023 were analyzed. The patients were stratified into two groups: 45-49 years (Group 1) and 50-54 years (Group 2). Demographic characteristics, polyp size, histological features, and prevalence of early-onset advanced colorectal neoplasia (EAO-aCRN) were assessed. RESULTS A total of 3943 patients were included, with 862 in Group 1 (45-49 years) and 3081 in Group 2 (50-54 years). The polyp detection rate was 16.6% in Group 1 and 22.9% in Group 2 (p < 0.001). The adenoma detection rates were 10.8% and 13.9% in Groups 1 and 2, respectively (p = 0.018). The advanced polyp detection rates were 3.2% and 7.3% in Groups 1 and 2, respectively (p < 0.001). Mean polyp size was 6.5 ± 5.1 mm in Group 1 and 8.8 ± 8.4 mm in Group 2 (p < 0.001). The mean number of polyps per patient was 1.5 ± 0.8 in Group 1 and 1.9 ± 1.6 in Group 2 (p = 0.023). Advanced neoplasia was detected in 16.6% of Group 1 patients compared with 22.9% of Group 2 patients (p < 0.001). CONCLUSIONS While CRC screening at age 45 demonstrated lower detection rates of polyps and advanced neoplasia than at age 50, the higher prevalence of EAO-CRN among 45-49-year-olds in Turkey underscores the importance of early screening in high-risk populations. Tailored regional strategies incorporating individual risk factors are crucial for optimizing CRC prevention policies.
Collapse
Affiliation(s)
- Cem Batuhan Ofluoğlu
- Department of Gastroenterology Surgery, Sancaktepe Training and Research Hospital, University of Health Sciences, 34785 Istanbul, Turkey
| | - Fırat Mülküt
- Department of General Surgery, Sancaktepe Training and Research Hospital, University of Health Sciences, 34785 Istanbul, Turkey;
| | - İsa Caner Aydın
- Ministry of Health, Department of Gastroenterology Surgery, Zonguldak Ataturk State Hospital, 34147 Zonguldak, Turkey;
| | - Mehmet Karahan
- Department of General Surgery, Kartal Dr. Lütfi Kırdar City Hospital, University of Health Sciences, 34130 Istanbul, Turkey;
| |
Collapse
|
26
|
Fan Y, Wang Q, Zhang Y, Wang Y, Li W, Jiang S, Duan JN. Mechanism of Guishao Yigong decoction in treating colorectal cancer based on network pharmacology and experimental validation. J Pharm Pharmacol 2025; 77:430-445. [PMID: 39352002 DOI: 10.1093/jpp/rgae045] [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: 11/17/2023] [Accepted: 04/05/2024] [Indexed: 03/06/2025]
Abstract
OBJECTIVES To explore the effective components of Guishao Yigong decoction (GYD) in the treatment of colorectal cancer and reveal its potential mechanism of action. METHODS Through network pharmacology, the main target and signaling pathway of GYD therapy for colorectal cancer (CRC) were found. Subsequently, the effect of GYD was verified by in vitro cell viability measurements, colony formation, and scratch healing tests. The effects of GYD on metabolic pathways in vivo were found through plasma metabolomics. Finally, flow cytometry and qPCR experiments were used to verify the cycle-blocking effect of GYD on CRC cells. KEY FINDINGS Based on the network pharmacological analysis and molecular docking technology, it was found that GYD could restrain the growth of CRC cells by affecting lipid metabolic pathways and mitogen-activated protein kinase (MAPK) signaling pathways. A series of cell experiments showed that GYD could inhibit the proliferation, migration and clonogenic ability of CRC cells. Furthermore, the plasma metabolomics results showed that GYD could affect the production of unsaturated fatty acids in mice. Flow cytometry and qPCR experiments further proved that GYD blocked the CRC cells in the G1 phase and modulated the expression of cell cycle-related targets, such as AKT, TP53, CDKN1A, and CDK2. CONCLUSIONS All the results indicated that GYD could regulate the related metabolism of unsaturated fatty acids. Thus, the cell cycle was blocked and the expressions of the key proteins such as AKT and TP53 were regulated, which achieved the purpose of intervention in colorectal cancer.
Collapse
Affiliation(s)
- Yuwen Fan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Quyi Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Yun Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Yu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Wenwen Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Ji-Nao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| |
Collapse
|
27
|
Demirci MG, Kesgin YM. Virtual Colonoscopy: Retrospective Comparison of the Findings in Supine and Prone Positions. Surg Innov 2025:15533506251325349. [PMID: 40033191 DOI: 10.1177/15533506251325349] [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: 03/05/2025]
Abstract
BACKGROUND Early detection of colorectal cancer (CRC) is significantly associated with reduced morbidity and mortality. Virtual colonoscopy (VC) is a minimally invasive, safe and well-tolerated alternative procedure to traditional colonoscopy. Therefore, we aimed to evaluate the findings of VC particularly in supine and prone positions as well as to contribute to the practical challenges of procedure. METHODS Total number of 20 patients who underwent VC were included in this retrospective study. After proper bowel cleansing was achieved, intestinal dilatation was performed by injecting air into the rectum. Two different shots were performed in the supine and prone positions. Additionally, intestinal diameters were measured from the cecum to the rectum at their widest point via 2-dimensional coronal reformat. RESULTS Polyps were detected in 3 patients which were confirmed by optical colonoscopy. The mean cecum diameter was detected as the largest diameter in the supine and prone examinations. In both supine and prone examinations, the distal descending colon was the most challenging site. Additionally, the mean descending colon diameter calculated in the prone position (40.9 ± 6.4 mm) was found to be statistically larger than descending colon diameter calculated in the supine position (36.1 ± 5.3 mm) (P = 0.001). CONCLUSIONS Our findings clearly demonstrated that combination of prone and supine scanning provides clear field of vision on narrow parts of the colon which improves accurate estimation for polyp detection. Furthermore, VC appears to be more comfortable, safe, fast, and cost-efffective procedure for CRC screening with advantages of low radiation exposure, extracolonic findings and lack of sedation requirements.
Collapse
Affiliation(s)
| | - Yasir Musa Kesgin
- Department of General Surgery, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, University of Health Sciences, Istanbul, Turkiye
| |
Collapse
|
28
|
Shan G, Jiang J, Ji L, Li S, Wang Z, Yang S, Shen Q. Evaluation of active substances in gamboge and their mechanisms for the treatment of colorectal cancer by UPLC-MS/MS integrated with network pharmacology. Anal Biochem 2025; 698:115747. [PMID: 39647807 DOI: 10.1016/j.ab.2024.115747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 12/10/2024]
Abstract
Gamboge exhibits anti-colorectal cancer (CRC) activity, however, its active compounds and the underlying mechanisms remain unclear. Herein, a liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method for determining gambogellic acid, β-morellic acid, isogambogenic acid, gambogenic acid, R-gambogic acid, S-gambogic acid, and hydroxygambogic acid in gamboge was established. The key parameters including ion transitions, voltages, LOD, and LOQ were determined, with LOD ranging from 0.8 to 2.0 ng mL-1 and LOQ from 2.7 to 6.7 ng mL-1. The recovery rates were found to be between 95.6 % and 103.5 %. Furthermore, the active compounds were successfully determined, and molecular mechanisms of gamboge in treating CRC were explored. Network pharmacology revealed a "compound-target-pathway" network where the seven compounds could target key proteins, modulate PI3K-Akt and JAK-STAT pathways, and inhibit CRC development. Molecular docking validated SRC, SATA3, PIK3CA, among others, as potential targets for the active compounds in CRC intervention. In conclusion, this method significantly reduces analysis time and improves efficiency relative to existing approaches, making it highly suitable for the effective determination of multiple compounds in the quality control of gamboge materials.
Collapse
Affiliation(s)
- Guodong Shan
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiajun Jiang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Liting Ji
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Shiyan Li
- Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Zejun Wang
- Laboratory of Medicine-Food Homology Innovation and Achievement Transformation, Linping Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, 311110, China.
| | - Shaohui Yang
- Department of Colorectal Surgery, Lihuili Hospital, Ningbo Medical Center, Ningbo, 315040, China.
| | - Qing Shen
- Panvascular Diseases Research Center, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China; Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China.
| |
Collapse
|
29
|
Shahzad M, Hameed H, Amjad A, Khan MA, Qureshi IS, Hameed A, Saeed A, Munir R. An updated landscape on nanopharmaceutical delivery for mitigation of colon cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2107-2125. [PMID: 39361171 DOI: 10.1007/s00210-024-03482-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/21/2024] [Indexed: 03/19/2025]
Abstract
Globally, colorectal cancer (CRC) continues to rank among the leading causes of cancer-related death. Systemic toxicity, multidrug resistance, and nonspecific targeting often pose challenges to conventional therapy for CRC. Because it is a complex disease with a complex genetic and environmental pathophysiology, advanced therapeutic strategies are needed. Nanotechnology presents a potential solution that may maximize therapeutic efficacy while minimizing negative effects by enabling personalized delivery of anticancer drugs. This review focuses on recent developments in colorectal drug delivery systems based on nanotechnology. Numerous nanomaterials, including liposomes, dendrimers, micelles, exosomes, and gold nanoparticles, are developed and used. Distinctive characteristics of mentioned nanocarriers are discussed along with strategies that can be employed for enhancing the delivery of drugs to colorectal cancer cells. The review also quotes the most relevant preclinical and clinical studies that show how these nanomaterials improve drug solubility, stability, and targeted delivery while overcoming the shortcomings of conventional therapies. Nanotechnology has made CRC treatment very efficient and advanced, which has opened up new possibilities for targeted drug delivery. Preclinical and clinical studies have also proved that the use of nano-formulations in colon-specific delivery systems have significant results, indicating potential for better patient outcomes. Future research can be done in order to overcome the hurdles regarding biocompatibility, expansion, and regulatory challenges. Large-scale clinical trials and nanomaterial formulation optimization should be the main goals of future research to confirm the efficacy and safety of these novel treatments.
Collapse
Affiliation(s)
- Maria Shahzad
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Ayesha Amjad
- Faculty of Food Technology and Nutrition Sciences, Lahore University of Biological and Applied Sciences, Lahore, 54000, Pakistan
| | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Inaba Shujaat Qureshi
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan
| | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan
| | - Asad Saeed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Rabia Munir
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| |
Collapse
|
30
|
Durr-e-Shahwar, Zubair H, Raza MK, Khan Z, Mansour L, Ali A, Imran M. Investigation of GSTP1 and PTEN gene polymorphisms and their association with susceptibility to colorectal cancer. Radiol Oncol 2025; 59:110-120. [PMID: 39754630 PMCID: PMC11867568 DOI: 10.2478/raon-2025-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/25/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND This study investigates the association of single nucleotide polymorphism in glutathione S transferase P1 (rs1695 and rs1138272) and phosphatase and TENsin homolog (rs701848 and rs2735343) with the risk of colorectal cancer (CRC). PATIENTS AND METHODS In this case-control study, 250 healthy controls and 200 CRC patients were enrolled. All subjects were divided into 3 groups: healthy control, patients, and overall (control + patients). Genotyping was performed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The demographic information, including age, gender, location, smoking status, cancer stage, and node involvement, were collected. RESULTS The allele frequencies of PTEN rs701848 in overall subjects were 0.78 for C and 0.22 for T. Similarly, in overall individuals, allele frequencies for PTEN rs2735343 were 0.65 and 0.35 for G and C alleles, respectively. The CC genotype or C allele of rs701848 and CG/GG genotype of rs2735343 were observed to be a risk factor for CRC. In overall individuals, a significant (p ≤ 0.05)) association was observed between rs701848 and rs2735343 polymorphisms CRC. Allele frequencies for GSTP1 rs1695 were 0.68 and 0.32 for the A and G alleles, respectively. Allele frequencies for GSTP1 rs1138272 were 0.68 and 0.32 for C and T alleles, respectively. However, a significant (p < 0.05) association was found in males for rs1695, while a non-significant difference was observed for the distribution of any genotypes or alleles at GSTP1 (rs1138272). CONCLUSIONS Both SNPs of PTEN rs701848 and rs2735343 polymorphisms were significantly associated with CRC. However, in GSTP1, rs1695 was significantly associated with CRC risk in males, and rs1138272 showed a non-significant association with colorectal cancer risk.
Collapse
Affiliation(s)
- Durr-e-Shahwar
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| | - Hina Zubair
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| | - Muhammad Kashif Raza
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
- Department of Chemistry, Shaheed Benazir Bhutto University Sheringal Dir upper, Sringeri, Pakistan
| | - Zahid Khan
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| | - Lamjed Mansour
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Aktar Ali
- Biological Screening Core, Warren Family Center for Drug Discovery, University of Notre Dame, Notre Dame, United States
| | - Muhammad Imran
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| |
Collapse
|
31
|
Jairoun AA, Al-Hemyari SS, Shahwan M, Zyoud SH, Saleh Jaber AA. Community pharmacist-led point-of-care colorectal cancer screening program: Early detection of colorectal cancer in high-risk patients. Res Social Adm Pharm 2025; 21:185-192. [PMID: 39694778 DOI: 10.1016/j.sapharm.2024.12.006] [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: 11/27/2023] [Revised: 08/08/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND The prevalence of colorectal cancer (CRC) is on the rise among the younger population, with an anticipated increase in new cases for individuals aged 20-49 years by 2030. The accessibility of community pharmacists and their strong community connections present unique opportunities to enhance patient engagement in a population-based CRC screening program. OBJECTIVES This study seeks to assess the effectiveness of a community pharmacist-led point-of-care CRC screening program utilizing fecal immunochemical test (FIT) kits to identify CRC prevalence in high-risk individuals. METHODS AND MATERIALS Over the course of a 10-month prospective intervention conducted in UAE community pharmacies, we evaluated the impact of a pharmacist-led point-of-care colorectal cancer screening program. Six pharmacies were selected based on their services and capabilities. Eligible participants were those identified during medication reviews as exhibiting colorectal cancer risk factors. Pharmacists provided communication materials, distributed FIT kits, and implemented reminders. Participants collected samples for hemoglobin analysis, which served as an indicator of colorectal bleeding. Collected data encompassed demographics, lifestyle, and health-related characteristics. Pharmacists performed medication reviews and offered recommendations. RESULTS A total of four hundred and one recruited int the study. The mean age of study cohort at baseline was 66.6 ± 11.3 years. In our study with 401 participants, 36.4 % had undiagnosed colorectal cancer (CRC). Univariate logistic regression identified older age, a history of Type 2 diabetes mellitus (DM), and inflammatory bowel disease (IBD) as significant factors associated with increased CRC prevalence, while aspirin users exhibited a lower likelihood of CRC. In the multivariate regression model, the history of Type 2 DM and IBD remained significant predictors for heightened CRC risk. CONCLUSION This study strengthens the plausibility of cause-and-effect relationships between colorectal cancer and demographic variables using epidemiological evidence. The significant relationships found between prevalence of CRC and age, type 2 diabetes, IBD and aspirin use support the effectiveness of using FIT kits in community pharmacist-led point-of-care CRC screening program to identify high-risk individuals. The finding highlights the significance of improving efforts on colorectal cancer prevention and control.
Collapse
Affiliation(s)
- Ammar Abdulrahman Jairoun
- Health and Safety Department, Dubai Municipality, Dubai, United Arab Emirates; Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences, Universiti Sains Malaysia (USM), Pulau Pinang, 11500, Malaysia.
| | - Sabaa Saleh Al-Hemyari
- Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences, Universiti Sains Malaysia (USM), Pulau Pinang, 11500, Malaysia; Pharmacy Department, Emirates Health Services, Dubai, United Arab Emirates.
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates; Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, 346, United Arab Emirates.
| | - Samer H Zyoud
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates; Department of Mathematics and Sciences, Ajman University, P.O. Box 346, Ajman, United Arab Emirates.
| | - Ammar Ali Saleh Jaber
- Department of Clinical Pharmacy & Pharmacotherapeutics, Dubai Pharmacy College for Girls, AlMuhaisanah 1, Al mizhar, Dubai, United Arab Emirates.
| |
Collapse
|
32
|
Vaghasiya MD, Sain A, Mendapara JV, Khamrai D, Naskar D, Kumari P. Thiazepine-Based Hybrids as Promising Anti-Colon Cancer Agents: Design, Synthesis, Computational and In Vitro Screening. Chem Biodivers 2025; 22:e202401550. [PMID: 39413279 DOI: 10.1002/cbdv.202401550] [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: 06/26/2024] [Revised: 09/20/2024] [Accepted: 10/15/2024] [Indexed: 10/18/2024]
Abstract
Novel thiazepine-based hybrids (9 a-d) were designed and synthesized to create lead molecules with exceptional anti-colon cancer efficacy. Analytical methods, including IR, NMR, and HR-MS, characterized the synthesized compounds. The in vitro colorectal study was carried out to compare the biological activity of newly developed compounds with the computational data. The tested compounds induced cytotoxicity in HT-29 cells for both 24 h and 48 h in a dose-dependent manner. However, compound 9 a induced cytotoxicity at much higher concentrations compared to the rest of the compounds. 9 b and 9 c caused 50 % cell death (compared to the untreated cells) at a dose of ~50 μM and 40 μM in case of 24-hour exposure, respectively. On the contrary, for 48 h exposure, both 9 b and 9 c induced 50 % cell death concerning untreated cells at a dose of around ~20 μM, whereas 9 d exhibited 50 % cell death at 5 μM in the case of 48 h exposure. In silico ADMET was also carried out to understand the pharmacokinetics and safety profiles of the drug candidates. We found some of the critical targets of these compounds, which eventually will be integral to exploring the mechanistic actions of these compounds in colon cancer.
Collapse
Affiliation(s)
- Mahesh D Vaghasiya
- Department of Chemistry, S.V. National Institute of Technology, Surat, Gujarat, 395007, India
| | - Arindam Sain
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, Nadia, West Bengal, 741249, India
| | - Jigarkumar V Mendapara
- Department of Chemistry, S.V. National Institute of Technology, Surat, Gujarat, 395007, India
| | - Dipshikha Khamrai
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, Nadia, West Bengal, 741249, India
| | - Debdut Naskar
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, Nadia, West Bengal, 741249, India
| | - Premlata Kumari
- Department of Chemistry, S.V. National Institute of Technology, Surat, Gujarat, 395007, India
| |
Collapse
|
33
|
Takechi F, Kawasoe Y, Tateno S, Ebata R, Hamada H, Niwa K. Risk of malignancy in adult patients with congenital heart disease: a clinical practice review. Cardiovasc Diagn Ther 2025; 15:195-206. [PMID: 40115083 PMCID: PMC11921439 DOI: 10.21037/cdt-24-388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/20/2024] [Indexed: 03/23/2025]
Abstract
This paper presents case studies of adult patients with congenital heart disease (CHD) who developed cancer and also discusses relevant epidemiological studies, risk factors, and reports on early detection. Herein, we describe three cases: a 63-year-old man with an atrial septal defect and multiple myeloma; a 48-year-old man with tetralogy of Fallot and colorectal cancer; and a 25-year-old man with Fontan circulation and hepatocellular carcinoma (HCC). Previous studies have found that the incidence of cancer in adult patients with CHD is higher than that in the general population. The management of adult patients with CHD complicated by cancer requires careful attention because cancer treatment alone can affect the survival prognosis and quality of life, as well as the pathophysiology and treatment of underlying heart disease. Apart from known risk factors in the non-CHD population, specific risk factors have been reported, such as genetic abnormalities, low-dose ionizing radiation exposure, early thymectomy, Fontan-associated liver disease, and hypoxia. Encouraging patients to participate in cancer screening and avoid known risk factors is essential in daily practice for the early diagnosis and prevention of cancer. It is also important to be vigilant for initial signs that are indicative of cancer as well as avoidable risk factors.
Collapse
Affiliation(s)
- Fumie Takechi
- Department of Pediatrics, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Section of Adult Congenital Heart Disease, Chiba Cerebral and Cardiovascular Center, Chiba, Japan
| | - Yasutaka Kawasoe
- Section of Adult Congenital Heart Disease, Chiba Cerebral and Cardiovascular Center, Chiba, Japan
| | - Shigeru Tateno
- Section of Adult Congenital Heart Disease, Chiba Cerebral and Cardiovascular Center, Chiba, Japan
| | - Ryota Ebata
- Department of Pediatrics, Chiba Kaihin Municipal Hospital, Chiba, Japan
| | - Hiromichi Hamada
- Department of Pediatrics, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Koichiro Niwa
- Department of Cardiology, Cardiovascular Center, St. Luke's International Hospital, Tokyo, Japan
| |
Collapse
|
34
|
Ponvilawan B, Sakornsakolpat P, Pongpaibul A, Roothumnong E, Akewanlop C, Pithukpakorn M, Korphaisarn K. Comprehensive genomic analysis in sporadic early-onset colorectal adenocarcinoma patients. BMC Cancer 2025; 25:349. [PMID: 40001005 PMCID: PMC11863557 DOI: 10.1186/s12885-025-13745-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND The incidence of colorectal cancer (CRC) in young adults has increased worldwide. Our study aimed to evaluate genomic alterations in early-onset (aged 15-39 years) sporadic CRC. METHODS Formalin-fixed, paraffin-embedded tissue samples from 90 patients with histologically confirmed colorectal adenocarcinoma with proficient mismatch repair status from Siriraj Hospital (Bangkok, Thailand) were extracted. Patients with clinically suspected familial adenomatous polyposis were excluded. A 517-gene mutational analysis was performed by next-generation sequencing using the Oncomine Comprehensive Assay Plus kit. The previously reported molecular data in adult-onset CRC from our group were used as a comparator group. RESULTS The five most frequently mutated genes were APC (66%), TP53 (51%), KRAS (47%), ARID1A (31%), and KMT2B (31%). When compare with adult-onset, NOTCH1 (11.1% vs. 1.9%), FBXW7 (23.3% vs. 14.8%), PIK3CA (20% vs. 12.1%), and FGFR3 (8.9% vs. 3.7%) mutations were more prevalent in early-onset. No differences were observed in other common mutations, such as TP53, EGFR, KRAS, NRAS and BRAF mutations. An increased prevalence in KRAS codon 12 mutations was also observed in early-onset patients compared to the adult-onset group (38.9% vs. 29.6%). CONCLUSIONS Overall, the genomic landscape between early- and adult-onset CRC appears similar. However, our study revealed the enrichment of NOTCH1, FBXW7, PIK3CA, and FGFR3 along with KRAS G12 mutations, were more frequent in early-onset compared to adult-onset cases. Further studies with a larger cohort of patients on the comprehensive analysis of genetic/epigenetic signatures are required.
Collapse
Affiliation(s)
- Ben Ponvilawan
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Phuwanat Sakornsakolpat
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok Noi, Bangkok, 10700, Thailand
| | - Ananya Pongpaibul
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ekkapong Roothumnong
- Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Medical Genetics, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Charuwan Akewanlop
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok Noi, Bangkok, 10700, Thailand
| | - Manop Pithukpakorn
- Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Medical Genetics, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Krittiya Korphaisarn
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok Noi, Bangkok, 10700, Thailand.
| |
Collapse
|
35
|
Haynes J, Manogaran P. Mechanisms and Strategies to Overcome Drug Resistance in Colorectal Cancer. Int J Mol Sci 2025; 26:1988. [PMID: 40076613 PMCID: PMC11901061 DOI: 10.3390/ijms26051988] [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: 01/10/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related mortality worldwide, with a significant impact on public health. Current treatment options include surgery, chemotherapy, radiotherapy, molecular-targeted therapy, and immunotherapy. Despite advancements in these therapeutic modalities, resistance remains a significant challenge, often leading to treatment failure, poor progression-free survival, and cancer recurrence. Mechanisms of resistance in CRC are multifaceted, involving genetic mutations, epigenetic alterations, tumor heterogeneity, and the tumor microenvironment. Understanding these mechanisms at the molecular level is crucial for identifying novel therapeutic targets and developing strategies to overcome resistance. This review provides an overview of the diverse mechanisms driving drug resistance in sporadic CRC and discusses strategies currently under investigation to counteract this resistance. Several promising strategies are being explored, including targeting drug transport, key signaling pathways, DNA damage response, cell death pathways, epigenetic modifications, cancer stem cells, and the tumor microenvironment. The integration of emerging therapeutic approaches that target resistance mechanisms aims to enhance the efficacy of current CRC treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Jennifer Haynes
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Drive, Huntington, WV 25701, USA;
| | | |
Collapse
|
36
|
Heydarian R, Divsalar A, Kouchesfehani HM, Rasouli M. Folic acid-targeted β-lactoglobulin nanocarriers for enhanced delivery of 5-fluorouracil and sodium butyrate in colorectal cancer treatment. Int J Pharm 2025; 671:125262. [PMID: 39870256 DOI: 10.1016/j.ijpharm.2025.125262] [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: 09/24/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Colorectal cancer (CRC) remains a significant public health concern, emphasizing the need for innovative therapeutic strategies to improve patient outcomes. This study aimed to develop a highly efficient nanocarrier for targeted drug delivery, enhancing drug efficacy while minimizing concentrations and limiting adverse effects. We synthesized protein-based β-lactoglobulin (βlg) nanoparticles (NPs), loaded with 5-fluorouracil (5-FU) and sodium butyrate (NaB), and further functionalized with folic acid (FA) for specific targeting of folate receptor-positive CRC cells. The βlg-5-FU-NaB-FA nanoplatforms exhibited a well-defined size of 208 nm with a narrow size distribution (PDI ≈ 0.5). Zeta potential measurements showed a value of -11.4 mV, indicating stability and suitability for drug delivery. Scanning electron microscopy (SEM) and atomic force microscopy (AFM) confirmed the nanocarrier's spherical morphology and efficient distribution. Drug release profiles demonstrated that the NPs released more drugs at neutral to alkaline pH levels, attributed to pectin's ionization properties. The efficacy of the prepared βlg-5-FU-NaB-FA nanoplatforms was investigated on HCT116 and Caco2 CRC cells, along with the normal cell line CRL-1831. The βlg-5-FU-NaB-FA nanoplatforms exhibited remarkable cytotoxicity against both HCT116 and Caco2 CRC cells compared to free drugs, highlighting the efficacy of targeted delivery in folate receptor-positive cells. These NPs induce cell apoptosis and cell cycle arrest more effectively than free drugs, demonstrating their potential for targeted cancer therapy. Furthermore, a decrease in the expression of crucial genes involved in the Wnt signaling pathway was observed, which offers a valuable understanding of their underlying mechanism. Collectively, our results suggest that the FA-targeted βlg nanocarriers represent a promising platform for the efficient and targeted delivery of 5-FU and NaB in folate receptor-positive CRC. This novel nanocarrier holds the potential to enhance therapeutic outcomes while minimizing side effects, providing a new avenue for the treatment of CRC.
Collapse
Affiliation(s)
- Ronak Heydarian
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Adeleh Divsalar
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | | | - Milad Rasouli
- Department of Physics, Kharazmi University, Tehran, Iran; Endocrinology and Metabolism Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Ghabra S, Chang D, Sugarbaker PH. Preoperative tumor marker elevations in colorectal cancer patients with peritoneal metastases should be used to help select patients for cytoreductive surgery. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109720. [PMID: 40023022 DOI: 10.1016/j.ejso.2025.109720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/04/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Tumor markers are used routinely for surveillance in patients treated for colorectal cancer (CRC). However, the prognostic implications of elevated preoperative tumor markers in patients treated for CRC and peritoneal metastases (PM) has not been well defined. The utility of carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 (CA19-9) and cancer antigen 125 (CA125) to predict outcome for these patients is reported. METHODS Clinical and histologic features plus preoperative tumor markers were recorded within 1 week prior to CRS. Impact on overall survival of these factors was analyzed by univariate and multivariate analysis. RESULTS Two hundred and four patients were in our database and 140 patients (75.3 %) had at least a single elevated preoperative tumor marker. In an analysis of clinical and histologic parameters preoperatively, a poorly differentiated tumor, signet ring morphology, a peritoneal cancer index (PCI) of ≥16 and an incomplete cytoreduction had a negative impact on median survival. In a multivariate analysis of clinical and histologic features together with tumor markers, an elevated CA19-9 and CA125 was independently associated with reduced overall survival (HR 2.7, p < 0.0001 and HR 2.2, p = 0.005), respectively. Quantitative assessment of CEA (HR 0.5, p = 0.0094) and CA19-9 (HR 4.9, p < 0.001) greater than x10 ULN showed reduced survival. CONCLUSION Preoperative assessment of symptoms and histopathology, PCI and a complete CRS combined with tumor markers CEA, CA19-9 and CA125 are independent prognostic indicators for selection by the multidisciplinary team of CRC PM patients for CRS and HIPEC. All three tumor markers are needed for a meaningful assessment.
Collapse
Affiliation(s)
- Shadin Ghabra
- Department of Surgery, MedStar Washington Hospital Center, Washington, DC, USA
| | | | - Paul H Sugarbaker
- Department of Surgery, MedStar Washington Hospital Center, Washington, DC, USA.
| |
Collapse
|
38
|
Wu H, Zhang W, Chang J, Wu J, Zhang X, Jia F, Li L, Liu M, Zhu J. Comprehensive analysis of mitochondrial-related gene signature for prognosis, tumor immune microenvironment evaluation, and candidate drug development in colon cancer. Sci Rep 2025; 15:6173. [PMID: 39979377 PMCID: PMC11842742 DOI: 10.1038/s41598-024-85035-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 12/30/2024] [Indexed: 02/22/2025] Open
Abstract
Colon adenocarcinoma (COAD), a common digestive system malignancy, involves crucial alterations in mitochondria-related genes influencing tumor growth, metastasis, and immune evasion. Despite limited studies on prognostic models for these genes in COAD, we established a mitochondrial-related risk prognostic model, including nine genes based on available TCGA and MitoCarta 3.0 databases, and validated its predictive power. We investigated the tumor microenvironment (TME), immune cell infiltration, complex cell communication, tumor mutation burden, and drug sensitivity of COAD patients using R language, CellChat, and additional bioinformatic tools from single-cell and bulk-tissue sequencing data. The risk model revealed significant differences in immune cell infiltration between high-risk and low-risk groups, with the strongest correlation found between tissue stem cells and macrophages in COAD. The risk score exhibited a robust correlation with TME signature genes and immune checkpoint molecules. Integrating the risk score with the immune score, microsatellite status, or TMB through TIDE analysis enhanced the accuracy of predicting immunotherapy benefits. Predicted drug efficacy offered options for both high- and low-risk group patients. Our study established a novel mitochondrial-related nine-gene prognostic signature, providing insights for prognostic assessment and clinical decision-making in COAD patients.
Collapse
Affiliation(s)
- Hao Wu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Wentao Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jingjia Chang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jin Wu
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Xintong Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Fengfeng Jia
- Taiyuan Technology Transfer Promotion Center, Taiyuan, 030006, China
| | - Li Li
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Ming Liu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China.
| | - Jianjun Zhu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
39
|
Haq MU, Noureldin K, Pritchard DM, Myint AS, Duckworth CA, Than NW, Hughes DM, Ahmed S, Javed MA. Long-Term Outcomes of Patients with Poor Prognostic Factors Following Transanal Endoscopic Microsurgery (TEMS) for Early Rectal Cancer. Biomedicines 2025; 13:521. [PMID: 40002934 PMCID: PMC11853461 DOI: 10.3390/biomedicines13020521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Transanal endoscopic microsurgery (TEMS) is an organ-preserving approach for treatment of early rectal cancer (ERC). However, adverse histopathological features identified post-TEMS often necessitate adjuvant therapy. This study aims to compare the long-term oncological outcomes of patients who underwent TEMS and were offered adjuvant treatments with total mesorectal excision (TME), chemoradiotherapy (CRT), radiotherapy (RT), active surveillance, or dose escalation with contact X-ray brachytherapy (CXB). Methods: This study included patients treated with TEMS for ERC between September 2012 and December 2022, with follow-up until December 2023. Patients with adverse histopathological features (extra-mural venous invasion, lympho-vascular invasion, R1 margins, tumour budding) were assigned to adjuvant treatments. Inverse probability of treatment weighting (IPTW) was applied to mitigate selection bias. Results: Of the 117 patients, 24 underwent TME, 17 received CRT, 25 received RT, 14 underwent active surveillance, and 37 patients received CXB boost along with CRT. The median follow-up was 60 months (IQR 52-73). During this time, 29 patients developed recurrence, and 15 died. The 5-year overall survival (OS) was 78.6%, and disease-free survival (DFS) was 70.9%. Compared to CXB, the mortality risk for CRT (HR = 0.81; 95% CI: 0.20-3.28; p = 0.77) and TME (HR = 3.68; 95% CI: 0.46-29.79; p = 0.22) was not significantly different. However, TME was associated with a significantly higher recurrence risk compared to CXB (HR = 7.57; 95% CI: 1.23-46.84; p = 0.029). Conclusions: An organ-preserving strategy with CRT or CRT combined with a CXB boost may offer comparable long-term outcomes and reduced recurrence risks for patients undergoing TEMS for ERC with poor prognostic features. Further research with larger cohorts is needed to validate these results.
Collapse
Affiliation(s)
- Muneeb Ul Haq
- Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Liverpool L69 7BE, UK; (D.M.P.); (A.S.M.); (C.A.D.); (N.W.T.); (M.A.J.)
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool L7 8YA, UK
| | - Khaled Noureldin
- Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8YE, UK (D.M.H.); (S.A.)
| | - David Mark Pritchard
- Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Liverpool L69 7BE, UK; (D.M.P.); (A.S.M.); (C.A.D.); (N.W.T.); (M.A.J.)
- Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8YE, UK (D.M.H.); (S.A.)
| | - Arthur Sun Myint
- Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Liverpool L69 7BE, UK; (D.M.P.); (A.S.M.); (C.A.D.); (N.W.T.); (M.A.J.)
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool L7 8YA, UK
| | - Carrie A. Duckworth
- Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Liverpool L69 7BE, UK; (D.M.P.); (A.S.M.); (C.A.D.); (N.W.T.); (M.A.J.)
- Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8YE, UK (D.M.H.); (S.A.)
| | - Ngu Wah Than
- Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Liverpool L69 7BE, UK; (D.M.P.); (A.S.M.); (C.A.D.); (N.W.T.); (M.A.J.)
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool L7 8YA, UK
| | - David M. Hughes
- Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8YE, UK (D.M.H.); (S.A.)
- Department of Health Data Science, Institute of Population Health, The University of Liverpool, Liverpool L69 3GF, UK
| | - Shakil Ahmed
- Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8YE, UK (D.M.H.); (S.A.)
| | - Muhammad Ahsan Javed
- Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Liverpool L69 7BE, UK; (D.M.P.); (A.S.M.); (C.A.D.); (N.W.T.); (M.A.J.)
- Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8YE, UK (D.M.H.); (S.A.)
| |
Collapse
|
40
|
Moniruzzaman M, Wong KY, Janjua TI, Martin JH, Begun J, Popat A. Cannabidiol Targets Colorectal Cancer Cells via Cannabinoid Receptor 2, Independent of Common Mutations. ACS Pharmacol Transl Sci 2025; 8:543-556. [PMID: 39974647 PMCID: PMC11833734 DOI: 10.1021/acsptsci.4c00644] [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: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 02/21/2025]
Abstract
Cannabidiol (CBD) is a non-neurotoxic, phytocannabinoid from cannabis with reported medicinal properties, including antiepileptic and anti-inflammatory activity. Several in vitro and in vivo studies have shown that CBD has antitumor potential against colorectal cancer (CRC), the third deadliest cancer in the world. However, as different mutations influence the antitumor effects and CBD can bind a variety of receptors, it is yet to be determined whether specific CRC mutations affect CBD's efficacy in treatment of CRC. To investigate this, we selected four CRC cell lines, including HCT116, HT-29, LS174T, and LS153, which harbor distinct mutations. Cells were treated with a range of concentrations of CBD to evaluate its cytotoxic effects and impact on cell proliferation, migration, and invasion by using a live-cell imaging system. IC50 values were then calculated for each parameter. The level of endoplasmic reticulum (ER) stress pathway markers was also measured using qRTPCR. The requirements for CB1 or CB2 receptor-medicated signaling were investigated using the selective inhibitors AM251 and SR144528, respectively. Our results demonstrate that CBD induces apoptosis and halts proliferation, migration, and invasion of CRC cell lines in a concentration-dependent manner. CBD showed potent antitumor effects in the tested cell lines with no obvious effect from different mutations such as KRAS, BRAF, APC, PTEN, etc. CBD also induced ER stress in CRC cells but not in healthy intestinal organoids. Cotreatment with SR144528 inhibited the effects of indicating involvement of CB2 receptor activation in the anticancer effects of CBD. Together, these results demonstrated that CBD could be effective for CRC regardless of the underlying mutation through CB2 receptor activation.
Collapse
Affiliation(s)
- Md Moniruzzaman
- School
of Pharmacy, The University of Queensland, Brisbane 4102, Australia
- Inflammatory
Bowel Diseases Group, Mater Research Institute—UQ at Translational
Research Institute, The University of Queensland, Brisbane 4102, Australia
- Faculty
of Medicine, The University of Queensland, Brisbane 4102, Australia
| | - Kuan Yau Wong
- Immunopathology
Group, Mater Research Institute—UQ at Translational Research
Institute, The University of Queensland, Brisbane 4102, Australia
| | | | - Jennifer H. Martin
- Clinical
Pharmacology, School of Medicine and Public Health, University of Newcastle, Hunter Medical Research Institute, Kookaburra Circuit, Newcastle 2308, Australia
| | - Jakob Begun
- Inflammatory
Bowel Diseases Group, Mater Research Institute—UQ at Translational
Research Institute, The University of Queensland, Brisbane 4102, Australia
- Faculty
of Medicine, The University of Queensland, Brisbane 4102, Australia
| | - Amirali Popat
- School
of Pharmacy, The University of Queensland, Brisbane 4102, Australia
| |
Collapse
|
41
|
Alkatheeri A, Salih S, Kamil N, Alnuaimi S, Abuzar M, Abdelrahman SS. Nano-Radiopharmaceuticals in Colon Cancer: Current Applications, Challenges, and Future Directions. Pharmaceuticals (Basel) 2025; 18:257. [PMID: 40006069 PMCID: PMC11859487 DOI: 10.3390/ph18020257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Colon cancer remains a significant global health challenge; however, the treatment outcome for colon patients can be improved through early detection and effective treatment. Nano-radiopharmaceuticals, combining nanotechnology with radiopharmaceuticals, are emerging as a revolutionary approach in both colon cancer diagnostic imaging and therapy, playing a significant role in the management of colon cancer patients. This review examines the use of nano-radiopharmaceuticals in the diagnosis and treatment of colon cancer, highlighting current applications, challenges, and future directions. Nanocarriers of radionuclides have shown potential in improving cancer treatment, including liposomes, microparticles, nanoparticles, micelles, dendrimers, and hydrogels, which are approved by the FDA. These nanocarriers can deliver targeted drugs into malignant cells without affecting normal cells, reducing side effects. Antibody-guided systemic radionuclide-targeted therapy has shown potential for treating cancer. Novel cancer nanomedicines, like Hensify and 32P BioSilicon, are under clinical development for targeted radiation delivery in percutaneous intratumoral injections. Although using nano-radiopharmaceuticals is a superior technique for diagnosing and treating colon cancer, there are limitations and challenges, such as the unintentional accumulation of nanoparticles in healthy tissues, which leads to toxicity due to biodistribution issues, as well as high manufacturing costs that limit their availability for patients. However, the future direction is moving toward providing more precise radiopharmaceuticals, which is crucial for enhancing the diagnosis and treatment of colon cancer and reducing production costs.
Collapse
Affiliation(s)
- Ajnas Alkatheeri
- Department of Radiography and Medical Imaging, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates;
| | - Suliman Salih
- Department of Radiography and Medical Imaging, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates;
- National Cancer Institute, University of Gezira, Wad Madani 2667, Sudan
| | - Noon Kamil
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates; (N.K.); (S.A.); (M.A.)
| | - Sara Alnuaimi
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates; (N.K.); (S.A.); (M.A.)
| | - Memona Abuzar
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates; (N.K.); (S.A.); (M.A.)
| | | |
Collapse
|
42
|
Chuang PK, Chang KF, Chang CH, Chen TY, Wu YJ, Lin HR, Wu CJ, Wu CC, Ho YC, Lin CC, Yuan CH, Wang CY, Lee YK, Chen TY. Comprehensive Bioinformatics Analysis of Glycosylation-Related Genes and Potential Therapeutic Targets in Colorectal Cancer. Int J Mol Sci 2025; 26:1648. [PMID: 40004112 PMCID: PMC11855181 DOI: 10.3390/ijms26041648] [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: 01/10/2025] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide, characterized by high incidence and poor survival rates. Glycosylation, a fundamental post-translational modification, influences protein stability, signaling, and tumor progression, with aberrations implicated in immune evasion and metastasis. This study investigates the role of glycosylation-related genes (Glycosylation-RGs) in CRC using machine learning and bioinformatics. Data from The Cancer Genome Atlas (TCGA) and the Molecular Signatures Database (MSigDB) were analyzed to identify 67 differentially expressed Glycosylation-RGs. These genes were used to classify CRC patients into two subgroups with distinct survival outcomes, highlighting their prognostic value. Weighted gene coexpression network analysis (WGCNA) revealed key modules associated with CRC traits, including pathways like glycan biosynthesis and PI3K-Akt signaling. A machine-learning-based prognostic model demonstrated strong predictive performance, stratifying patients into high- and low-risk groups with significant survival differences. Additionally, the model revealed correlations between risk scores and immune cell infiltration, providing insights into the tumor immune microenvironment. Drug sensitivity analysis identified potential therapeutic agents, including Trametinib, SCH772984, and Oxaliplatin, showing differential efficacy between risk groups. These findings enhance our understanding of glycosylation in CRC, identifying it as a critical factor in disease progression and a promising target for future therapeutic strategies.
Collapse
Affiliation(s)
- Po-Kai Chuang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (P.-K.C.); (C.-H.C.); (T.-Y.C.)
| | - Kai-Fu Chang
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan; (K.-F.C.); (C.-H.Y.)
| | - Chih-Hsuan Chang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (P.-K.C.); (C.-H.C.); (T.-Y.C.)
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan; (K.-F.C.); (C.-H.Y.)
| | - Ting-Yu Chen
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (P.-K.C.); (C.-H.C.); (T.-Y.C.)
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan; (K.-F.C.); (C.-H.Y.)
| | - Yueh-Jung Wu
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
| | - Hui-Ru Lin
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
- Nursing Department, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
| | - Chi-Jen Wu
- Nursing Department, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- College of Nursing, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Cheng-Chun Wu
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan; (C.-C.W.); (Y.-C.H.)
| | - Yu-Cheng Ho
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan; (C.-C.W.); (Y.-C.H.)
| | - Chih-Chun Lin
- Department of Physical Therapy, I-Shou University, Kaohsiung 824005, Taiwan;
| | - Chien-Han Yuan
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan; (K.-F.C.); (C.-H.Y.)
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
- Department of Otolaryngology, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
- Department of Otolaryngology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Chih-Yang Wang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yung-Kuo Lee
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan; (K.-F.C.); (C.-H.Y.)
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
- Division of Experimental Surgery Center, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| | - Tung-Yuan Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
| |
Collapse
|
43
|
Randisi F, Perletti G, Marras E, Gariboldi MB. Green Tea Components: In Vitro and In Vivo Evidence for Their Anticancer Potential in Colon Cancer. Cancers (Basel) 2025; 17:623. [PMID: 40002218 PMCID: PMC11853328 DOI: 10.3390/cancers17040623] [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: 01/12/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Green tea consumption has been implicated in various biological activities, with particular emphasis on its anticancer properties. The antineoplastic effects of green tea are primarily attributed to its rich polyphenol content, among which, epigallocatechin-3-gallate (EGCG) is recognized as the most bioactive and potent catechin, responsible for the majority of its anticancer activity. This review provides a detailed examination of the in vitro and in vivo effects of green tea components, focusing on their potential therapeutic implications in colorectal cancer. The molecular mechanisms of action and bioactive constituents of green tea are systematically discussed, alongside an evaluation of experimental evidence supporting their efficacy. Furthermore, insights into the relationship between green tea dietary intake and colorectal cancer risk are analyzed, with a particular emphasis on clinical data and findings from meta-analyses involving patients diagnosed with colon cancer. The aggregated evidence underscores the necessity for well-designed randomized controlled trials and longitudinal cohort studies to substantiate the role of green tea as a chemopreventive agent. Additionally, future investigations should prioritize determining the optimal dosages, the appropriate durations of consumption, and the potential modulatory effects of dietary or lifestyle factors on green tea's anticancer efficacy.
Collapse
Affiliation(s)
| | | | | | - Marzia Bruna Gariboldi
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy; (F.R.); (G.P.); (E.M.)
| |
Collapse
|
44
|
Ahmadinejad M, Parvizi A, Sheikhi S, Eghbal F, Navabian S, Chaboki F, Bahri MH, Bozorgmehr R, Bagherpour JZ, Ziaie S. Optimal timing of surgery after neoadjuvant chemoradiotherapy in rectal cancer: A retrospective analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109702. [PMID: 40009935 DOI: 10.1016/j.ejso.2025.109702] [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: 11/05/2024] [Revised: 01/12/2025] [Accepted: 02/12/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Determining the optimal interval between neoadjuvant chemoradiotherapy (NCRT) and surgery in patients with locally advanced rectal cancer (LARC) remains crucial for improving treatment outcomes. Extending the interval may increase rates of pathological complete response (pCR), potentially enhancing survival and reducing recurrence. METHODS This retrospective cohort study included 226 patients with LARC who underwent NCRT followed by surgery. ROC analysis was used to establish the optimal interval between NCRT and surgery for achieving pCR, and multivariate logistic regression assessed independent predictors of pCR. Spline regression further analyzed the relationship between surgery timing and the probability of pCR. RESULTS ROC analysis identified 10.5 weeks as the optimal interval, showing increased pCR rates within this period. Multivariate analysis confirmed that surgery interval (OR = 2.603, P = 0.045) significantly predicted pCR. Both ROC and spline regression indicated that a 9-11-week interval maximizes pCR probability. Notably, the comparison of postoperative complications between groups with surgery intervals ≤10 weeks and >10 weeks showed no statistically significant differences (P = 0.518). CONCLUSION An interval of 9-11 weeks between NCRT and surgery optimizes pCR rates without increasing postoperative risks. This timeframe may serve as a favorable window for surgical intervention to enhance outcomes in rectal cancer patients.
Collapse
Affiliation(s)
- Mojtaba Ahmadinejad
- Department of Surgery, School of Medicine, Alborz University of Medical Science, Karaj, Iran.
| | - Arash Parvizi
- Radiation Oncology, Independent Practice, Karaj, Iran.
| | - Saman Sheikhi
- Department of Surgery, School of Medicine, Alborz University of Medical Science, Karaj, Iran.
| | - Fatemeh Eghbal
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran.
| | - Susan Navabian
- General Practitioner, Independent Practice, Karaj, Iran.
| | - Faranak Chaboki
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran.
| | - Mohammad Hadi Bahri
- Department of Surgery, School of Medicine, Alborz University of Medical Science, Karaj, Iran.
| | - Ramin Bozorgmehr
- Department of Surgery, School of Medicine, Alborz University of Medical Science, Karaj, Iran.
| | | | - Shirin Ziaie
- Internal Medicine, Independent Practice, Tehran, Iran.
| |
Collapse
|
45
|
Tetrick MG, Emon MAB, Doha U, Marcellus M, Symanski J, Ramanathan V, Saif MTA, Murphy CJ. Decoupling chemical and mechanical signaling in colorectal cancer cell migration. Sci Rep 2025; 15:4952. [PMID: 39929899 PMCID: PMC11811049 DOI: 10.1038/s41598-025-89152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Colorectal cancer metastasis is governed by a variety of chemical and mechanical signaling that are largely influenced by cancer-associated fibroblasts (CAFs) in the tumor microenvironment. Here, we deconvolute the chemical from mechanical signaling in the case of the colon cancer cell line HCT-116 and CAFs. We examined three chemoattractants (CXCL12, TGF-β, and activin A) which allegedly are secreted by CAFs and induce HCT-116 cell migration. None of the chemoattractants tested resulted in enhanced migration of HCT-116 in a 2D transwell assay, at low cell density. Similarly, CAF-conditioned media also did not lead to enhanced HCT-116 migration, while CAFs co-cultured in the transwell assay did lead to increased HCT-116 migration. This result suggests that either high cell densities are required for chemotaxis, and/or a reciprocal two-way signaling network between CAFs and HCT-116 is necessary to induce chemotaxis. Surprisingly, we find that HCT-116 cells exhibit enhanced migration along the axis of mechanical stress in a 3D collagen matrix, at very high cell densities. This migration is independent of whether the strain is induced mechanically or by CAFs. By comparing purely mechanical and purely chemical migration to a 3D co-culture of CAFs and HCT-116 containing both chemical and mechanical cues, it is concluded that HCT-116 migration is dominated by mechanical signaling, while chemical cues are less influential.
Collapse
Affiliation(s)
- Maxwell G Tetrick
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Md Abul Bashar Emon
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Umnia Doha
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Marsophia Marcellus
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Joseph Symanski
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Valli Ramanathan
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - M Taher A Saif
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Catherine J Murphy
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
46
|
Sharma D, Arumugam S. Pharmacophore-based identification and in Silico characterization of microbial metabolites as potential modulators of Wnt signaling pathway in colorectal cancer therapy. Mol Divers 2025:10.1007/s11030-024-11103-4. [PMID: 39921842 DOI: 10.1007/s11030-024-11103-4] [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: 11/23/2024] [Accepted: 12/31/2024] [Indexed: 02/10/2025]
Abstract
Aberrant activation of the Wnt/β-catenin signaling pathway, primarily driven by APC mutation and AXIN degradation via Tankyrase, contributes significantly to colorectal cancer (CRC) progression and metastasis. The accumulation of β-catenin, resulting from the dysregulated ubiquitination, underscores the need for alternative therapeutic strategies targeting Tankyrase and β-catenin. This present study explores microbial metabolites as a source of novel anti-cancer agents, leveraging their unique bioactivity and structural diversity, often exhibiting superior target specificity and lower toxicity than synthetic drugs. Through a computational drug discovery pipeline, a large library of 27641 microbial metabolites was initially screened based on multiple drug-likeliness criteria, resulting in the selection of 2527 compounds. Among the screened compounds, an integrated computational workflow comprising molecular docking, molecular dynamic simulations (MDS), MM/PBSA analysis, and Principal component analysis (PCA) identified Terreustoxin I (T1) as a potential Tankyrase inhibitor. In contrast, compound 10- phenyl-[12]-cytochalasin Z16 (B1) demonstrated a strong binding affinity within the β-catenin active site. Under physiological conditions, these lead compounds were evaluated for conformational stability, binding efficacy, and dynamic behavior. Additionally, ADMET profiling, physiochemical properties, and bioactivity score predictions confirmed the identified compounds' pharmacokinetic suitability and reduced toxicity profile. In silico, cytotoxicity predictions showed significant activity against SW480 and HCT90 colorectal cell lines, with additional anti-neoplastic and anti-leukemic properties, strengthening their candidacy as effective anti-cancer agents. These findings provide a foundation for further experimental validation and development of novel CRC therapies with improved safety and efficacy potential.
Collapse
Affiliation(s)
- Divya Sharma
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Sivakumar Arumugam
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
47
|
Sun M, Monahan K, Moquet J, Barnard S. Ionizing Radiation May Induce Tumors Partly Through the Alteration or Regulation of Mismatch Repair Genes. Cancers (Basel) 2025; 17:564. [PMID: 40002162 PMCID: PMC11852753 DOI: 10.3390/cancers17040564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Ionizing radiation is mutagenic and carcinogenic, and it is reported to induce primary and secondary tumors with intestinal tumors being one of the most commonly observed. However, the pathological and molecular mechanism(s) underlying the radiation-associated tumorigenesis remain unclear. A link between radiation and somatic tumorigenesis partly through genetic, epigenetic alteration and/or regulation of mismatch repair (MMR) genes has been hypothesized for the first time within this review. Clinical observations and experimental findings provide significant support for this association including MMR mutations as well as altered MMR RNA and protein expressions that occurred post-exposure, although existing evidence in published literature is sparse in this niche area. Some speculative mechanisms are suggested with this review to inform future research. Further studies are needed to understand the roles of the MMR system in response to radiation and to test this possible connection which could potentially provide useful and urgently needed information for clinical guidance.
Collapse
Affiliation(s)
- Mingzhu Sun
- UK Health Security Agency (UKHSA), Cytogenetics Group, Radiation Effects Department, Radiation, Chemical, Climate and Environmental Hazards Directorate, Chilton, Didcot OX11 0RQ, UK
| | - Kevin Monahan
- Lynch Syndrome Clinic, Centre for Familial Intestinal Cancer, St Mark’s Hospital, London North West University Healthcare NHS Trust, Watford Road, Harrow HA1 3UJ, UK
- Department of Surgery and Cancer, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Jayne Moquet
- UK Health Security Agency (UKHSA), Cytogenetics Group, Radiation Effects Department, Radiation, Chemical, Climate and Environmental Hazards Directorate, Chilton, Didcot OX11 0RQ, UK
| | - Stephen Barnard
- UK Health Security Agency (UKHSA), Cytogenetics Group, Radiation Effects Department, Radiation, Chemical, Climate and Environmental Hazards Directorate, Chilton, Didcot OX11 0RQ, UK
| |
Collapse
|
48
|
Tsukanov VV, Vasyutin AV, Tonkikh JL. Risk factors, prevention and screening of colorectal cancer: A rising problem. World J Gastroenterol 2025; 31:98629. [PMID: 39926213 PMCID: PMC11718609 DOI: 10.3748/wjg.v31.i5.98629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/06/2024] [Accepted: 12/04/2024] [Indexed: 12/30/2024] Open
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the second leading cause of cancer death worldwide. The leading risk factors for CRC include male gender, age over 50, family history, obesity, tobacco smoking, alcohol consumption, and unhealthy diet. CRC screening methods vary considerably between countries and depend on incidence, economic resources and healthcare structure. Important aspects of screening include adherence, which can vary significantly across ethnic and socioeconomic groups. Basic concepts of CRC screening include pre-stratification of patients by identifying risk factors and then using fecal immunochemical test or guaiac-based fecal occult blood test and/or colonoscopy or radiologic imaging techniques. Technological capabilities for CRC screening are rapidly evolving and include stool DNA test, liquid biopsy, virtual colonography, and the use of artificial intelligence. A CRC prevention strategy should be comprehensive and include active patient education along with targeted implementation of screening.
Collapse
Affiliation(s)
- Vladislav V Tsukanov
- Clinical Department of The Digestive System Pathology of Adults and Children, Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, Krasnoyarsk 660022, Russia
| | - Alexander V Vasyutin
- Clinical Department of The Digestive System Pathology of Adults and Children, Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, Krasnoyarsk 660022, Russia
| | - Julia L Tonkikh
- Clinical Department of The Digestive System Pathology of Adults and Children, Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, Krasnoyarsk 660022, Russia
| |
Collapse
|
49
|
Brocco D, Simeone P, Marino PD, De Bellis D, D’Ascanio F, Colasante G, Grassadonia A, De Tursi M, Florio R, Di Ianni M, Cama A, Tinari N, Lanuti P. Low Phosphatidylserine+ Cells Within the CD34+/CD45dim/CD117(c-kit)+ Subpopulation Are Associated with Poor Outcomes in Metastatic Colorectal Cancer. Cancers (Basel) 2025; 17:499. [PMID: 39941866 PMCID: PMC11816280 DOI: 10.3390/cancers17030499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Colorectal cancer is among the most prevalent causes of tumor-related deaths worldwide. Antiangiogenic therapy represents a cornerstone of metastatic CRC treatment, and biomarkers are advocated for the optimization of this therapeutic strategy. METHODS In this observational prospective study, we employed an optimized flow cytometry protocol to investigate the prognostic and predictive potential of blood circulating endothelial cells (CECs), circulating endothelial progenitor cells (CEPCs), and related subsets in a cohort of patients with metastatic colorectal cancer (n = 40). RESULTS Computational FC analysis revealed a differential enrichment of blood cell clusters with a CD34+/CD45dim/CD117(c-kit)+ phenotype between responders and non-responders both to antiangiogenic and non-antiangiogenic treatments. Intriguingly, our results show that a high percentage of annexin V-negative cells in a putative circulating progenitor population with a CD34+/CD45dim/CD117+ phenotype was correlated with a reduced response to systemic anticancer treatments (p = 0.015) and worse overall survival (log-rank p = 0.03). In addition, we observed increased blood concentrations of CD34+/CD45dim/CD117+/annexin V- cells in patients with a higher number of metastatic sites (p = 0.03). CONCLUSIONS Overall, these findings hold promise for the identification of novel circulating biomarkers to develop more personalized treatment approaches in patients with metastatic colorectal cancer.
Collapse
Affiliation(s)
- Davide Brocco
- Department of Medical, Oral & Biotechnological Sciences, University "G. D’Annunzio", 66100 Chieti, Italy;
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy; (P.S.); (D.D.B.); (F.D.); (G.C.); (P.L.)
| | - Pasquale Simeone
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy; (P.S.); (D.D.B.); (F.D.); (G.C.); (P.L.)
- Department of Medicine and Aging Sciences, University “G. D’Annunzio”, 66100 Chieti, Italy
| | - Pietro Di Marino
- Clinical Oncology Unit, S.S. Annunziata Hospital, 66100 Chieti, Italy
| | - Domenico De Bellis
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy; (P.S.); (D.D.B.); (F.D.); (G.C.); (P.L.)
- Department of Medicine and Aging Sciences, University “G. D’Annunzio”, 66100 Chieti, Italy
| | - Francesca D’Ascanio
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy; (P.S.); (D.D.B.); (F.D.); (G.C.); (P.L.)
- Department of Medicine and Aging Sciences, University “G. D’Annunzio”, 66100 Chieti, Italy
- Department of Humanities, Law and Economics, “Leonardo da Vinci” University, 66010 Torrevecchia Teatina, Italy
| | - Giulia Colasante
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy; (P.S.); (D.D.B.); (F.D.); (G.C.); (P.L.)
- Department of Medicine and Aging Sciences, University “G. D’Annunzio”, 66100 Chieti, Italy
| | - Antonino Grassadonia
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” 66100 Chieti, Italy; (A.G.); (M.D.T.)
| | - Michele De Tursi
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” 66100 Chieti, Italy; (A.G.); (M.D.T.)
| | - Rosalba Florio
- Department of Pharmacy, University “G. D’Annunzio”, 66100 Chieti, Italy;
| | - Mauro Di Ianni
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy; (P.S.); (D.D.B.); (F.D.); (G.C.); (P.L.)
- Department of Medicine and Aging Sciences, University “G. D’Annunzio”, 66100 Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, University “G. D’Annunzio”, 66100 Chieti, Italy;
| | - Nicola Tinari
- Department of Medical, Oral & Biotechnological Sciences, University "G. D’Annunzio", 66100 Chieti, Italy;
| | - Paola Lanuti
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy; (P.S.); (D.D.B.); (F.D.); (G.C.); (P.L.)
- Department of Medicine and Aging Sciences, University “G. D’Annunzio”, 66100 Chieti, Italy
| |
Collapse
|
50
|
Bhoomandla S, Chennuri BK, Sirisha S, Ganji S, Trivedi R, Karunasri A, Pandiri S. Design, Synthesis of Flurbiprofen Based 1,3,4-Oxadiazoles and Constrained Anticancer, Antioxidant Agents: In silico Docking Analysis. Chem Biodivers 2025; 22:e202401313. [PMID: 39365710 DOI: 10.1002/cbdv.202401313] [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: 05/25/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/06/2024]
Abstract
Flurbiprofen, a primary component of a nonsteroidal anti-inflammatory drug (NSAID) used to relieve symptoms of arthritis, and is a considerable interest in medicinal chemistry due to its demonstrated potential as an effective agent in various therapeutic applications. In consideration of the 1,3,4-oxadiazole therapeutic potential and anticancer activity, a new series of flurbiprofen scaffolds have been prepared through a straightforward reaction between 5-(1-(2-fluoro-[1,1'-biphenyl]-4-yl)ethyl)-1,3,4-oxadiazole-2-thiol (4) and various organic active 2-chloro-N-phenyl acetamides (5). The synthesized series (6a-6k) was characterized using a combination of spectroscopic techniques, including FT-IR, mass, 1H-NMR, and 13C NMR, followed by physical data. The cytotoxicity of the newly synthesized congeners was investigated against MCF-7 (human breast cancer cell line) and A-549 (human lung carcinoma epithelial) cell lines and anti-inflammatory activity as DPPH and H2O2 radical scavenging ability. In the series, analogues 6c, 6e, 6h, and 6k showed excellent inhibitory activity against MCF-7 cells in the range of IC50 values of 9.10-13.67 μg mL-1 compared to DXN (IC50=9.24 μg mL-1). In this series, analogues 6c, 6f, 6h, and 6j show remarkable H2O2 radical scavenging inhibition IC50 of 48.25±0.21, 47.33±0.15, 51.10±0.25, and 44.40±0.07 μM by using ascorbic acid as a standard, whose IC50 is 49.90±0.27 μM. According to the docking results, the most potent cytotoxic compounds have a stronger binding affinity with the Flurbiprofen complex (PDB: 1R9O) because of their interactions with residues such as Arg416(A), Trp103(A), Phe97(A), Gly279(A), Ile188(A), Glu283(A), Thr287(A), Val462(A), Phe459(A), Leu345(A), Ile417(A), and Cys418(A). Furthermore, in silico drug-likeness prediction analysis suggested that the majority of the synthesized compounds exhibit good oral bioavailability based on their Lipinski's Rule of Five and toxicity using ADME/Tox predictions.
Collapse
Affiliation(s)
- Srinu Bhoomandla
- Department of Chemistry, Geethanjali College of Engineering and Technology, Cheeryal, Keesara, Medchal, Telanagana, 501301, India
- Department of Chemistry, School of Science, GITAM (Deemed to be University), Hyderabad, Telangana, 502329, India
| | - Bharath Kumar Chennuri
- Department of Chemistry, BVRIT Hyderabad College of Engineering for Women, Bachupally, Hyderabad, Telangana, 500090, India
| | - Surapaneni Sirisha
- Department of Chemistry, Gitam School of Science, Gitam University (Deemed to be University), Bengaluru Campus, Karnataka, 561203, India
- Department of Chemistry-H & S, CMRTC, Kandlakoya, Hyderabad, Telangana, 501401, India
| | - Saidulu Ganji
- Department of Chemistry, Chaitanya Bharathi Institute of Technology (A), Hyderabad, Telangana, 500075, India
| | - Rashmi Trivedi
- Department of Chemistry, Nalla Narsimha Reddy Education Society's Group of Institutions, Hyderabad, Telangana, India
| | - Ananthoju Karunasri
- Department of Chemistry, Mallareddy College of Engineering, Maisammaguda, Telangana, 500100, India
| | - Sreedhar Pandiri
- Department of Chemistry, Geethanjali College of Engineering and Technology, Cheeryal, Keesara, Medchal, Telanagana, 501301, India
- Department of Chemistry, Osmania University, Tarnaka, Hyderabad, Telangana, 500007, India
| |
Collapse
|