1
|
Lin J, Yin Y, Cao J, Zou B, Han K, Chen Y, Li S, Huang C, Chen J, Lv Y, Xu S, Xie D, Wang F. Nudix Hydrolase 13 Impairs the Initiation of Colorectal Cancer by Inhibiting PKM1 ADP-Ribosylation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410058. [PMID: 39921866 PMCID: PMC11967829 DOI: 10.1002/advs.202410058] [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: 08/21/2024] [Revised: 01/06/2025] [Indexed: 02/10/2025]
Abstract
Metabolic dysregulation has been implicated as a key factor in colorectal cancer (CRC) initiation, however, the underlying driving forces and mechanisms remain poorly understood. Herein, transcriptome profiling of paired early-stage CRCs and adenomas identifies Nudix hydrolase 13 (NUDT13) as a critical suppressor. Elevated NUDT13 expression impedes the proliferation of CRC cells under hypoxic conditions and markedly inhibits CRC initiation by upregulating PKM1. Mechanistically, NUDT13 directly binds and stabilizes PKM1 protein by reducing its poly ADP-ribosylation (PARylation), which is catalyzed by PARP1 at E275/D281/E282/E285/D296, thereby inducing an oxidative phosphorylation (OXPHOS) phenotype in CRC cells. Moreover, spatiotemporal knockout of Nudt13 enhances intestinal tumorigenesis in mice, which can be significantly suppressed by PARP1 inhibitor Olaparib. Notably, residues E245/E248/E249 within the Nudix box motif of NUDT13 are essential for PKM1 PARylation, and a mimic peptide derived from this motif is sufficient to stabilize PKM1 protein and robustly inhibit CRC tumorigenesis. Collectively, this study reveals a previously unknown PARylation-dependent mechanism that regulates PKM1 protein stability and switches the metabolic pathway of CRC cells, providing a promising target for CRC treatment.
Collapse
Affiliation(s)
- Jinlong Lin
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
- Department of Thoracic SurgerySun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Yixin Yin
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
- Department of AnesthesiologySun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinghua Cao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Bingxu Zou
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Kai Han
- Department of Colorectal SurgerySun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Yufan Chen
- Department of EndoscopySun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Siyu Li
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Cijun Huang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Jiewei Chen
- Department of PathologySun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Yongrui Lv
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Shuidan Xu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Dan Xie
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
- Department of PathologySun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Fengwei Wang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
2
|
Sultan S, Shung DL, Kolb JM, Foroutan F, Hassan C, Kahi CJ, Liang PS, Levin TR, Siddique SM, Lebwohl B. AGA Living Clinical Practice Guideline on Computer-Aided Detection-Assisted Colonoscopy. Gastroenterology 2025; 168:691-700. [PMID: 40121061 DOI: 10.1053/j.gastro.2025.01.002] [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] [Indexed: 03/25/2025]
Abstract
BACKGROUND & AIMS This American Gastroenterological Association (AGA) guideline is intended to provide an overview of the evidence and support endoscopists and patients on the use of computer-aided detection (CADe) systems for the detection of colorectal polyps during colonoscopy. METHODS A multidisciplinary panel of content experts and guideline methodologists used the Grading of Recommendations Assessment, Development and Evaluation framework and relied on the following sources of evidence: (1) a systematic review examining the desirable and undesirable effects (ie, benefits and harms) of CADe-assisted colonoscopy, (2) a microsimulation study estimating the effects of CADe on longer-term patient-important outcomes, (3) a systematic search of evidence evaluating the values and preferences of patients undergoing colonoscopy, and (4) a systematic review of studies evaluating health care providers' trust in artificial intelligence technology in gastroenterology. RESULTS The panel reached the conclusion that no recommendation could be made for or against the use of CADe-assisted colonoscopy in light of very low certainty of evidence for the critical outcomes, desirable and undesirable (11 fewer colorectal cancers per 10,000 individuals and 2 fewer colorectal cancer deaths per 10,000 individuals), increased burden of more intensive surveillance colonoscopies (635 more per 10,000 individuals), and cost and resource implications. The panel acknowledged the 8% (95% CI, 6%-10%) increase in adenoma detection rate and 2% (95% CI, 0%-4%) increase in advanced adenoma and/or sessile serrated lesion detection rate. CONCLUSIONS This guideline highlights the close tradeoff between desirable and undesirable effects and the limitations in the current evidence to support a recommendation. The panel acknowledged the potential for CADe to continually improve as an iterative artificial intelligence application. Ongoing publications providing evidence for critical outcomes will help inform a future recommendation.
Collapse
Affiliation(s)
- Shahnaz Sultan
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, Minnesota; Minneapolis Veterans Affairs Healthcare System, Minneapolis, Minnesota
| | - Dennis L Shung
- Department of Medicine, Section of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut
| | - Jennifer M Kolb
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California; Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Farid Foroutan
- MAGIC Evidence Ecosystem Foundation, Oslo, Norway; Ted Rogers Centre for Heart Research, University Health Network, Toronto, Ontario, Canada
| | - Cesare Hassan
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Charles J Kahi
- Department of Gastroenterology, Indiana University Medical Center, Indianapolis, Indiana
| | - Peter S Liang
- Department of Medicine, Division of Gastroenterology and Hepatology, NYU Langone Health, New York, New York; Department of Medicine, Veterans Affairs New York Harbor Health Care System, New York, New York
| | - Theodore R Levin
- Division of Research, Kaiser Permanente Northern California, Pleasanton, California; Department of Gastroenterology, Kaiser Permanente Walnut Creek, Walnut Creek, California
| | - Shazia Mehmood Siddique
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania; Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Healthcare Improvement and Patient Safety, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Benjamin Lebwohl
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| |
Collapse
|
3
|
Meyer CE, Vukelic N, Mariadason JM, Kipp AP. Connecting concentrations of copper, selenium, and zinc with transcriptomic and proteomic data of well-characterized human colorectal cancer cell lines. J Trace Elem Med Biol 2025; 89:127638. [PMID: 40179449 DOI: 10.1016/j.jtemb.2025.127638] [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: 09/12/2024] [Revised: 02/18/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) incidence is associated with lower circulating selenium and zinc and elevated copper concentrations. Moreover, copper and selenium accumulate within tumor tissue, indicating a disturbed homeostasis of these essential trace elements in CRC. OBJECTIVE This study aimed to identify associations between CRC characteristics (based on genomic, transcriptomic and proteomic data) and trace element concentrations. METHODS The concentrations of copper, selenium, and zinc were measured in 83 human CRC cell lines and correlated with transcript and protein expression levels to identify trace element-related gene signatures. By using publicly available gene expression data from The Cancer Genome Atlas we investigated the association between those signatures with the survival probability of CRC patients. RESULTS The CRC cell lines differed in their copper (fold change 7.3), selenium (fold change 6), and zinc (fold change 2.6) concentrations. The concentrations were not associated with genetic or cellular characteristics, except for lower copper concentrations in KRAS mutant cells. Expression levels of known copper- and zinc-related proteins correlated significantly with the respective trace element concentrations, serving as a proxy for trace element concentrations in tumors, and with patient survival. This was not the case for selenium and selenoproteins. In addition, an unbiased approach identified novel high and low copper- and zinc-related gene expression signatures significantly associated with patient's outcome. CONCLUSION Herein we identify gene signatures associated with intracellular copper and zinc concentrations in CRC cell lines. Extrapolating these signatures to primary colorectal tumors revealed that they can inform outcome of CRC patients.
Collapse
Affiliation(s)
- Caroline E Meyer
- Department of Nutritional Physiology, Friedrich Schiller University Jena, Jena, Germany
| | - Natalia Vukelic
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia
| | | | - Anna P Kipp
- Department of Nutritional Physiology, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
4
|
Kahn CL, Petersen MM, Kleif J, Mansvelders MSE, Rasmussen M, Jørgensen LN, Vilandt J, Seidelin JB, Jaensch C, Bondeven P, Gotschalck KA, Løve US, Andersen B, Christensen IJ, LaPoint LC, Therkildsen C. Circulating Tumor DNA in Addition to Fecal Immunochemical Test in a Dual-Test Colorectal Cancer Screening Approach. Clin Colorectal Cancer 2025:S1533-0028(25)00028-3. [PMID: 40204621 DOI: 10.1016/j.clcc.2025.03.001] [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: 11/13/2024] [Revised: 02/03/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Early detection is paramount when reducing incidence and mortality of colorectal cancer (CRC). Current population-based screening programs primarily use fecal immunochemical test (FIT) to allocate individuals for colonoscopy although low specificity challenges colonoscopy capacities. We aimed to assess the potential of circulating tumor (ct)DNA markers for early CRC detection in a dual-test CRC screening approach among FIT positive individuals. METHODS Plasma samples from 774 FIT positive (≥100 ng Hemoglobin/mL) individuals from the Danish CRC screening program were analyzed for hypermethylated DNA in the genes Branched Chain Amino-acid Transaminase 1 (BCAT1), Ikaros-Family Zinc Finger transcription 1 (IKZF1), and Interferon Regulator Factor 4 (IRF4). Multivariate logistic regression models were generated adding the ctDNA markers and age to the FIT value. The dual-test approach was benchmarked to FIT at specific thresholds. RESULTS The dual-test approach improved CRC detection compared to the FIT alone (AUC of 87.2 [95% CI, 82.9-91.4] vs AUC of 72.5 [95% CI, 67.0-77.9]). This was also seen when adding advanced adenomas to the outcome resulting in AUCs of 71.8 [95% CI, 67.8-75.8] for the dual-test approach compared to 65.5 [95% CI, 61.3-69.7] for the FIT model alone. Benchmarking the dual-test approach at FIT cut-offs between 100 and 600 ng Hb/mL showed a potential for either reducing the colonoscopy requirement by up to 56% or increasing CRC detection by up to 28%. CONCLUSIONS As increasing FIT cutoff will decrease CRC detection rate, application of the ctDNA panel can increase the sensitivity and specificity in a dual-test approach among asymptomatic individuals.
Collapse
Affiliation(s)
| | - Mathias M Petersen
- Gastro Unit, Hvidovre Hospital, Hvidovre, Denmark; Institute for Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Kleif
- Gastro Unit, Hvidovre Hospital, Hvidovre, Denmark; Institute for Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Surgery, Nordsjællands Hospital, Hillerød, Denmark
| | | | - Morten Rasmussen
- Institute for Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Digestive Disease Center, Bispebjerg Hospital, Copenhagen, Denmark
| | - Lars N Jørgensen
- Institute for Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Digestive Disease Center, Bispebjerg Hospital, Copenhagen, Denmark
| | - Jesper Vilandt
- Department of Surgery, Nordsjællands Hospital, Hillerød, Denmark
| | - Jakob B Seidelin
- Gastro Unit, Section for Gastroenterology, Herlev Hospital, Herlev, Denmark
| | - Claudia Jaensch
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam UMC, AMC & VUMC, Amsterdam, The Netherlands
| | - Peter Bondeven
- Department of Surgery, Randers Hospital, Randers, Denmark
| | - Kåre A Gotschalck
- Department of Surgery, Horsens Hospital, Horsens, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Uffe S Løve
- Department of Surgery, Viborg Hospital, Viborg, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Berit Andersen
- Department of Public Health Programmes and University Research Clinic for Cancer Screening, Randers Regional Hospital, Randers, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | | | | |
Collapse
|
5
|
Silinskaite U, Valciukiene J, Jakubauskas M, Poskus T. The Immune Environment in Colorectal Adenoma: A Systematic Review. Biomedicines 2025; 13:699. [PMID: 40149674 PMCID: PMC11940254 DOI: 10.3390/biomedicines13030699] [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: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Research on colorectal adenoma is significantly less comprehensive compared to studies on colorectal carcinoma. Although colorectal adenoma is a precursor of the majority of sporadic colorectal cancers, not all adenomas develop into carcinomas. The complex interaction of immune responses in the premalignant tumor microenvironment might be a factor for that. Methods: In this systematic review, we aim to provide a thorough analysis of the current research examining the immune infiltration patterns in sporadic colorectal adenoma tissues in the context of immune cell-based, cytokine-based, and other immunological factor-related changes along the conventional adenoma-carcinoma sequence. The articles included in the review extend up to December 2024 in PubMed and Web of Science databases. Results: Most included studies have shown significant differences in immune cell counts, densities, and cytokine expression levels associated with premalignant colorectal lesions (and/or colorectal cancer). No consensus on the immune-related tendencies concerning CD4+T cells and CD8+T cells was reached. Decreasing expression of mDCs and plasma and naïve B cells were detected along the ACS. The increased density of tissue eosinophils in the adenoma tissue dramatically diminishes after the transition to carcinoma. As the adenoma progresses, the increasing expression of IL-1α, IL-4, IL-6, IL-8, IL-10, IL-17A, IL-21, IL-23, IL-33, and TGF-β and decreasing levels of IL-12A, IL-18, IFN-γ, and TNFα cytokines in the invasive carcinoma stage is being detected. The over-expression of COX-2, PD-1/PD-L1, CTLA-4, and ICOS/ICOSLG in the colorectal adenomatous and cancerous tissues was also observed. Conclusions: Further studies are needed for a better understanding of the whole picture of colorectal adenoma-associated immunity and its impact on precancerous lesion's potential to progress.
Collapse
|
6
|
Zengin Kurt B, Özmen Ö, Öztürk Civelek D, Şenol H, Sonmez F. Exploring anticancer properties of new triazole-linked benzenesulfonamide derivatives against colorectal carcinoma: Synthesis, cytotoxicity, and in silico insights. Bioorg Med Chem 2025; 119:118060. [PMID: 39793404 DOI: 10.1016/j.bmc.2025.118060] [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: 11/19/2024] [Revised: 12/10/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025]
Abstract
This study reports the design, synthesis, and characterization of a novel series of benzene sulfonamide-triazole hybrid derivatives, to evaluate their anticancer potential against colorectal cancer. The synthesized compounds were characterized using NMR and HRMS spectroscopic techniques. In vitro cytotoxicity assessments revealed that compounds 5g and 5j exhibited significant anticancer effects. 5g showed the highest potency in the DLD-1 cell line (IC50 = 11.84 µM), while 5j demonstrated robust activity in the HT-29 cell line (IC50 = 9.35 µM). Apoptotic analysis indicated that compound 5g effectively induced early and total apoptosis, surpassing the chemotherapeutic agent 5-fluorouracil (5-FU), highlighting its therapeutic potential. Molecular docking studies showed strong binding interactions with key proteins involved in colorectal cancer progression, such as TGFβ2 and VEGFR1. 5j displayed a high binding affinity for TGFβ2 (MM-GBSA ΔG = -92.52 kcal/mol) and 5g showed promising interactions with VEGFR1 (ΔG = -70.63 kcal/mol). Molecular dynamics simulations confirmed the stability of the ligand-protein complexes, indicating potential as targeted therapeutic agents. Compounds 5g and 5j demonstrate significant promise for further development in colorectal cancer treatment.
Collapse
Affiliation(s)
- Belma Zengin Kurt
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 34093 Istanbul, Turkiye.
| | - Özge Özmen
- Bezmialem Vakif University, Faculty of Pharmacy, 34093 Istanbul, Turkiye; Kartal Dr. Lutfi Kirdar City Hospital, 34865 Istanbul, Turkiye
| | - Dilek Öztürk Civelek
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmacology, 34093 Istanbul, Turkiye
| | - Halil Şenol
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 34093 Istanbul, Turkiye
| | - Fatih Sonmez
- Sakarya University of Applied Sciences, Pamukova Vocational School, 54055 Sakarya, Turkiye
| |
Collapse
|
7
|
Peng L, Zhang X, Zhu Y, Shi L, Ai K, Huang G, Ma W, Wei Z, Wang L, Ma Y, Wang L. T2WI and ADC radiomics combined with a nomogram based on clinicopathologic features to quantitatively predict microsatellite instability in colorectal cancer. Acad Radiol 2025; 32:1431-1450. [PMID: 39490321 DOI: 10.1016/j.acra.2024.10.002] [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: 05/14/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024]
Abstract
RATIONALE AND OBJECTIVES Microsatellite instability (MSI) stratification can guide the clinical management of patients with colorectal cancer (CRC). This study aimed to establish a radiomics model for predicting the MSI status of patients with CRC before treatment. MATERIALS AND METHODS This retrospective study was performed on 366 patients diagnosed with CRC who underwent preoperative magnetic resonance imaging (MRI) and immunohistochemical staining between February 2016 and September 2023. The participants were divided randomly into training and testing cohorts in a 7:3 ratio. The tumor volume of interest (VOI) was manually delineated on T2-weighted imaging (T2WI) and apparent diffusion coefficient (ADC) sequences using 3D Slicer software, and radiomics features were extracted. Feature selection was performed using the least absolute shrinkage and selection operator method. A radiomics nomogram was developed using multiple logistic regression, and the predictive performance of the models was evaluated and compared using receiver operating characteristic curves. The calibration curve, clinical decision curve analysis (DCA) and clinical impact curve (CIC) were used to evaluate the clinical application value of the model. RESULTS The radiomics normogram combined with history of chronic enteritis, tumor location, MR-reported inflammatory response, D2-40, carcinoembryonic antigen, tumor protein 53, and monocyte was an excellent predictive tool. The area under the curve for the training and testing cohorts were 0.927 and 0.984, respectively. The DCA and CIC demonstrated favorable clinical application and net benefit. CONCLUSIONS A radiomics nomogram based on T2WI and ADC sequences and clinicopathologic features can effectively and noninvasively predict the MSI status in CRC. This approach helps clinicians in stratifying CRC patients and making clinical decisions for personalized treatment.
Collapse
Affiliation(s)
- Leping Peng
- Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Xiuling Zhang
- Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Yuanhui Zhu
- Department of Radiology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Liuyan Shi
- Department of Radiology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Kai Ai
- Department of Clinical and Technical Support, Philips Healthcare, Xi'an 710065, China
| | - Gang Huang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Wenting Ma
- Department of Radiology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Zhaokun Wei
- Department of Radiology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Ling Wang
- Department of Pathology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Yaqiong Ma
- Department of Radiology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Lili Wang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou 730000, China.
| |
Collapse
|
8
|
Rong Y, Zhang G, Ye W, Qi L, Hao X, Li X, Zhang W, Chao Y, Gu S. Uncovering the Effects and Molecular Mechanisms of Shaoyao Decoction Against Colorectal Cancer Using Network Pharmacology Analysis Coupled With Experimental Validation and Gut Microbiota Analysis. Cancer Med 2025; 14:e70813. [PMID: 40119640 PMCID: PMC11928771 DOI: 10.1002/cam4.70813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Chronic gut inflammation and dysbiosis contribute significantly to colorectal cancer (CRC) development. Shaoyao decoction (SYD) is a well-established Chinese medicine prescription. Besides ameliorating CRC via anti-inflammatory effects, SYD modulates gut microbiota (GM) to improve inflammatory responses in ulcerative colitis (UC). However, whether and how SYD suppresses CRC by regulating GM remains largely unknown. METHODS SD rats were orally administered SYD for 7 days to obtain medicated serum. We utilized liquid chromatography-mass spectrometry (LC-MS) analysis, GeneCards, DisGeNET, and SwissTargetPrediction databases to analyze blank and SYD-medicated rat serum, comparing the findings with those of SYD aqueous extract in previous studies to identify SYD circulating compounds/components with predictable target genes. Using network pharmacology, the potential active compounds and corresponding hub genes associated with modulating GM to suppress CRC were selected for molecular docking. In vivo experiments, a CRC transplantation tumor model was established in BALB/c mice using CT26 cells, with SYD gavage for 14 days. To investigate the mechanism of SYD-regulated GM against CRC, HE and IHC staining, Western blotting, and 16S rRNA sequencing were employed. RESULTS LC-MS identified 26 SYD compounds with computationally predicted target genes. Network pharmacology prioritized 13 compounds targeting 8 inflammation/immunity-related genes (IL-17/TNF pathways), validated by molecular docking. In vivo experiments, SYD dose-dependently suppressed tumor growth (p < 0.05, medium/high doses), as confirmed by HE staining and IHC analysis of Ki-67. Notably, SYD potentially delayed CRC liver metastasis and alleviated hepatic injury in tumor-bearing mice. Western blotting demonstrated SYD's inhibition of the IL-17/TNF/NF-κB axis, aligning with computational predictions. 16S rRNA sequencing revealed SYD-enriched Akkermansia and GM structural shifts, mechanistically linking microbiota remodeling to anti-tumor efficacy. CONCLUSIONS SYD combats CRC via dual modulation of IL-17/TNF/NF-κB signaling and GM ecosystems (e.g., Akkermansia enrichment). This microbiota-immune crosstalk positions SYD as a potential adjunct to conventional therapies, particularly for CRC patients with dysbiosis.
Collapse
Affiliation(s)
- Yaojun Rong
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Guiyu Zhang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wenhao Ye
- The Seventh Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Linhua Qi
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xiaojiang Hao
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xiaolin Li
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wuhong Zhang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yangfa Chao
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Shaodong Gu
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Emile SH, Horesh N, Strassmann V, Garoufalia Z, Gefen R, Zhou P, Wexner SD. A National Cancer Database Analysis of the Characteristics and Outcome of Colon Cancer According to Type of Preexisting Adenoma. Clin Colorectal Cancer 2025; 24:56-63. [PMID: 39443244 DOI: 10.1016/j.clcc.2024.09.002] [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: 07/26/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND The vast majority of colon cancers occur in pre-existing adenomas. Little is known about the impact of adenoma type on behavior and outcome of subsequent carcinomas. The present study aimed to assess characteristics, behavior, and outcome of colon adenocarcinoma based on histologic type of pre-existing adenoma. METHODS US-National Cancer Database was searched between 2005 and 2019 for patients with colonic adenocarcinoma with known adenoma type who underwent colectomy. Patients were divided into 3 groups according to type of adenoma in which carcinoma developed: tubular adenoma (TA), villous adenoma (VA), and tubulovillous adenoma (TVA)-associated carcinomas. The main outcome of the study was 5-year overall survival (OS). RESULTS 66,854 patients were included. 79.3% of carcinomas originated from TVA, 10.2% from VA, and 0.5% from TA. Patients with adenocarcinoma in VA were more often female whereas carcinomas in TA affected patients of Asian race more often. Approximately one-third of carcinomas in villous and tubulovillous adenomas were in the cecum whereas one-third of carcinomas in tubular adenomas were in the sigmoid colon. More TA-associated carcinomas were of clinical T1-2 stage (30.2% vs. 20.8%; P < .001), clinical N0 stage (69% vs. 62.2%, P < .001), and high grade (15.9% vs. 11.5%, P < .001) compared to VA-associated carcinomas. Patients with TA-associated carcinomas had longer mean OS than patients with VA and TVA-associated carcinomas (130.1 vs. 116.9 vs. 123.5 months, P < .0001). CONCLUSIONS Adenocarcinomas that arose in TA had more T1-2 stage and N0 stage, higher grade, and longer OS than did adenocarcinomas that arose in VA and TVA.
Collapse
Affiliation(s)
- Sameh Hany Emile
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL; Colorectal Surgery Unit, General Surgery Department, Mansoura University Hospitals, Mansoura, Egypt
| | - Nir Horesh
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL
| | - Victor Strassmann
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL
| | - Zoe Garoufalia
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL
| | - Rachel Gefen
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL; Department of General Surgery, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Peige Zhou
- Georgia Colon and Rectal Surgical Associates, Northside Hospital, Atlanta, GA
| | - Steven D Wexner
- Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL.
| |
Collapse
|
10
|
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
|
11
|
Jin L, Jin HY, Kim Y, Cho NY, Bae JM, Kim JH, Han SW, Kim TY, Kang GH. Clinicopathological and molecular features of genome-stable colorectal cancers. Histol Histopathol 2025; 40:381-388. [PMID: 38993017 DOI: 10.14670/hh-18-785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Colorectal cancers (CRCs) are traditionally divided into those with either chromosomal instability (CIN) or microsatellite instability (MSI). By utilizing TCGA data, the Laird team found a subset of CRCs, namely, genome-stable CRCs (GS CRCs), which lack both CIN and MSI. Although the molecular features of GS CRCs have been described in detail, the clinicopathological features are not well defined. A total of 437 CRCs were analyzed for copy number variation (CNV) statuses in eight genes (ARID1A, EGFR, FGFR1, KDM5B, MYBL2, MYC, SALL4, and SETDB1) using droplet-digital PCR. CRCs that showed CNV in ≤ one gene and no MSI were defined as GS-like CRCs. Clinicopathological and molecular features of GS-like CRCs were compared with those of CIN-like CRCs. GS-like CRCs comprised 4.6% of CRCs and showed a predilection toward the proximal colon, lower nuclear optical density, KRAS mutation, PIK3CA mutation, and aberrant expression of KRT7. Survival analysis showed no significant difference between the three subgroups. Through our study, the GS-like subtype was found to comprise a minor proportion of CRCs and have proclivity toward a proximal bowel location, hypochromatic tumor nuclei, aberrant KRT7 expression, and a high frequency of KRAS and PIK3CA mutations.
Collapse
Affiliation(s)
- Lingyan Jin
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hye-Yeong Jin
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Younghoon Kim
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Nam-Yun Cho
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Mo Bae
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Sae-Won Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Tae-You Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Gyeong Hoon Kang
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
12
|
Mehanna MG, El-Halawany AM, Al-Abd AM, Alqurashi MM, Bukhari HA, Kazmi I, Al-Qahtani SD, Bawadood AS, Anwar F, Al-Abbasi FA. 6-Paradol enhances sorafenib's effects against colorectal cancer and promotes active metabolite formation. Pathol Res Pract 2025; 269:155876. [PMID: 40086339 DOI: 10.1016/j.prp.2025.155876] [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: 12/04/2024] [Revised: 01/03/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
AIM This research aimed to investigate the effect of 6-Paradol on sorafenib cytotoxicity in colorectal cancer (CRC) cell lines (HT-29, HCT-116, LS-174T, CaCo-2) through its effects on cellular processes. BACKGROUND CRC, which ranks as the third most prevalent cancer, arises in the colon or rectum due to a multifaceted interplay of dietary, lifestyle, genetic, and age-related factors. Sorafenib (multikinase inhibitor), a drug targeting multiple cancer signals, shows promise against liver, thyroid, and kidney tumors but faces resistance in CRC. 6-Paradol (Zingiberaceae), a natural compound, offers the potential for overcoming this resistance. OBJECTIVE The current study investigates whether 6-paradol potentiates sorafenib's anti-cancer activity in CRC cells by promoting increased drug entrapment, uptake, and metabolism. METHOD Sulpharodamine B assay, caspase-3 activity, PARP cleavage, cell cycle distribution analysis, and P-gp efflux activity were assessed for Cytotoxicity using CRC cell lines (HT-29, HCT-116, LS-174T, CaCo-2). RESULT Sorafenib showed potent cytotoxicity across CRC cell lines (IC50: 2.0-11.1 µM). Notably, 6-Paradol synergistically enhanced its effect in HT-29 and HCT-116 cells, reducing IC50 from 338 ± 55.7 to 107 ± 5.3 µM, respectively. 6-Paradol synergized with sorafenib, reducing HCT-116 cell proliferation from from 14.25 ± 0.62 to 20.7 ± 0 fg/cell and Paradol didn't show any significant change in c-PARP concentration which equaled 289.82 ± 27.32 pg/cell, suggesting enhanced cytotoxicity. 6-Paradol exposure resulted in a concentration-dependent (1-100 µM) increase in intracellular p-gp accumulation in CaCo-2, HCT-116, and HT-29. Co-incubation with 6-paradol significantly enhanced sorafenib cellular internalization within CaCo-2 cells, resulting in a concomitant decrease in its extracellular concentration from 1491 ± 132 ng/ml to 216 ± 39 ng/ml. 6-Paradol increased sorafenib intracellular accumulation ranges from 239 ± 16 to 327 ± 34 pg/cell, respectively in HCT-116 cells. 6-Paradol significantly increased the intracellular conversion of sorafenib to its active N-oxide metabolite and facilitated its retention within the CaCo-2 cell compartment. CONCLUSION In conclusion, 6-Paradol marked elevation in sorafenib's cytotoxic profile by influencing its cellular uptake and metabolism. Future research should expand in vitro studies to diverse cell lines, conducting in vivo elucidating underlying mechanisms and rigorously evaluating safety and toxicity.
Collapse
Affiliation(s)
- Mohamed G Mehanna
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Ali M El-Halawany
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr-El-Ainy Street, Cairo 11562, Egypt.
| | - Ahmed M Al-Abd
- Pharmacology Department, Medical Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt.
| | - May M Alqurashi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Hussam A Bukhari
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Salwa D Al-Qahtani
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia.
| | - Azizah Salim Bawadood
- Basic Medical Sciences Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| | - Firoz Anwar
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
13
|
Liu Z, Li Y, Wang S, Wang Y, Sui M, Liu J, Chen P, Wang J, Zhang Y, Dang C, Hou P. Genome-wide CRISPR screening identifies PHF8 as an effective therapeutic target for KRAS- or BRAF-mutant colorectal cancers. J Exp Clin Cancer Res 2025; 44:70. [PMID: 40001243 PMCID: PMC11853609 DOI: 10.1186/s13046-025-03338-2] [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: 09/24/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Mutations in KRAS and BRAF genes are prevalent in colorectal cancer (CRC), which strikingly promote tumorigenesis and lead to poor response to a variety of treatments including immunotherapy by activating the MAPK/ERK pathway. Thus, there is an urgent need to discover effective therapeutic targets and strategies. METHODS CRISPR-Cas9 lentiviral knockout library was used to screen the suppressors of anti-PD1 immunotherapy. Bioinformatic analysis was used to analyze the correlation between PHF8 expression and immune indicators in CRC. In vitro and in vivo experiments were utilized to determine the effects of PHF8 on the immune indexes and malignant phenotypes of CRC cells. qRT-PCR, western blotting, immunohistochemical (IHC) staining, and chromatin immunoprecipitation (ChIP)-qPCR assays were used to determine the regulatory effects of PHF8 on PD-L1, KRAS, BRAF, and c-Myc and the regulatory effect c-Myc/miR-22-3p signaling axis on PHF8 expression in CRC cells. RESULTS This study identified histone lysine demethylase PHF8 as a negative regulator for the efficacy of anti-PD1 therapy and found that it was highly expressed in CRCs and strongly associated with poor patient survival. Functional studies showed that PHF8 played an oncogenic role in KRAS- or BRAF-mutant CRC cells, but not in wild-type ones. Mechanistically, PHF8 up-regulated the expression of PD-L1, KRAS, BRAF, and c-Myc by increasing the levels of transcriptional activation marks H3K4me3 and H3K27ac and decreasing the levels of transcriptional repression mark H3K9me2 within their promoter regions, promoting immune escape and tumor progression. Besides, our data also demonstrated that PHF8 was up-regulated by the c-Myc/miR-22-3p signaling axis to form a positive feedback loop. Targeting PHF8 substantially improved the efficacy of anti-PD1 therapy and inhibited the malignant phenotypes of KRAS- or BRAF-mutant CRC cells. CONCLUSION Our data demonstrate that PHF8 may be an effective therapeutic target for KRAS- or BRAF-mutant CRCs.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Yiqi Li
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, P.R. China
| | - Simeng Wang
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Yubo Wang
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Mengjun Sui
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Jiaxin Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Pu Chen
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Jianling Wang
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Yuchen Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Chengxue Dang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China.
| | - Peng Hou
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China.
| |
Collapse
|
14
|
Li W, Wang N, Ye H, Chen M. Correlation Between Skeletal Muscle Mass and Different Pathological Types of Colorectal Polyp in Chinese Asymptomatic Population. Int J Gen Med 2025; 18:927-938. [PMID: 39995636 PMCID: PMC11849526 DOI: 10.2147/ijgm.s503137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Background Low relative muscle mass was identified to be related to ascending risk of pre-cancerous polyps (adenoma) in recent cohort study. Our study aimed to dig out the correlation between muscle mass and different pathological types of colorectal polyps in Chinese asymptomatic population. Methods In all, 5923 adults were included. The effects of low skeletal muscle mass index (SMI) on colorectal polyp occurrence, including different pathological types, and the effects modification of age and BMI were analyzed using univariate and multivariate logistic regression. Results Lower SMI was connected with the lower occurrence of colorectal polyp (OR: 0.810, 95% CI: 0.683~0.960, p=0.015). Considering different pathological types of colorectal polyps, lower SMI was associated with lower occurrence of inflammatory polyp (OR: 0.633, 95% CI: 0.434~0.898, p=0.013), rather than conventional adenoma and serrated polyp (all p>0.05). Besides, SMI was positively related to the occurrence of 2 pathological types of colorectal polyp in males: inflammatory polyp (OR: 1.237, 95% CI: 1.058~1.444, p=0.007) and serrated polyp (OR: 1.288, 95% CI: 1.143~1.456, p<0.001). The interaction effect of BMI and SMI on occurrence of inflammatory polyp after adjusting age and smoking status was significant (p=0.015). For individuals with low SMI (compared with the normal SMI group), the incidence of inflammatory polyp was reduced from 8.95% to 3.50% in the low BMI quartile (Q1) in the adjusted model (OR of 0.332, 95% CI: 0.005-0.061, p<0.001). It was noticeable for males rather than females that individuals with colorectal polyps had higher levels of SMI (p=0.003). In addition, individuals with inflammatory polyps as well as serrated polyps possessed higher levels of SMI in males (all p<0.05). Conclusion Generally, especially in Chinese asymptomatic males, low SMI kept independent effect on the presence of inflammatory polyp and serrated polyp, rather than conventional adenoma.
Collapse
Affiliation(s)
- Wenya Li
- Ultrasound Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Na Wang
- Healthcare Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Huajun Ye
- Gastroenterology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Mengjun Chen
- Gastroenterology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
15
|
Camandona A, Gagliardi A, Licheri N, Tarallo S, Francescato G, Budinska E, Carnogurska M, Zwinsová B, Martinoglio B, Franchitti L, Gallo G, Cutrupi S, De Bortoli M, Pardini B, Naccarati A, Ferrero G. Multiple regulatory events contribute to a widespread circular RNA downregulation in precancer and early stage of colorectal cancer development. Biomark Res 2025; 13:30. [PMID: 39980011 PMCID: PMC11844049 DOI: 10.1186/s40364-025-00744-8] [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: 11/21/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Early detection of colorectal cancer (CRC) significantly improves its management and patients' survival. Circular RNAs (circRNAs) are peculiar covalently closed transcripts involved in gene expression modulation whose dysregulation has been extensively reported in CRC cells. However, little is known about their alterations in the early phases of colorectal carcinogenesis. METHODS In this study, we performed an integrative analysis of circRNA profiles in RNA-sequencing (RNA-Seq) data of 96 colorectal cancers, 27 adenomas, and matched adjacent mucosa tissues. We also investigated the levels of cognate linear transcripts and those of regulating RNA-binding proteins (RBPs). Levels of circRNA-interacting microRNAs (miRNAs) were explored by integrating data of small RNA-Seq performed on the same samples. RESULTS Our results revealed a significant dysregulation of 34 circRNAs (paired adj. p < 0.05), almost exclusively downregulated in tumor tissues and, prevalently, in early disease stages. This downregulation was associated with decreased expression of circRNA host genes and those encoding for RBPs involved in circRNA biogenesis, including NOVA1, RBMS3, and MBNL1. Guilt-by-association analysis showed that dysregulated circRNAs correlated with increased predicted activity of cell proliferation, DNA repair, and c-Myc signaling pathways. Functional analysis showed interactions among dysregulated circRNAs, RBPs, and miRNAs, which were supported by significant correlations among their expression levels. Findings were validated in independent cohorts and public datasets, and the downregulation of circLPAR1(2,3) and circLINC00632(5) was validated by ddPCR. CONCLUSIONS These results support that multiple altered regulatory mechanisms may contribute to the reduction of circRNA levels that characterize early colorectal carcinogenesis.
Collapse
Affiliation(s)
- Alessandro Camandona
- Department of Clinical and Biological Sciences, University of Torino, Turin, 10100, Italy
| | - Amedeo Gagliardi
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Turin, 10060, Italy
- Candiolo Cancer Institute, FPO IRCCS, Candiolo, Turin, 10060, Italy
| | - Nicola Licheri
- Department of Clinical and Biological Sciences, University of Torino, Turin, 10100, Italy
| | - Sonia Tarallo
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Turin, 10060, Italy
- Candiolo Cancer Institute, FPO IRCCS, Candiolo, Turin, 10060, Italy
| | - Giulia Francescato
- Department of Clinical and Biological Sciences, University of Torino, Turin, 10100, Italy
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Turin, 10060, Italy
| | - Eva Budinska
- RECETOX, Faculty of Science, Masaryk University, Brno, 61137, Czech Republic
| | - Martina Carnogurska
- RECETOX, Faculty of Science, Masaryk University, Brno, 61137, Czech Republic
| | - Barbora Zwinsová
- RECETOX, Faculty of Science, Masaryk University, Brno, 61137, Czech Republic
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, 62500, Czech Republic
| | | | - Lorenzo Franchitti
- Department of Clinical and Biological Sciences, University of Torino, Turin, 10100, Italy
| | - Gaetano Gallo
- Department of Surgery, "La Sapienza" University of Rome, Rome, 00161, Italy
- Department of Colorectal Surgery, Clinica S. Rita, Vercelli, 13100, Italy
| | - Santina Cutrupi
- Department of Clinical and Biological Sciences, University of Torino, Turin, 10100, Italy
| | - Michele De Bortoli
- Department of Clinical and Biological Sciences, University of Torino, Turin, 10100, Italy
| | - Barbara Pardini
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Turin, 10060, Italy
- Candiolo Cancer Institute, FPO IRCCS, Candiolo, Turin, 10060, Italy
| | - Alessio Naccarati
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Turin, 10060, Italy
- Candiolo Cancer Institute, FPO IRCCS, Candiolo, Turin, 10060, Italy
| | - Giulio Ferrero
- Department of Clinical and Biological Sciences, University of Torino, Turin, 10100, Italy.
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Turin, 10060, Italy.
| |
Collapse
|
16
|
Xiao C, Wu H, Long J, You F, Li X. Olink Profiling of Intestinal Tissue Identifies Novel Biomarkers For Colorectal Cancer. J Proteome Res 2025; 24:599-611. [PMID: 39757570 PMCID: PMC11812010 DOI: 10.1021/acs.jproteome.4c00728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/09/2024] [Accepted: 12/25/2024] [Indexed: 01/07/2025]
Abstract
Comprehensive protein profiling in intestinal tissues provides detailed information about the pathogenesis of colorectal cancer (CRC). This study quantified the expression levels of 92 oncology-related proteins in tumors, paired para-carcinoma tissues, and remote normal tissues from a cohort of 52 CRC patients utilizing the Olink technology. The proteomic profile of normal tissues closely resembled that of para-carcinoma tissues while distinctly differing from that of tumors. Among the 68 differentially expressed proteins (DEPs) identified between the tumor and normal tissues, WISP-1, ESM-1, and TFPI-2 showed the most pronounced alterations and exhibited relatively strong correlations. These markers also presented the highest AUC values for distinguishing between tissue types. Bioinformatic analysis of the DEPs revealed that the plasma membrane and the PI3K-AKT signaling pathway were among the most enriched GO terms and KEGG pathways. Furthermore, although TFPI-2 is typically recognized as a tumor suppressor, both Olink and enzyme linked immunosorbent assay (ELISA) analyses have demonstrated that its expression is significantly elevated in tumors compared with paired normal tissues. To the best of our knowledge, this is the first study to profile the proteome of intestinal tissue using the Olink technology. This work offers valuable insights into potential biomarkers and therapeutic targets for CRC, complementing the Olink profiling of circulating proteins.
Collapse
Affiliation(s)
- Chong Xiao
- TCM
Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese
Medicine, Chengdu 610075, Sichuan, China
- Oncology
Teaching and Research Department, Chengdu
University of Traditional Chinese Medicine, Chengdu 610075, Sichuan, China
| | - Hao Wu
- TCM
Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese
Medicine, Chengdu 610075, Sichuan, China
| | - Jing Long
- TCM
Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese
Medicine, Chengdu 610075, Sichuan, China
| | - Fengming You
- TCM
Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese
Medicine, Chengdu 610075, Sichuan, China
- Institute
of Oncology, Chengdu University of Traditional
Chinese Medicine, Chengdu 610075, Sichuan, China
| | - Xueke Li
- TCM
Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese
Medicine, Chengdu 610075, Sichuan, China
- Oncology
Teaching and Research Department, Chengdu
University of Traditional Chinese Medicine, Chengdu 610075, Sichuan, China
| |
Collapse
|
17
|
Liu Z, Liu Q, Zeng A, Song L. Regulatory function of endoplasmic reticulum stress in colorectal cancer: Mechanism, facts, and perspectives. Int Immunopharmacol 2025; 147:114024. [PMID: 39764998 DOI: 10.1016/j.intimp.2025.114024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/29/2025]
Abstract
Colorectal cancer (CRC) is an exceedingly common and profoundly impactful malignancy of the digestive system, posing a grave threat to human health. Endoplasmic reticulum stress (ERS) is an intracellular biological reaction that mobilizes the unfolded protein response (UPR) to tackling dysregulation in protein homeostasis. This process subtly modulates the cell to either restore normal cellular function or steer it towards apoptosis. The high metabolic demands of CRC cells sculpt a rigorous tumor microenvironment (TME), compelling CRC cells to experience ERS. Adaptive responses induced by mild ERS furnish the necessary conditions for the survival of CRC cells, whereas the cell death mechanisms triggered by sustained ERS could be considered a prospective strategy for cancer therapy. Considering the complex regulation of ERS in cancer development, this article offers a comprehensive review of the molecular mechanisms through which ERS influences CRC fate. It provides crucial insights for exploring the role of ERS in the occurrence and progression of CRC, laying a new theoretical foundation for devising precise therapeutic strategies targeting ERS. Furthermore, by synthesizing extensive clinical and preclinical studies, we delve into therapeutic strategies targeting ERS, including the potential of targeting ERS in immunotherapy, the utilization of native compounds, advancements in proteasome inhibitors, and the potential synergies of these strategies with traditional chemotherapy agents and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Zihan Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan 610041, China.
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
18
|
Hunter E, Alshaker H, Weston C, Issa M, Bautista S, Gebregzabhar A, Virdi A, Dring A, Powell R, Green J, Lal R, Velchuru V, Aryal K, Bin Abu Hassan MR, Meng GT, Patel JS, Mohamed Gani SP, Lim CR, Guiel T, Akoulitchev A, Pchejetski D. A New Blood-Based Epigenetic Diagnostic Biomarker Test (EpiSwitch ®® NST) with High Sensitivity and Positive Predictive Value for Colorectal Cancer and Precancerous Polyps. Cancers (Basel) 2025; 17:521. [PMID: 39941889 PMCID: PMC11816175 DOI: 10.3390/cancers17030521] [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: 12/08/2024] [Revised: 01/25/2025] [Accepted: 02/02/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Colorectal cancer (CRC) arises from the epithelial lining of the colon or rectum, often following a progression from benign adenomatous polyps to malignant carcinoma. Screening modalities such as colonoscopy, faecal immunochemical tests (FIT), and FIT-DNA are critical for early detection and prevention, but non-invasive methods lack sensitivity to polyps and early CRC. Chromosome conformations (CCs) are potent epigenetic regulators of gene expression. We have previously developed an epigenetic assay, EpiSwitch®®, that employs an algorithmic-based CCs analysis. Using EpiSwitch®® technology, we have shown the presence of cancer-specific CCs in peripheral blood mononuclear cells (PBMCs) and primary tumours of patients with melanoma and prostate cancer. EpiSwitch®®-based commercial tests are now available to diagnose prostate cancer with 94% accuracy (PSE test) and response to immune checkpoint inhibitors across 14 cancers with 85% accuracy (CiRT test). Methods/Results/Conclusions: Using blood samples collected from n = 171 patients with CRC, n = 44 patients with colorectal polyps and n = 110 patients with a 'clear' colonoscopy we performed whole Genome DNA screening for CCs correlating to CRC diagnosis. Our findings suggest the presence of two eight-marker CC signatures (EpiSwitch®® NST) in whole blood that allow diagnosis of CRC and precancerous polyps, respectively. Independent validation cohort testing demonstrated high accuracy in identifying colorectal polyps and early versus late stages of CRC with an exceptionally high sensitivity of 79-90% and a high positive prediction value of 60-84%. Linking the top diagnostic CCs to nearby genes, we have built pathways maps that likely underline processes contributing to the pathology of polyp and CRC progression, including TGFβ, cMYC, Rho GTPase, ROS, TNFa/NFκB, and APC.
Collapse
Affiliation(s)
- Ewan Hunter
- Oxford BioDynamics Plc., Oxford OX4 2WB, UK (A.A.)
| | - Heba Alshaker
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | | | - Mutaz Issa
- Oxford BioDynamics Plc., Oxford OX4 2WB, UK (A.A.)
| | | | | | - Anya Virdi
- Oxford BioDynamics Plc., Oxford OX4 2WB, UK (A.A.)
| | - Ann Dring
- Oxford BioDynamics Plc., Oxford OX4 2WB, UK (A.A.)
| | - Ryan Powell
- Oxford BioDynamics Plc., Oxford OX4 2WB, UK (A.A.)
| | - Jayne Green
- Oxford BioDynamics Plc., Oxford OX4 2WB, UK (A.A.)
| | - Roshan Lal
- James Paget University Hospitals NHS Trust, Great Yarmouth NR31 6LA, UK
| | - Vamsi Velchuru
- James Paget University Hospitals NHS Trust, Great Yarmouth NR31 6LA, UK
| | - Kamal Aryal
- James Paget University Hospitals NHS Trust, Great Yarmouth NR31 6LA, UK
| | | | - Goh Tiong Meng
- Island Hospital Penang, Jalan Macalister, George Town 10450, Malaysia
| | - Janisha Suriakant Patel
- Penang Reference Laboratory, Oxford BioDynamics Plc., Jalan Tanjung Tokong, George Town 10470, Malaysia
| | | | - Chun Ren Lim
- Penang Reference Laboratory, Oxford BioDynamics Plc., Jalan Tanjung Tokong, George Town 10470, Malaysia
| | - Thomas Guiel
- Oxford BioDynamics Inc., Frederick, MD 21703, USA
| | | | - Dmitri Pchejetski
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| |
Collapse
|
19
|
Zou J, Shi X, Wu Z, Zuo S, Tang X, Zhou H, Huang Y. MRTX1133 attenuates KRAS G12D mutated-colorectal cancer progression through activating ferroptosis activity via METTL14/LINC02159/FOXC2 axis. Transl Oncol 2025; 52:102235. [PMID: 39657309 PMCID: PMC11683245 DOI: 10.1016/j.tranon.2024.102235] [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: 08/07/2024] [Revised: 11/30/2024] [Accepted: 12/07/2024] [Indexed: 12/12/2024] Open
Abstract
Colorectal cancer (CRC) ranks as the third most commonly diagnosed cancer and the second leading cause of cancer-related deaths worldwide. Studies have shown that CRC patients with KRAS mutations, especially KRASG12D, have an increased risk of metastasis. Emerging evidence indicates that long non-coding RNAs (lncRNAs) are crucial in the carcinogenesis and progression of various cancers, regulating multiple biological processes but the link between KRASG12D mutations and lncRNAs in CRC remains unclear. Therefore, this study was designed to identify a novel lncRNA involved in KRASG12D-mutated CRC and to elucidate its molecular mechanisms. The analysis of differentially expressed lncRNAs in the GSE201412 dataset revealed that LINC02159 was significantly upregulated following treatment with the KRASG12D inhibitor MTRX1133 Data from the GTEx database indicated that LINC02159 is highly expressed in CRC tumour tissues and is associated with better patient outcomes. In vitro and in vivo experiments suggest that LINC02159 acts as a tumour suppressor in CRC progression. Specifically, LINC02159 knockdown negated the inhibitory effects of MRTX1133 on tumourigenesis and its promotive effect on ferroptosis in KRASG12D-mutated CRC cells. LINC02159 expression is regulated by METTL14, with METTL14 knockdown decreasing m6A methylation of LINC02159, leading to its increased expression in CRC cells. Additionally, LINC02159 stabilised FOXC2 expression through de-ubiquitination. Rescue experiments further clarified that the METTL14/LINC02159/FOXC2 signalling axis is crucial for the inhibitory effects of MRTX1133 in KRASG12D-mutated CRC. Our study provides novel insights into the therapeutic potential of MRTX1133 in treating KRASG12D-mutated CRC by identifying a METTL14/LINC02159/FOXC2 signalling axis that mediates drug response. Our findings highlight the importance of understanding the molecular mechanisms of lncRNAs in cancer to develop effective targeted therapies.
Collapse
Affiliation(s)
- Junwei Zou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Xiuhua Shi
- Department of Radiotherapy & Oncology, The No.2 People's Hospital of Wuhu City, Wuhu, Anhui, China
| | - Zhaoying Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Siyuan Zuo
- School of Clinical Medicine, Wannan Medical College, Wuhu, Anhui, China
| | - Xiaolei Tang
- Center for Translational Medicine, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Hailang Zhou
- Department of Gastroenterology, Lianshui People's Hospital of kangda college Affiliated to Nanjing Medical University, Huai'an, Jiangsu, China.
| | - Yong Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China.
| |
Collapse
|
20
|
Lei X, Zhang L, Liu Y, Sun H, Yan J, Liu S. ZNF26-Associated Genes as Prognostic Signatures in Colorectal Cancer with Broad Therapeutic Implications. J Appl Genet 2025; 66:141-153. [PMID: 38568413 DOI: 10.1007/s13353-024-00854-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 01/25/2025]
Abstract
The identification of biomarkers correlated with colorectal cancer (CRC) prognosis holds substantial importance from both clinical and scientific perspectives. Zinc finger protein 26 (ZNF26) has not been previously investigated or documented in solid tumors; thus, further research is necessary to ascertain its prognostic value in CRC. Gene expression profiles and clinicopathological data were acquired from The Cancer Genome Atlas (TCGA) database. Subsequently, expression correlation was assessed utilizing the TCGA CRC cohort. The prognostic value of ZNF26 was evaluated through Kaplan-Meier (KM) and ROC curve analyses. Following this, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were conducted to perform enrichment analysis between high- and low-ZNF26 expression groups. The association between immune cells, immune checkpoint genes, and ZNF26 expression levels was examined. Lastly, the research findings were further validated using CRC tissue samples. The results revealed that, in comparison to healthy controls, CRC significantly reduced ZNF26 expression. Elevated ZNF26 expression was associated with poorer overall survival in CRC patients. Additionally, high ZNF26 expression exhibited an inverse relationship with the immunological score and immune checkpoint gene expression in CRC patients. The findings from the TCGA data analysis were corroborated by the PCR results obtained from CRC tissue samples. ZNF26 is markedly upregulated in colorectal cancer tissues, potentially serving as a biomarker for CRC.
Collapse
Affiliation(s)
- Xue Lei
- Department of Clinical Specialty of Integrated Traditional Chinese and Western Medicine, Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Lijia Zhang
- Ethics Committee Office, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Ye Liu
- Department of Intensive Care Unit, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Heng Sun
- Department of Oncology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Jun Yan
- Department of Oncology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Songjiang Liu
- Department of Oncology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
21
|
Coelho D, Estêvão D, Oliveira MJ, Sarmento B. Radioresistance in rectal cancer: can nanoparticles turn the tide? Mol Cancer 2025; 24:35. [PMID: 39885557 PMCID: PMC11784129 DOI: 10.1186/s12943-025-02232-x] [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: 12/22/2024] [Accepted: 01/14/2025] [Indexed: 02/01/2025] Open
Abstract
Rectal cancer accounts for over 35% of the worldwide colorectal cancer burden representing a distinctive subset of cancers from those arising in the colon. Colorectal cancers exhibit a continuum of traits that differ with their location in the large intestine. Due to anatomical and molecular differences, rectal cancer is treated differently from colon cancer, with neoadjuvant chemoradiotherapy playing a pivotal role in the control of the locally advanced disease. However, radioresistance remains a major obstacle often correlated with poor prognosis. Multifunctional nanomedicines offer a promising approach to improve radiotherapy response rates, as well as to increase the intratumoral concentration of chemotherapeutic agents, such as 5-Fluorouracil. Here, we revise the main molecular differences between rectal and colon tumors, exploring the complex orchestration beyond rectal cancer radioresistance and the most promising nanomedicines reported in the literature to improve neoadjuvant therapy response rates.
Collapse
Affiliation(s)
- Diogo Coelho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| | - Diogo Estêvão
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Cancer Research Institute, Ghent University, Ghent, Belgium
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, Porto, 4200-319, Portugal
| | - Maria José Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, Porto, 4200-319, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal.
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal.
- IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal.
| |
Collapse
|
22
|
Grosu S, Fabritius MP, Winkelmann M, Puhr-Westerheide D, Ingenerf M, Maurus S, Graser A, Schulz C, Knösel T, Cyran CC, Ricke J, Kazmierczak PM, Ingrisch M, Wesp P. Effect of artificial intelligence-aided differentiation of adenomatous and non-adenomatous colorectal polyps at CT colonography on radiologists' therapy management. Eur Radiol 2025:10.1007/s00330-025-11371-0. [PMID: 39862248 DOI: 10.1007/s00330-025-11371-0] [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: 10/16/2024] [Revised: 12/01/2024] [Accepted: 12/22/2024] [Indexed: 01/27/2025]
Abstract
OBJECTIVES Adenomatous colorectal polyps require endoscopic resection, as opposed to non-adenomatous hyperplastic colorectal polyps. This study aims to evaluate the effect of artificial intelligence (AI)-assisted differentiation of adenomatous and non-adenomatous colorectal polyps at CT colonography on radiologists' therapy management. MATERIALS AND METHODS Five board-certified radiologists evaluated CT colonography images with colorectal polyps of all sizes and morphologies retrospectively and decided whether the depicted polyps required endoscopic resection. After a primary unassisted reading based on current guidelines, a second reading with access to the classification of a radiomics-based random-forest AI-model labelling each polyp as "non-adenomatous" or "adenomatous" was performed. Performance was evaluated using polyp histopathology as the reference standard. RESULTS 77 polyps in 59 patients comprising 118 polyp image series (47% supine position, 53% prone position) were evaluated unassisted and AI-assisted by five independent board-certified radiologists, resulting in a total of 1180 readings (subsequent polypectomy: yes or no). AI-assisted readings had higher accuracy (76% +/- 1% vs. 84% +/- 1%), sensitivity (78% +/- 6% vs. 85% +/- 1%), and specificity (73% +/- 8% vs. 82% +/- 2%) in selecting polyps eligible for polypectomy (p < 0.001). Inter-reader agreement was improved in the AI-assisted readings (Fleiss' kappa 0.69 vs. 0.92). CONCLUSION AI-based characterisation of colorectal polyps at CT colonography as a second reader might enable a more precise selection of polyps eligible for subsequent endoscopic resection. However, further studies are needed to confirm this finding and histopathologic polyp evaluation is still mandatory. KEY POINTS Question This is the first study evaluating the impact of AI-based polyp classification in CT colonography on radiologists' therapy management. Findings Compared with unassisted reading, AI-assisted reading had higher accuracy, sensitivity, and specificity in selecting polyps eligible for polypectomy. Clinical relevance Integrating an AI tool for colorectal polyp classification in CT colonography could further improve radiologists' therapy recommendations.
Collapse
Affiliation(s)
- Sergio Grosu
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany.
| | - Matthias P Fabritius
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Michael Winkelmann
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Daniel Puhr-Westerheide
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Maria Ingenerf
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Stefan Maurus
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- Department for Diagnostic and Interventional Radiology and Neuroradiology, Klinikum Kempten, Robert-Weixler-Straße 50, 87439, Kempten, Germany
| | - Anno Graser
- Gemeinschaftspraxis Radiologie München, Burgstraße 7, 80331, Munich, Germany
| | - Christian Schulz
- Department of Medicine II, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Thomas Knösel
- Department of Pathology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Clemens C Cyran
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Jens Ricke
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Philipp M Kazmierczak
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Michael Ingrisch
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- Munich Center for Machine Learning (MCML), Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Philipp Wesp
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- Munich Center for Machine Learning (MCML), Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| |
Collapse
|
23
|
Singer M, Valerin J, Zhang Z, Zhang Z, Dayyani F, Yaghmai V, Choi A, Imagawa D, Abi-Jaoudeh N. Promising Cellular Immunotherapy for Colorectal Cancer Using Classical Dendritic Cells and Natural Killer T Cells. Cells 2025; 14:166. [PMID: 39936958 DOI: 10.3390/cells14030166] [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: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related morbidity and mortality around the world. Despite advances in surgery, chemotherapy, and targeted therapies, the prognosis for patients with metastatic or advanced CRC remains poor. Immunotherapies comprising immune checkpoint inhibitors showed disappointing responses in metastatic CRC (mCRC). However, cellular immunotherapy, specifically using classical dendritic cells (cDCs), may hold unique promise in immune recognition for CRC antigens. cDCs are substantial players in immune recognition and are instrumental in orchestrating innate and adaptive immune responses by processing and presenting tumor antigens to effector cells. Natural killer T (NKT) cells are insufficiently studied but unique effector cells because of their ability to bridge innate and adaptive immune reactions and the crosstalk with dendritic cells in cancer. This review explores the therapeutic potential of using both cDCs and NKT cells as a synergistic therapy in CRC, focusing on their biological roles, strategies for harnessing their capabilities, clinical applications, and the challenges within the tumor microenvironment. Both cDCs and NKT cells can be used as a new effective approach for cell-based therapies in cancers to provide a new hope for CRC patients that are challenging to treat.
Collapse
Affiliation(s)
- Mahmoud Singer
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Jennifer Valerin
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA
| | - Zhuoli Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Zigeng Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Farshid Dayyani
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA
| | - Vahid Yaghmai
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - April Choi
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA
| | - David Imagawa
- Department of Surgery, University of California Irvine, Orange, CA 92697, USA
| | - Nadine Abi-Jaoudeh
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| |
Collapse
|
24
|
Taha SR, Karimi M, Mahdavi B, Yousefi Tehrani M, Bemani A, Kabirian S, Mohammadi J, Jabbari S, Hushmand M, Mokhtar A, Pourhanifeh MH. Crosstalk between non-coding RNAs and programmed cell death in colorectal cancer: implications for targeted therapy. Epigenetics Chromatin 2025; 18:3. [PMID: 39810224 PMCID: PMC11734566 DOI: 10.1186/s13072-024-00560-8] [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: 09/17/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the most common causes of cancer-related mortality worldwide. Its progression is influenced by complex interactions involving genetic, epigenetic, and environmental factors. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), have been identified as key regulators of gene expression, affecting diverse biological processes, notably programmed cell death (PCD). OBJECTIVE This review aims to explore the relationship between ncRNAs and PCD in CRC, focusing on how ncRNAs influence cancer cell survival, proliferation, and treatment resistance. METHODS A comprehensive literature analysis was conducted to examine recent findings on the role of ncRNAs in modulating various PCD mechanisms, including apoptosis, autophagy, necroptosis, and pyroptosis, and their impact on CRC development and therapeutic response. RESULTS ncRNAs were found to significantly regulate PCD pathways, impacting tumor growth, metastasis, and treatment sensitivity in CRC. Their influence on these pathways highlights the potential of ncRNAs as biomarkers for early CRC detection and as targets for innovative therapeutic interventions. CONCLUSION Understanding the involvement of ncRNAs in PCD regulation offers new insights into CRC biology. The targeted modulation of ncRNA-PCD interactions presents promising avenues for personalized cancer treatment, which may improve patient outcomes by enhancing therapeutic effectiveness and reducing resistance.
Collapse
Affiliation(s)
- Seyed Reza Taha
- Department of Pathology and Immunology, Washington University School of Medicine, St. LouisWashington, MO, USA
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Karimi
- Faculty of Medicine, Bogomolets National Medical University (NMU), Kiev, Ukraine.
| | - Bahar Mahdavi
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Ali Bemani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahriar Kabirian
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Javad Mohammadi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sina Jabbari
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Meysam Hushmand
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mokhtar
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
- PAKAN Institute, Tehran, Iran.
| |
Collapse
|
25
|
Mroczkowski P, Kusian H, Jannasch O, Lippert H, Zajdel R, Zajdel K, Sadowski A, Merecz-Sadowska A. Treatment Quality of Rectal Cancer Patients in Certified Colorectal Cancer Centers Versus Non-Certified Hospitals: A Comparative Analysis. Cancers (Basel) 2025; 17:120. [PMID: 39796747 PMCID: PMC11720547 DOI: 10.3390/cancers17010120] [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: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: The certification of hospitals as colorectal cancer centers aims to improve treatment quality, but evidence supporting its effectiveness remains limited. This study evaluated the impact of certification on treatment outcomes for rectal cancer patients in Germany. Methods: We conducted a retrospective analysis of 14,905 patients with primary rectal cancer (UICC Stages I-III) treated at 271 hospitals. Treatment outcomes were compared between certified colorectal cancer centers (3624 patients in 55 hospitals) and non-certified hospitals (11,281 patients in 216 hospitals). Additionally, a subset analysis examined outcomes before and after certification within the same institutions. Results: Certified centers demonstrated higher utilization of preoperative imaging (endorectal ultrasound: 70.7% vs. 58.2%, p < 0.001; pelvic MRI: 39.1% vs. 28.5%, p < 0.001) and lower rates of intraoperative complications (4.6% vs. 6.2%, p < 0.001). Surgical quality indicators, including M.E.R.C.U.R.Y. classification (Grade 1: 86.5% both groups, p = 0.620) and anastomotic leakage rates (11.3% vs. 11.9%, p = 0.407), were comparable between certified and non-certified hospitals. Despite treating patients with more favorable tumor stages, certified centers showed no significant advantage in 5-year overall survival (82.8% vs. 82.0%, p = 0.880) or 30-day mortality (2.6% both groups, p = 0.869). Hospital stays were marginally shorter in certified centers (19.46 vs. 20.24 days, p < 0.001). Conclusions: While certification was associated with improved adherence to diagnostic protocols and reduced intraoperative complications, it did not significantly impact surgical quality or long-term survival outcomes. These findings suggest that certification alone may not guarantee superior treatment quality, as hospitals participating in quality assurance programs achieved comparable results without formal certification.
Collapse
Affiliation(s)
- Paweł Mroczkowski
- Department for General and Colorectal Surgery, Medical University of Lodz, Pl. Hallera 1, 90-647 Lodz, Poland;
- Institute for Quality Assurance in Operative Medicine Ltd., Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany; (H.K.); (O.J.); (H.L.)
- Department for Surgery, University Hospital Knappschaftskrankenhaus, Ruhr-University, In der Schornau 23-25, 44892 Bochum, Germany
| | - Henry Kusian
- Institute for Quality Assurance in Operative Medicine Ltd., Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany; (H.K.); (O.J.); (H.L.)
| | - Olof Jannasch
- Institute for Quality Assurance in Operative Medicine Ltd., Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany; (H.K.); (O.J.); (H.L.)
| | - Hans Lippert
- Institute for Quality Assurance in Operative Medicine Ltd., Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany; (H.K.); (O.J.); (H.L.)
- Department for General, Visceral and Vascular Surgery, Otto-von-Guericke-University, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Radosław Zajdel
- Department of Economic and Medical Informatics, University of Lodz, 90-214 Lodz, Poland; (R.Z.); (A.S.)
- Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland;
| | - Karolina Zajdel
- Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland;
| | - Arkadiusz Sadowski
- Department of Economic and Medical Informatics, University of Lodz, 90-214 Lodz, Poland; (R.Z.); (A.S.)
| | - Anna Merecz-Sadowska
- Department of Economic and Medical Informatics, University of Lodz, 90-214 Lodz, Poland; (R.Z.); (A.S.)
- Department of Allergology and Respiratory Rehabilitation, Medical University of Lodz, 90-725 Lodz, Poland
| |
Collapse
|
26
|
Tang YC, Ou JJ, Hsu SC, Huang CH, Lin LM, Chang HH, Wang YH, Huang ZT, Sun M, Liu KJ, Hung YM, Lai CY, Shih C, Chen CT, Chang JY, Hsieh HP, Jiaang WT, Kuo CC. Advancing precision therapy for colorectal cancer: Developing clinical indications for multi-target kinase inhibitor BPR1J481 using patient-derived xenograft models. Pharmacol Res 2025; 211:107556. [PMID: 39709137 DOI: 10.1016/j.phrs.2024.107556] [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: 06/17/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
The large and rapid increase in the incidence and mortality of colorectal cancer (CRC) demonstrates the urgent need for new drugs with higher efficacy to treat CRC. However, the lack of applicable and reliable preclinical models significantly hinders the progress of drug development. Patient-derived xenograft (PDX) models are currently considered reliable in vivo preclinical models for predicting drug efficacy in cancer patients. This study successfully uses the CRC PDX model to develop clinical indications for the new multi-target kinase inhibitor BPR1J481 and demonstrated the anti-cancer mechanism and competitive advantages of this drug candidate. The results demonstrate that BPR1J481 exhibits significant anticancer efficacy by inducing apoptosis in CRC PDX tumor tissues and corresponding PDX-derived CRC cells. Through kinase competitive binding and kinase activity assays, we discover that BPR1J481 effectively inhibits SRC kinase activity by directly binding to its active site. The reduction in SRC phosphorylation observed in CRC PDX tumor tissues and derived cells upon treatment with BPR1J481 further validates its inhibitory potential. Furthermore, the decrease in viable cells after SRC knockout and the poorer prognosis observed in patients with higher SRC expression, emphasizes the critical significance and clinical relevance of SRC in CRC. Additionally, BPR1J481 exhibits robust anti-angiogenic effects by suppressing VEGF- and PDGF-induced endothelial cell proliferation, migration, and capillary-like tube formation through inhibition of VEGFR2 and PDGFRβ phosphorylation. Remarkably, BPR1J481 appears to demonstrate greater efficacy against CRC compared to regorafenib. These findings highlight the therapeutic potential of BPR1J481 for patients with CRC.
Collapse
Affiliation(s)
- Ya-Chu Tang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Jing-Jim Ou
- Department of Surgery, Chang Bing Show Chwan Memorial Hospital, Changhua County 505029, Taiwan
| | - Shu-Ching Hsu
- Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Chih-Hsiang Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Li-Mei Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Hsin-Huei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Yi-Hsin Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Zih-Ting Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Manwu Sun
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, National Health Research Institutes, Tainan City 704016, Taiwan
| | - Yi-Mei Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan City 704016, Taiwan
| | - Chi-Yun Lai
- Pathology Core Laboratory, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Chuan Shih
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Jang-Yang Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan; Taipei Medical University Hospital, College of Medicine, Taipei Medical University, Taipei City 110301, Taiwan; Taipei Cancer Center, Taiwan; TMU Research Center of Cancer Translational Medicine, 110301, Taiwan
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Weir-Torn Jiaang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan.
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan.
| |
Collapse
|
27
|
Xiong L, Yang X, Liu H, Wu X, Cai T, Yuan M, Huang L, Zhou C, Zheng X, Li W, Zeng Z, Li S, Lan P, Kang L, Liang Z. Glutamic-pyruvic transaminase 1 deficiency-mediated metabolic reprogramming facilitates colorectal adenoma-carcinoma progression. Sci Transl Med 2025; 17:eadp9805. [PMID: 39742507 DOI: 10.1126/scitranslmed.adp9805] [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: 04/22/2024] [Revised: 09/10/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
The tumorigenesis of colorectal cancer (CRC) often follows the normal-adenoma-carcinoma (N-A-C) sequence. However, the molecular mechanisms underlying colorectal adenoma carcinogenesis remain largely unknown. Here, we analyzed transcriptomic profile changes in normal, advanced adenoma, and carcinoma tissues from patients with CRC, revealing that glutamic-pyruvic transaminase 1 (GPT1) in colorectal tissues was down-regulated during the N-A-C process and correlated with poor CRC prognosis. Mechanistically, GPT1 was transcriptionally activated by Krüppel-like factor 4 (KLF4). GPT1 reprogrammed metabolism and suppressed CRC tumorigenesis in cells and mouse models not only through enzyme-dependent α-ketoglutarate (α-KG) production and WNT signaling inhibition but also through enzyme-independent disruption of the folate cycle through binding with methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L). Furthermore, we identified poliumoside as a GPT1 activator that restrained CRC progression in cells, patient-derived CRC organoids, and patient-derived xenograft (PDX) models of CRC. Our study uncovers a role for GPT1 in CRC tumorigenesis and shows that poliumoside is a potential drug for the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Li Xiong
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xin Yang
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Huashan Liu
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xianrui Wu
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Tanxing Cai
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Ming Yuan
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Liang Huang
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Chi Zhou
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510050, China
| | - Xiaobin Zheng
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Wenxin Li
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Ziwei Zeng
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Shujuan Li
- Department of Pharmacy, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ping Lan
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Liang Kang
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Zhenxing Liang
- Department of Colorectal Surgery (General Surgery), Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
- Biomedical Innovation Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| |
Collapse
|
28
|
Mandal S, Baker AM, Graham TA, Bräutigam K. The tumour histopathology "glossary" for AI developers. PLoS Comput Biol 2025; 21:e1012708. [PMID: 39847582 PMCID: PMC11756763 DOI: 10.1371/journal.pcbi.1012708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025] Open
Abstract
The applications of artificial intelligence (AI) and deep learning (DL) are leading to significant advances in cancer research, particularly in analysing histopathology images for prognostic and treatment-predictive insights. However, effective translation of these computational methods requires computational researchers to have at least a basic understanding of histopathology. In this work, we aim to bridge that gap by introducing essential histopathology concepts to support AI developers in their research. We cover the defining features of key cell types, including epithelial, stromal, and immune cells. The concepts of malignancy, precursor lesions, and the tumour microenvironment (TME) are discussed and illustrated. To enhance understanding, we also introduce foundational histopathology techniques, such as conventional staining with hematoxylin and eosin (HE), antibody staining by immunohistochemistry, and including the new multiplexed antibody staining methods. By providing this essential knowledge to the computational community, we aim to accelerate the development of AI algorithms for cancer research.
Collapse
Affiliation(s)
- Soham Mandal
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
- Data Science Team, Institute of Cancer Research, London, United Kingdom
| | - Ann-Marie Baker
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
| | - Trevor A. Graham
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
| | - Konstantin Bräutigam
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
29
|
Tian TT, Chen G, Sun K, Wang XY, Liu Y, Wang FQ, Yang B, Liu J, Han JY, Tang DX. ChanLingGao alleviates intestinal mucosal barrier damage and suppresses the onset and progression of Colorectal cancer in AOM/DSS murine model. Int Immunopharmacol 2024; 143:113193. [PMID: 39368132 DOI: 10.1016/j.intimp.2024.113193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/26/2024] [Accepted: 09/14/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND The occurrence of Colorectal Cancer (CRC) is influenced by various factors, including host susceptibility, immune imbalance, and environmental triggers. Numerous studies have underscored the critical role of chronic intestinal inflammation and dysbiosis in the development of CRC. Traditional Chinese Medicine (TCM) holds unique advantages in regulating the intricate process of and comprehensive treatment for systemic disease. Previous investigations by our team have confirmed the anti-cancer properties of the TCM compound ChanLingGao (CLG), including inhibiting cancer cell migration, and alleviating bone cancer pain. However, the mechanisms underlying its efficacy in alleviating chronic intestinal inflammation, modulating the gut microbiota, and protecting the intestinal mucosal barrier remain largely unknown. PURPOSE This study aims to explore the inhibitory effects of CLG on CRC tumors in mice and its potential mechanisms. METHODS A chronic inflammation-related CRC mouse model was established using AOM/DSS. The study examined the mechanisms of intestinal inflammation and tumor cell proliferation through intestinal histological morphology. High-throughput sequencing was employed to analyze changes in gut microbiota diversity and intestinal mucosal barrier integrity in CRC mice. Based on network pharmacology target prediction and Wnt/β-catenin signaling pathway analysis, the study analyzed and discussed the potential mechanisms of CLG on CRC. RESULTS CLG significantly ameliorated weight loss and increased survival rates in CRC mice, while suppressing tumor growth in the intestinal tract. Post-CLG treatment improved intestinal inflammation in CRC mice, with a significant reduction in inflammatory factors IL-6, IL-23 and LCN2, and inhibition of tumor cell proliferation markers Proliferating Cell Nuclear Antigen (PCNA), Recombinant Ki-67 Protein (Ki-67), and CCND1. 16sV3-V4 region microbiota sequencing results indicated that CLG improved dysbiosis, and significantly increased the abundance of Akkermansia bacteria, further promoting the expression of MUC-2 protein and mucin secretion. Additionally, CLG prevented the disruption of intestinal epithelial cell junction proteins Occludin, Claudin-1, ZO-1, and E-cadherin, restored the number of goblet cells, and preserved the integrity of the intestinal mucosal barrier. Further experiments suggested that CLG inhibited abnormal activation of the Wnt/β-catenin pathway, and its potential mechanism in maintaining mucosal barrier integrity might be related to blocking Wnt/β-catenin pathway. CONCLUSIONS This study demonstrates that CLG can inhibit CRC tumor growth by regulating the gut microbiota structure, reducing intestinal inflammation, improving intestinal mucosal barrier function, and inhibiting the complex process of cancer cell proliferation. This provides new clinical insights into the "membrane-oriented" treatment of CRC with CLG.
Collapse
Affiliation(s)
- Ting-Ting Tian
- Scientific Research Department, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Guo Chen
- Scientific Research Department, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Kai Sun
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiao-Yi Wang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yang Liu
- Scientific Research Department, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Fei-Qing Wang
- Scientific Research Department, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Bing Yang
- Scientific Research Department, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Jian Liu
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Dong-Xin Tang
- Scientific Research Department, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China.
| |
Collapse
|
30
|
Opperman RCM, Bosch S, Nazmi K, Bikker FJ, Brand HS, Jimenez CR, de Meij TGJ, Dekker E, de Boer NKH, Kaman WE. Detecting Colorectal Neoplasia Using Specific Fecal Fluorogenic Protease-Sensitive Substrates: A Pilot Study. Anal Chem 2024; 96:20239-20246. [PMID: 39665576 DOI: 10.1021/acs.analchem.4c04586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
BACKGROUND identification and removal of advanced adenomas (AA) reduce colorectal cancer (CRC) incidence and potentially mortality. CRC screening often uses fecal immunochemical testing to select high-risk individuals for colonoscopy, despite its low sensitivity for AA and relatively high false-positivity rate. Previous studies have linked proteases to CRC development through their ability to facilitate angiogenesis and immunoregulation. This study aims to identify colorectal neoplasia-associated proteases and their substrates as a potential noninvasive screening test, introducing an innovative application of fecal protease profiling, which has previously been limited to tissue samples. METHODS eighteen fluorogenic substrates were designed based on literature. Proteolytic degradation of these substrates was measured in fecal samples of patients with CRC (n = 12), AA (n = 9), nonadvanced adenomas (n = 10), and controls (n = 14). Substrate degradation was correlated to a matched human proteome data set, and underlying proteases were identified based on their recognition patterns. Experiments with protease inhibitors and ZnCl2 were performed to further characterize the involved proteases. RESULTS in total, 7 of the 18 substrates tested showed a significantly decreased proteolytic degradation in feces from patients with any colorectal neoplasia compared to the control group. The l-aspartic acid-l-glutamic acid substrate (ED) showed significantly decreased degradation in AA and CRC patients. ED degradation significantly decreased with the addition of ZnCl2 and the cysteine protease inhibitor NEM. CONCLUSION we successfully developed colorectal neoplasia-specific fluorogenic substrates, highlighting the ED substrate as a potential substrate for the detection of AA and CRC. Although the responsible proteases require further identification, our results suggest an association with calcium-dependent cysteine proteases.
Collapse
Affiliation(s)
- Roza C M Opperman
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, 1081 HV Amsterdam, The Netherlands
- Research Program, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sofie Bosch
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, 1081 HV Amsterdam, The Netherlands
| | - Kamran Nazmi
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Floris J Bikker
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Henk S Brand
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Connie R Jimenez
- Research Program, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Tim G J de Meij
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, 1081 HV Amsterdam, The Netherlands
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam UMC, Academic Medical Centre, 1105 AZ Amsterdam, The Netherlands
| | - Evelien Dekker
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, 1081 HV Amsterdam, The Netherlands
- Research Program, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Nanne K H de Boer
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, 1081 HV Amsterdam, The Netherlands
| | - Wendy E Kaman
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| |
Collapse
|
31
|
Thomas CE, Takashima Y, Wesselink E, Ugai T, Steinfelder RS, Buchanan DD, Qu C, Hsu L, Dias Costa A, Gallinger S, Grant RC, Huyghe JR, Thomas SS, Ogino S, Phipps AI, Nowak JA, Peters U. Association between somatic microsatellite instability, hypermutation status, and specific T cell subsets in colorectal cancer tumors. Front Immunol 2024; 15:1505896. [PMID: 39763680 PMCID: PMC11701007 DOI: 10.3389/fimmu.2024.1505896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Background Microsatellite instability-high (MSI-high) tumors comprise ~15% of sporadic colorectal cancers (CRC) and are associated with elevated T cell infiltration. However, the universality of this response across T cell subtypes with distinct functions is unknown. Methods Including 1,236 CRC tumors from three observational studies, we conducted in-situ T cell profiling using a customized 9-plex (CD3, CD4, CD8, CD45RA, CD45RO, FOXP3, KRT, MKI67, and DAPI) multispectral immunofluorescence assay. MSI status was assessed through polymerase chain reaction or immunohistochemical assays. We used multivariable ordinal logistic regression to estimate odds ratios (OR per increasing quantile) and 95% confidence intervals (CIs) for the association of MSI status with quantiles of T cell densities in either tumor epithelial or stromal tissue areas. Results Compared to microsatellite instability low or microsatellite stable (MSI-low/MSS) tumors, MSI-high status was associated with higher density for the majority of immune subsets (twelve out of eighteen) in both epithelial and stromal tissue areas. The strongest associations were for CD3+CD8+ T cells in epithelial areas [OR (95% CI) for naive, memory, and regulatory subsets = 3.49 (2.57, 4.75); 2.82 (2.10, 3.78); 3.04 (2.24, 4.13), respectively]. Conversely, stromal area CD3+CD4+ memory T cells were inversely associated [OR (95% CI) = 0.68 (0.51, 0.91)]. Discussion MSI-high status was strongly associated with higher densities of most T cell subsets in both epithelial and stromal tissue areas. Our investigation supports efforts to identify patients who may be more likely to respond to current immunotherapy treatments. Significance This study helps us better understand how a clinically relevant tumor phenotype, microsatellite instability status, is related to different functioning T cell densities in colorectal tumors, which may impact future immunotherapy strategies.
Collapse
Affiliation(s)
- Claire E. Thomas
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Yasutoshi Takashima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Evertine Wesselink
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, Netherlands
| | - Tomotaka Ugai
- Program in Molecular Pathological Epidemiology (MPE), Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
- Cancer Epidemiology Program, Dana-Farber/Harvard Cancer Center, Boston, MA, United States
| | - Robert S. Steinfelder
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Daniel D. Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Parkville, VIC, Australia
- University of Melbourne Centre for Cancer Research, The University of Melbourne, Parkville, VIC, Australia
- Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Conghui Qu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, United States
| | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Robert C. Grant
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jeroen R. Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Sushma S. Thomas
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Shuji Ogino
- Program in Molecular Pathological Epidemiology (MPE), Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Institute of Science Tokyo, Tokyo, Japan
| | - Amanda I. Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Jonathan A. Nowak
- Program in Molecular Pathological Epidemiology (MPE), Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| |
Collapse
|
32
|
Wang S, Peng F, Dang M, Jiao H, Zhang H, Zhou K, Guo W, Gong Z, Guo L, Lu R, Li D, Liu B, Guo X, Xing J, Liu Y. Early detection of colorectal cancer using aberrant circulating cell-free mitochondrial DNA fragmentomics. Gut 2024:gutjnl-2024-333533. [PMID: 39694683 DOI: 10.1136/gutjnl-2024-333533] [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: 08/05/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Early detection of colorectal cancer (CRC) is crucial for improving the survival rates of patients. OBJECTIVE We aimed to develop a novel strategy for early CRC detection using the fragmentomic features of circulating cell-free mitochondrial DNA (ccf-mtDNA). DESIGN Here, a total of 1147 participants, including 478 healthy controls (HCs), 112 patients with advanced adenomas (AAs) and 557 patients with CRC, were enrolled from five hospitals and plasma samples were collected for capture-based ccf-mtDNA sequencing. RESULTS Our data analysis revealed significantly aberrant ccf-mtDNA fragmentomic features in patients with CRC and AA when compared with HCs. Then, a CRC detection (CD) model was constructed based on the fragmentomic features of ccf-mtDNA from 246 patients with CRC and 168 HC in the training cohort, showing area under the curve of 0.9863, sensitivity of 92.68% and specificity of 93.45%. Both internal and two external validation cohorts demonstrated the excellent capacity of CD model in distinguishing patients with early-stage CRC from HCs, greatly surpassing the performance of serum biomarkers. Furthermore, our CD model can also detect patients with AA with a sensitivity of 79.35% in AA cohort 1 and 85.00% in AA cohort 2. CONCLUSION In conclusion, based on aberrant ccf-mtDNA fragmentomic features, a novel and non-invasive approach was established for the detection of patients with early-stage CRC or AA, with high performance.
Collapse
Affiliation(s)
- Siyuan Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Fan Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Miao Dang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huanmin Jiao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huanqin Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Kaixiang Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenjie Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhiyun Gong
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Renquan Lu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Deliang Li
- Department of Gatroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bingrong Liu
- Department of Gatroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xu Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jinliang Xing
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yang Liu
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
33
|
Obeidat IM, Yahia Y, Chandra P, Altaiam A, Mohamed E, Saffo H, Abualsuod R, Mousa AAD, Shalatouni D, Alsa'ed K, Arabyat MY. Evaluating the necessity of colonoscopy in patients under 40 with rectal bleeding: insights from a large-scale retrospective analysis. Int J Colorectal Dis 2024; 39:202. [PMID: 39680187 PMCID: PMC11649774 DOI: 10.1007/s00384-024-04784-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/08/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE Bleeding per rectum (BPR) is a common clinical presentation, and colonoscopy is the gold standard for evaluating patients aged ≥ 45 years. However, its role in younger patients remains unclear. This study evaluated the appropriateness of colonoscopy in patients < 40 years of age who presented with BPR. METHODS This retrospective observational study was conducted over 10 years, including 3422 patients aged 18-40 years who underwent colonoscopy for BPR. The cohort was divided into two age groups: younger (aged 18-30 years) and older (31-40 years). The patients' baseline characteristics, colonoscopy findings, and histopathological results were analyzed. RESULTS Hemorrhoids were the most common finding (48%), with a higher prevalence in younger age groups (50.7%). Polyps were detected in 12.5% of patients, with 1.75% having advanced adenoma polyps (AAP) and 1.3% diagnosed with colorectal cancer (CRC). A family history of CRC/AAP was significantly associated with increased CRC risk (adjusted OR 6.35, 95% CI 2.24-18.02, p = 0.001) in explorative logistic regression analysis. CONCLUSION AAP and CRC were detected in a small but significant proportion of patients, particularly among those aged 18-30 years. The detection of significant lesions in this age group highlights the need for targeted colonoscopy based on specific risk factors such as family history and clinical presentation. Future research should prioritize the creation of targeted assessment models to improve clinical decision making in this context.
Collapse
Affiliation(s)
- Ibrahim M Obeidat
- Gastroenterology and Hepatology Department, Hamad Medical Corporation, Doha, Qatar.
| | - Yousef Yahia
- Gastroenterology and Hepatology Department, Hamad Medical Corporation, Doha, Qatar.
| | - Prem Chandra
- Medical Research Centre, Hamad Medical Corporation, Doha, Qatar
| | - Amani Altaiam
- Family Medicine Department, Primary Health Care Corporation, Doha, Qatar
| | - Ethar Mohamed
- Gastroenterology and Hepatology Department, Hamad Medical Corporation, Doha, Qatar
| | - Husam Saffo
- Gastroenterology and Hepatology Department, Hamad Medical Corporation, Doha, Qatar
| | - Raya Abualsuod
- Internal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | | | - Duha Shalatouni
- Internal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Khaled Alsa'ed
- Internal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Mahmoud Y Arabyat
- Internal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
34
|
Vasquez DL, Kreft C, Latka I, Popp J, Mantke R, Schie IW. Colon Tumor Discrimination Combining Independent Endoscopic Probe-Based Raman Spectroscopy and Optical Coherence Tomography Modalities with Bayes Rule. Int J Mol Sci 2024; 25:13306. [PMID: 39769072 PMCID: PMC11677020 DOI: 10.3390/ijms252413306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Colorectal cancer is one of the most prevalent forms of cancer globally. The most common routine diagnostic methods are the examination of the interior of the colon during colonoscopy or sigmoidoscopy, which frequently includes the removal of a biopsy sample. Optical methods, such as Raman spectroscopy (RS) and optical coherence tomography (OCT), can help to improve diagnostics and reduce the number of unnecessary biopsies. For in vivo use, we have developed fiber-optic probes, one for single-point Raman measurements and one for volumetric OCT. Here, we present the results of a clinical study using these fiber-optic probes in an ex vivo setting. The goal was to evaluate the beneficial effect of combining these two modalities on the AUC ROC score of the machine learning models for the discrimination of cancerous and healthy tissue. In the initial stage of the investigation, both modalities were validated separately using linear discriminant analysis. RS was subjected to spectral preprocessing, while OCT underwent texture feature extraction. Subsequently, both modalities were integrated using the Bayes rule, resulting in an enhanced area under the curve score of 0.93, representing an improvement over the 0.77 score for Raman spectroscopy and 0.86 for OCT.
Collapse
Affiliation(s)
- David L. Vasquez
- Leibniz-Institute of Photonic Technology (Leibniz-IPHT), Albert-Einstein-Str. 9, 07745 Jena, Germany; (D.L.V.); (I.L.); (J.P.)
| | - Calvin Kreft
- Department of Medical Engineering and Biotechnology, University of Applied Sciences—Jena, Carl-Zeiss-Promenade 2, 07745 Jena, Germany;
| | - Ines Latka
- Leibniz-Institute of Photonic Technology (Leibniz-IPHT), Albert-Einstein-Str. 9, 07745 Jena, Germany; (D.L.V.); (I.L.); (J.P.)
| | - Jürgen Popp
- Leibniz-Institute of Photonic Technology (Leibniz-IPHT), Albert-Einstein-Str. 9, 07745 Jena, Germany; (D.L.V.); (I.L.); (J.P.)
- Institute of Physical Chemistry (IPC), Abbe Center of Photonics (ACP), Friedrich-Schiller-University Jena, Helmholtzweg 4, 07743 Jena, Germany
| | - René Mantke
- Clinic for General and Visceral Surgery, University Hospital Brandenburg an der Havel, Brandenburg Medical School, 14770 Brandenburg an der Havel, Germany;
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School, 14770 Brandenburg an der Havel, Germany
| | - Iwan W. Schie
- Leibniz-Institute of Photonic Technology (Leibniz-IPHT), Albert-Einstein-Str. 9, 07745 Jena, Germany; (D.L.V.); (I.L.); (J.P.)
- Department of Medical Engineering and Biotechnology, University of Applied Sciences—Jena, Carl-Zeiss-Promenade 2, 07745 Jena, Germany;
| |
Collapse
|
35
|
Aspesi A, La Vecchia M, Sala G, Ghelardi E, Dianzani I. Study of Microbiota Associated to Early Tumors Can Shed Light on Colon Carcinogenesis. Int J Mol Sci 2024; 25:13308. [PMID: 39769073 PMCID: PMC11677268 DOI: 10.3390/ijms252413308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
An increasingly important role for gut microbiota in the initiation and progression of colorectal cancer (CRC) has been described. Even in the early stages of transformation, i.e., colorectal adenomas, changes in gut microbiota composition have been observed, and several bacterial species, such as pks+Escherichia coli and enterotoxigenic Bacteroides fragilis, have been proposed to drive colon tumorigenesis. In recent years, several strategies have been developed to study mucosa-associated microbiota (MAM), which is more closely associated with CRC development than lumen-associated microbiota (LAM) derived from fecal samples. This review summarizes the state of the art about the oncogenic actions of gut bacteria and compares the different sampling strategies to collect intestinal microbiota (feces, biopsies, swabs, brushes, and washing aspirates). In particular, this article recapitulates the current knowledge on MAM in colorectal adenomas and serrated polyps, since studying the intestinal microbiota associated with early-stage tumors can elucidate the molecular mechanisms underpinning CRC carcinogenesis.
Collapse
Affiliation(s)
- Anna Aspesi
- Department of Health Sciences, Università Del Piemonte Orientale, 28100 Novara, Italy; (A.A.); (M.L.V.); (G.S.)
| | - Marta La Vecchia
- Department of Health Sciences, Università Del Piemonte Orientale, 28100 Novara, Italy; (A.A.); (M.L.V.); (G.S.)
| | - Gloria Sala
- Department of Health Sciences, Università Del Piemonte Orientale, 28100 Novara, Italy; (A.A.); (M.L.V.); (G.S.)
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56123 Pisa, Italy;
| | - Irma Dianzani
- Department of Health Sciences, Università Del Piemonte Orientale, 28100 Novara, Italy; (A.A.); (M.L.V.); (G.S.)
| |
Collapse
|
36
|
Li Y, Wang S, Han C, Li XL, Min JZ. Unlocking the future of colorectal cancer detection: Advances in screening glycosylation-based biomarkers on biological mass spectrometry technology. J Chromatogr A 2024; 1738:465501. [PMID: 39504704 DOI: 10.1016/j.chroma.2024.465501] [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: 09/21/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
The incidence of colorectal cancer (CRC) is increasingly affecting younger populations, with its mortality rate rising annually. However, current clinical diagnostic techniques, such as colonoscopy and CEA antigen testing, remain invasive and prone to false-positive results, complicating early diagnosis and intervention. Glycosylation, a key post-translational modification, plays an essential role in cellular function, physiological regulation, and disease processes. In recent years, mass spectrometry technology has emerged as a powerful tool for screening glycan biomarkers, owing to its exceptional separation capabilities and sensitivity. This review encompasses the advancements in CRC glycan biomarkers from 2016 to 2024, with particular emphasis placed on N/O-glycan biomarkers identified through mass spectrometry. Nonetheless, the intrinsic low abundance and polyhydroxy nature of glycans hinder the specificity and sensitivity of current glycan biomarkers. To overcome these limitations, this article outlines pretreatment strategies for N/O-glycans, including glycan release, enrichment, purification, and derivatization, in conjunction with relative quantification techniques and high-throughput bioinformatics tools for biomarker screening. These strategies are anticipated to enhance the efficiency and precision of glycan biomarker identification through mass spectrometry. These advancements hold significant promise for enhancing CRC prevention, diagnosis, and treatment, thereby potentially improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Yuxuan Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Department of Pharmaceutical Analysis, College of Pharmacy Yanbian University, Yanji, 133002, Jilin Province, China
| | - Songze Wang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Department of Pharmaceutical Analysis, College of Pharmacy Yanbian University, Yanji, 133002, Jilin Province, China
| | - Chengqiang Han
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Department of Pharmaceutical Analysis, College of Pharmacy Yanbian University, Yanji, 133002, Jilin Province, China
| | - Xi-Ling Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Department of Pharmaceutical Analysis, College of Pharmacy Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jun Zhe Min
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Department of Pharmaceutical Analysis, College of Pharmacy Yanbian University, Yanji, 133002, Jilin Province, China.
| |
Collapse
|
37
|
Ezenkwa US, Omenai SA, Iyapo O, Ezekekwu CA, Adetona AE, Akunwata CU, Ale AO, Ebili HO. Low prostaglandin-endoperoxide synthase-2 gene expression in colorectal carcinomas may predict poorer survival. Ecancermedicalscience 2024; 18:1814. [PMID: 40171466 PMCID: PMC11959140 DOI: 10.3332/ecancer.2024.1814] [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: 03/13/2024] [Indexed: 04/03/2025] Open
Abstract
Introduction Prostaglandin-endoperoxide synthase-2 (ptgs2), otherwise called Cyclooxygenase 2, is overexpressed in colorectal carcinoma (CRC) compared to normal tissues. However, the impact of differential expression among ptgs2-positive tumours on CRC prognosis has not been well investigated. By sub-stratifying positive tumour expression, this study determined its potential influence on patients' outcomes. Methods The Cancer Genome Atlas database was explored to determine CRC cases with RNA-Sequence (RNA-Seq) transcript data and matched clinicopathological data alongside gene copy number variation and methylation status. Descriptive, chi-square, Fisher exact, Linear-by-Linear associations, logistic and Kaplan-Meier statistics were used to determine proportions, associations, predictors and survival between ptgs2 and tumour parameters using Statistical Package for Social Sciences version 20. Two-tailed p-value <0.05 was accepted as statistically significant. Results There were 534 CRC classified predominantly as adenocarcinoma not otherwise specified (86.3%) and mucinous carcinoma (12.4) histologically included in this study. Marker (ptgs2) expression ranged from 0.02 FPKM-131.89 FPKM, (Median 1.4 FPKM). The majority of the cases (53.4%) were diagnosed at an early stage and showed high ptgs2 RNA-Sequence (RNA-seq) expression in 51.5% (275/534). Significant associations were seen between ptgs2 expression and histological subtype (p < 0.001), lymphovascular invasion (p = 0.013), pN2 stage (> 6 positive lymph nodes) (p = 0.011) and American Joint Committee on Cancer Staging stage (p = 0.028), and these all had lower ptgs2 expression. On regression analysis, histological differentiation emerged as a predictor of ptgs2 expression (Odds ratio 2.749, 95% confidence interval 1.479-5.108, p < 0.001). Also, gene methylation was associated with reduced ptgs2 expression. Overall survival was significantly inferior among individuals with low ptgs2 tumours (p = 0.018) while that for disease-free survival was non-significant (p = 0.327). Conclusion CRCs with low ptgs2 transcripts are associated with poorer survival. This finding suggests a need for closer follow up and tailored adjuvant therapy for these patients.
Collapse
Affiliation(s)
- Uchenna Simon Ezenkwa
- Department of Histopathology, Federal University of Health Sciences Azare, Azare 751101, Bauchi, Nigeria
- Department of Pathology, Federal Medical Centre Azare, Azare 751101, Bauchi, Nigeria
| | | | - Oluwadamilare Iyapo
- Department of Pathology, Federal Medical Centre, Ebute Metta, Lagos 101212, Nigeria
| | - Chinedu Anthony Ezekekwu
- Bristol Haematology and Oncology Centre, University Hospital, Bristol and Weston NHS Trust, Bristol BS28ED, UK
| | - Adesoji E Adetona
- Department of Morbid Anatomy & Histopathology, Olabisi Onabanjo University, Ago-Iwoye 121101, Ogun State, Nigeria
| | - Chima Uzoma Akunwata
- Department of Haematology, University College Hospital Ibadan, Ibadan 200221, Oyo, Nigeria
| | - Ayotunde Oladunmi Ale
- Department of Medicine, Olabisi Onabanjo University/University Teaching Hospital, Sagamu 121101, Ogun, Nigeria
| | - Henry Okwuchukwu Ebili
- Department of Morbid Anatomy & Histopathology, Olabisi Onabanjo University, Ago-Iwoye 121101, Ogun State, Nigeria
| |
Collapse
|
38
|
Nguyen L, Shanmugan S. A Review Article: The Relationship Between Obesity and Colorectal Cancer. Curr Diab Rep 2024; 25:8. [PMID: 39621160 PMCID: PMC11611961 DOI: 10.1007/s11892-024-01556-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 12/06/2024]
Abstract
PURPOSE OF REVIEW This article aims to review the recent literature assessing the relationship between obesity and colorectal carcinogenesis, the effect of obesity on the treatment of colorectal cancer (CRC), tools available to help augment the increased risk, and outcomes for patients who are affected by both obesity and colorectal cancer. RECENT FINDINGS The biochemical mechanisms contributing to CRC carcinogenesis are not well understood but are suspected to be related to adipose tissue leading to a pro-inflammatory state and changes in the gut microbiome. Individuals with obesity are at higher risk for CRC development, worse oncologic outcomes, and increased rates of post-operative complications. Bariatric surgery decreases CRC risk but results with GLP-1 agonists are heterogeneous. Prehabilitation is the only weight loss method that has been demonstrated to decrease risks of post-operative morbidity in this population. Obesity augments CRC risk and outcomes. There are persistent knowledge gaps in etiology and epidemiology for the increased CRC risk in obese patients and more research is required to identify the therapeutic advantage of weight loss on CRC risk.
Collapse
Affiliation(s)
- Lily Nguyen
- Department of Surgery, Division of Colon and Rectal Surgery, University of California, 333 The City Blvd West, Suite 1600, Suite 1600, Irvine, CA, USA, 92868-3298
| | - Skandan Shanmugan
- Department of Surgery, Division of Colon and Rectal Surgery, University of California, 333 The City Blvd West, Suite 1600, Suite 1600, Irvine, CA, USA, 92868-3298.
| |
Collapse
|
39
|
Hemade A, Hallit S. The risk and distribution of second primary cancers according to subsite of primary stomach cancer: a retrospective cohort population-based study. Ann Med Surg (Lond) 2024; 86:6944-6950. [PMID: 39649899 PMCID: PMC11623845 DOI: 10.1097/ms9.0000000000002695] [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: 08/22/2024] [Accepted: 10/17/2024] [Indexed: 12/11/2024] Open
Abstract
Background The development of second primary cancers (SPCs) following a diagnosis of stomach cancer presents a significant clinical challenge, with varying risks depending on the anatomic subsite of the primary tumor, patient demographics, and treatment modalities. This study aims to assess the risk of SPCs in stomach cancer survivors, focusing on differences across anatomic subsites, sex, age, and treatment periods. Methods The authors conducted a retrospective cohort study using data from stomach cancer patients, analyzing the incidence of SPCs based on the anatomic location of the primary tumor, with stratifications by sex, age, latency period, and year of diagnosis. Standardized incidence ratios (SIRs) were calculated to compare the observed SPC rates with those expected in the general population. Results Elevated stomach SPC risk was observed across most anatomic subsites, particularly in the body (SIR 8.84) and fundus (SIR 7.34). Females exhibited higher SIRs compared to males, especially in the fundus (SIR 13.33 for females vs. 4.55 for males). Younger patients (<50 years) had significantly higher SPC risks, particularly for cancers originating in the fundus (SIR 49.56). Notably, patients diagnosed after 2010 showed the highest SIRs, indicating a potential impact of advances in diagnostic and therapeutic modalities. Nonstomach SPCs, including colorectal, lung, and thyroid cancers, were significantly elevated, with distinct patterns based on the primary tumor site. Conclusions The study highlights the critical role of primary tumor location, sex, age, and treatment era in determining SPC risk in stomach cancer survivors. These findings underscore the need for tailored surveillance strategies to manage long-term cancer risks in this population.
Collapse
Affiliation(s)
- Ali Hemade
- Faculty of Medicine, Lebanese University, Hadat, Lebanon
| | - Souheil Hallit
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| |
Collapse
|
40
|
Koh GE, Ng B, Lagström RM, Foo FJ, Chin SE, Wan FT, Kam JH, Yeung B, Kwan C, Hassan C, Gögenur I, Koh FH. Real-World Assessment of the Efficacy of Computer-Assisted Diagnosis in Colonoscopy: A Single Institution Cohort Study in Singapore. MAYO CLINIC PROCEEDINGS. DIGITAL HEALTH 2024; 2:647-655. [PMID: 40206538 PMCID: PMC11976013 DOI: 10.1016/j.mcpdig.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Objective To review the efficacy and accuracy of the GI Genius Intelligent Endoscopy Module Computer-Assisted Diagnosis (CADx) program in colonic adenoma detection and real-time polyp characterization. Patients and Methods Colonoscopy remains the gold standard in colonic screening and evaluation. The incorporation of artificial intelligence (AI) technology therefore allows for optimized endoscopic performance. However, validation of most CADx programs with real-world data remains scarce. This prospective cohort study was conducted within a single Singaporean institution between April 1, 2023 and December 31, 2023. Videos of all AI-enabled colonoscopies were reviewed with polyp-by-polyp analysis performed. Real-time polyp characterization predictions after sustained polyp detection were compared against final histology results to assess the accuracy of the CADx system at colonic adenoma identification. Results A total of 808 videos of CADx colonoscopies were reviewed. Out of the 781 polypectomies performed, 543 (69.5%) and 222 (28.4%) were adenomas and non-adenomas on final histology, respectively. Overall, GI Genius correctly characterized adenomas with 89.4% sensitivity, 61.7% specificity, a positive predictive value of 85.4%, a negative predictive value of 69.8%, and 81.5% accuracy. The negative predictive value for rectosigmoid lesions (80.3%) was notably higher than for colonic lesions (54.2%), attributed to the increased prevalence of hyperplastic rectosigmoid polyps (11.4%) vs other colonic regions (5.4%). Conclusion Computer-Assisted Diagnosis is therefore a promising adjunct in colonoscopy with substantial clinical implications. Accurate identification of low-risk non-adenomatous polyps encourages the adoption of "resect-and-discard" strategies. However, further calibration of AI systems is needed before the acceptance of such strategies as the new standard of care.
Collapse
Affiliation(s)
- Gabrielle E. Koh
- Department of Surgery, Sengkang General Hospital, Singapore, Singapore
| | - Brittany Ng
- Department of Surgery, Sengkang General Hospital, Singapore, Singapore
| | - Ronja M.B. Lagström
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Koege, Denmark
| | - Fung-Joon Foo
- Colorectal Service, Department of Surgery, Sengkang General Hospital, Singapore, Singapore
| | - Shuen-Ern Chin
- Department of Surgery, Sengkang General Hospital, Singapore, Singapore
| | - Fang-Ting Wan
- Department of Surgery, Sengkang General Hospital, Singapore, Singapore
| | - Juinn Huar Kam
- Department of Surgery, Sengkang General Hospital, Singapore, Singapore
| | - Baldwin Yeung
- Department of Surgery, Sengkang General Hospital, Singapore, Singapore
| | - Clarence Kwan
- Department of Gastroenterology, Sengkang General Hospital, Singapore, Singapore
| | - Cesare Hassan
- Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
- Endoscopy Unit, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Koege, Denmark
| | - Frederick H. Koh
- Colorectal Service, Department of Surgery, Sengkang General Hospital, Singapore, Singapore
- Department of Surgery, Duke-National University of Singapore (NUS) Medical School, National University of Singapore, Singapore, Singapore
- Department of Surgery, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
41
|
Xu R, Balmer L, Chen G, Song M. Role of N-Glycosylation in Gastrointestinal Cancers. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:596-607. [PMID: 39514331 DOI: 10.1089/omi.2024.0174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Gastrointestinal cancers pose a significant global health challenge. N-glycosylation modulates various cellular processes, including key cancer-related mechanisms. Elucidating its involvement in the onset and advancement of these cancers can offer critical insights for enhancing diagnostic and therapeutic approaches. This review outlines the core process of protein N-glycosylation and highlights its contribution to the progression of gastrointestinal cancers, encompassing cell proliferation, survival, invasion, metastasis, and immune evasion, mainly through its impact on critical signaling pathways. Notably, aberrant N-glycosylation patterns have emerged as crucial biomarkers for the diagnosis and prognosis of various gastrointestinal cancers, providing the foundation for more personalized therapeutic approaches. Therapeutic strategies targeting N-glycosylation, such as glycosyltransferase inhibitors and glycoengineering, show significant promise in mitigating tumor aggressiveness and enhancing immune recognition. However, the clinical implementation of N-glycosylation biomarkers requires the standardization of glycosylation analysis techniques and solutions to challenges in sample processing and data interpretation. Future research efforts should concentrate on overcoming these obstacles to unlock the full potential of N-glycosylation in enhancing cancer management and advancing patient outcomes.
Collapse
Affiliation(s)
- Ruirui Xu
- Center for Precision Health, Edith Cowan University, Western Australia, Australia
- School of Medical and Health Science, Edith Cowan University, Western Australia, Australia
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Shantou University Medical College, Guangdong, China
| | - Lois Balmer
- Center for Precision Health, Edith Cowan University, Western Australia, Australia
- School of Medical and Health Science, Edith Cowan University, Western Australia, Australia
| | - Gengzhen Chen
- Digestive Disease Prevention and Treatment Center, Chenghai District People's Hospital, Guangdong, China
| | - Manshu Song
- School of Medical and Health Science, Edith Cowan University, Western Australia, Australia
| |
Collapse
|
42
|
Permain J, Hock B, Eglinton T, Purcell R. Functional links between the microbiome and the molecular pathways of colorectal carcinogenesis. Cancer Metastasis Rev 2024; 43:1463-1474. [PMID: 39340753 PMCID: PMC11554747 DOI: 10.1007/s10555-024-10215-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
Colorectal cancer (CRC) is a common cancer, with a concerning rise in early-onset CRC cases, signalling a shift in disease epidemiology. Whilst our understanding of the molecular underpinnings of CRC has expanded, the complexities underlying its initiation remain elusive, with emerging evidence implicating the microbiome in CRC pathogenesis. This review synthesizes current knowledge on the intricate interplay between the microbiome, tumour microenvironment (TME), and molecular pathways driving CRC carcinogenesis. Recent studies have reported how the microbiome may modulate the TME and tumour immune responses, consequently influencing cancer progression, and whilst specific bacteria have been linked with CRC, the underlying mechanisms remains poorly understood. By elucidating the functional links between microbial landscapes and carcinogenesis pathways, this review offers insights into how bacteria orchestrate diverse pathways of CRC development, shedding light on potential therapeutic targets and personalized intervention strategies.
Collapse
Affiliation(s)
- Jessica Permain
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand
| | - Barry Hock
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Timothy Eglinton
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand
| | - Rachel Purcell
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
43
|
Duta-Ion SG, Juganaru IR, Hotinceanu IA, Dan A, Burtavel LM, Coman MC, Focsa IO, Zaruha AG, Codreanu PC, Bohiltea LC, Radoi VE. Redefining Therapeutic Approaches in Colorectal Cancer: Targeting Molecular Pathways and Overcoming Resistance. Int J Mol Sci 2024; 25:12507. [PMID: 39684219 DOI: 10.3390/ijms252312507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Colorectal cancer (CRC) arises through a combination of genetic and epigenetic alterations that affect key pathways involved in tumor growth and progression. This review examines the major molecular pathways driving CRC, including Chromosomal Instability (CIN), Microsatellite Instability (MSI), and the CpG Island Methylator Phenotype (CIMP). Key mutations in genes such as APC, KRAS, NRAS, BRAF, and TP53 activate signaling pathways like Wnt, EGFR, and PI3K/AKT, contributing to tumorigenesis and influencing responses to targeted therapies. Resistance mechanisms, including mutations that bypass drug action, remain challenging in CRC treatment. This review highlights the role of molecular profiling in guiding the use of targeted therapies such as tyrosine kinase inhibitors and immune checkpoint inhibitors. Novel combination treatments are also discussed as strategies to improve outcomes and overcome resistance. Understanding these molecular mechanisms is critical to advancing personalized treatment approaches in CRC and improving patient prognosis.
Collapse
Affiliation(s)
- Simona Gabriela Duta-Ion
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ioana Ruxandra Juganaru
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Iulian Andrei Hotinceanu
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Andra Dan
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Livia Malina Burtavel
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Madalin Codrut Coman
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ina Ofelia Focsa
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Andra Giorgiana Zaruha
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Patricia Christina Codreanu
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Laurentiu Camil Bohiltea
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- "Alessandrescu-Rusescu" National Institute for Maternal and Child Health, 20382 Bucharest, Romania
| | - Viorica Elena Radoi
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- "Alessandrescu-Rusescu" National Institute for Maternal and Child Health, 20382 Bucharest, Romania
| |
Collapse
|
44
|
Zajkowska M, Orywal K, Gryko M. Potential Utility of A Proliferation-Inducing Ligand (APRIL) in Colorectal Cancer. Int J Mol Sci 2024; 25:12496. [PMID: 39684206 DOI: 10.3390/ijms252312496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
APRIL (A proliferation-inducing ligand) is a member of the tumor necrosis factor superfamily that is overexpressed in a variety of malignant tumors, including colorectal cancer (CRC). Its key physiological roles include inducing the immunoglobulin switch and ensuring plasmocyte survival. In terms of pathological roles, APRIL antagonism has been identified as a key target in autoimmune diseases and immunoglobulin disorders. As previously demonstrated, several inflammatory processes occur at the site of neoplastic initial stages, and their local symptoms are difficult to detect, particularly in the early stages. That is why we chose to study the current literature on APRIL's role in the development of colorectal cancer. The main objective of our research was to investigate the role of APRIL in cancer initiation and its usefulness in the detection and therapy of CRC. Interestingly, the findings conducted so far show that the selected protein has a significant potential as a CRC biomarker and treatment target. Importantly, based on its concentration, it is possible to identify CRC patients, but whether the lesion has a benign or malignant nature, indicating the possibility of rapid detection of an ongoing disease process.
Collapse
Affiliation(s)
- Monika Zajkowska
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok Clinical Hospital, 15-269 Białystok, Poland
| | - Karolina Orywal
- Department of Biochemical Diagnostics, Medical University of Bialystok Clinical Hospital, 15-269 Białystok, Poland
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland
| | - Mariusz Gryko
- Department of Surgical Nursing, Medical University of Białystok, 15-274 Białystok, Poland
- 1st Clinical Department of General and Endocrine Surgery, Medical University of Bialystok Clinical Hospital, 15-276 Białystok, Poland
| |
Collapse
|
45
|
Zinkeng A, Taylor FL, Cheong SH, Song H, Merchant JL. Early Onset Colorectal Cancer: Molecular Underpinnings Accelerating Occurrence. Cell Mol Gastroenterol Hepatol 2024; 19:101425. [PMID: 39510499 PMCID: PMC11731505 DOI: 10.1016/j.jcmgh.2024.101425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
The onset of colorectal cancer (CRC) in patients younger than 50 continues to rapidly increase. This study highlights the epidemiologic changes, risk factors, clinical characteristics, and molecular profiles prevalent in early onset CRC patients, and identifies key areas for future research. It has been noted that only a small fraction of early onset CRC cases is attributed to known hereditary mutations and fit the canonical pathway of late-onset colorectal cancer development. To highlight this, we review the genetic and epigenetic modifications specific to early onset CRC. We also discuss the synergetic effect of single-nucleotide polymorphisms and environmental factors on the early onset of CRC. Additionally, we discuss the potential of noninvasive biomarker assays to enhance early detection, screening, diagnosis, and prognostic outcome predictions.
Collapse
Affiliation(s)
- Atehkeng Zinkeng
- Medical Scientist Training Program, University of Arizona College of Medicine, Tucson, Arizona
| | | | | | | | - Juanita L Merchant
- Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona.
| |
Collapse
|
46
|
Li D, Peng M, Zhou J. Noncoding RNA Linc00475 promotes the proliferation of colorectal cancer cells by targeting miR-107/CDK6 axis. J Biochem Mol Toxicol 2024; 38:e70012. [PMID: 39434447 DOI: 10.1002/jbt.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/11/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024]
Abstract
Colorectal cancer (CRC) represents a substantial challenge to public health. Despite extensive research, the pathogenesis of CRC is not yet fully elucidated, hindering the development of effective therapeutic strategies. Recent advancements have underscored the importance of Non-coding RNAs in tumor biology. Our research identified a significant upregulation of Linc00475 in CRC, which correlated with reduced survival rates among CRC patients. Consequently, this study aimed to elucidate the mechanisms by which Linc00475 contributed to CRC progression. Employing a comprehensive array of experimental techniques-including CCK-8 assays, colony formation assays, flow cytometry, quantitative PCR (qPCR), western blot analysis, and in vivo tumorigenesis assays-we have demonstrated that Linc00475 enhances CRC cell proliferation. Further analysis revealed that Linc00475 directly interacted with miR-107, leading to its downregulation. Moreover, our findings confirmed that miR-107 directly targeted CDK6, which was markedly downregulated following Linc00475 silencing. In vivo experiments further indicated that the silencing of Linc00475 markedly inhibited the proliferation of CRC cells. Collectively, our findings suggested that Linc00475 facilitated CRC cell proliferation through the regulation of the miR-107/CDK6 axis, thereby providing a novel perspective for understanding the molecular mechanisms underlying CRC development.
Collapse
Affiliation(s)
- Dongqing Li
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
- The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Mingya Peng
- Nuclear Medicine Department, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Juying Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
47
|
Gallardo‐Gómez M, Costas‐Ríos L, Garcia‐Prieto CA, Álvarez‐Rodríguez L, Bujanda L, Barrero M, Castells A, Balaguer F, Jover R, Esteller M, Tardío Baiges A, González‐Carreró Fojón J, Cubiella J, De Chiara L. Serum DNA methylome of the colorectal cancer serrated pathway enables non-invasive detection. Mol Oncol 2024; 18:2696-2713. [PMID: 38129291 PMCID: PMC11547225 DOI: 10.1002/1878-0261.13573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 12/23/2023] Open
Abstract
The clinical relevance of the colorectal cancer serrated pathway is evident, but the screening of serrated lesions remains challenging. We aimed to characterize the serum methylome of the serrated pathway and to evaluate circulating cell-free DNA (cfDNA) methylomes as a potential source of biomarkers for the non-invasive detection of serrated lesions. We collected serum samples from individuals with serrated adenocarcinoma (SAC), traditional serrated adenomas, sessile serrated lesions, hyperplastic polyps and individuals with no colorectal findings. First, we quantified cfDNA methylation with the MethylationEPIC array. Then, we compared the methylation profiles with tissue and serum datasets. Finally, we evaluated the utility of serum cfDNA methylation biomarkers. We identified a differential methylation profile able to distinguish high-risk serrated lesions from no serrated neoplasia, showing concordance with tissue methylation from SAC and sessile serrated lesions. Serum methylation profiles are pathway-specific, clearly separating serrated lesions from conventional adenomas. The combination of ninjurin 2 (NINJ2) and glutamate-rich 1 (ERICH1) methylation discriminated high-risk serrated lesions and SAC with 91.4% sensitivity (64.4% specificity), while zinc finger protein 718 (ZNF718) methylation reported 100% sensitivity for the detection of SAC (96% specificity). This is the first study exploring the serum methylome of serrated lesions. Differential methylation of cfDNA can be used for the non-invasive detection of colorectal serrated lesions.
Collapse
Affiliation(s)
- María Gallardo‐Gómez
- CINBIO, Universidade de VigoSpain
- Department of Biochemistry, Genetics and ImmunologyUniversidade de VigoSpain
- Galicia Sur Health Research Institute (IIS Galicia Sur)SERGAS‐UVIGOSpain
| | - Lara Costas‐Ríos
- CINBIO, Universidade de VigoSpain
- Department of Biochemistry, Genetics and ImmunologyUniversidade de VigoSpain
| | - Carlos A. Garcia‐Prieto
- Josep Carreras Leukaemia Research Institute (IJC)BadalonaSpain
- Life Sciences DepartmentBarcelona Supercomputing Center (BSC)Spain
| | - Lara Álvarez‐Rodríguez
- CINBIO, Universidade de VigoSpain
- Department of Biochemistry, Genetics and ImmunologyUniversidade de VigoSpain
| | - Luis Bujanda
- Department of Gastroenterology, Biodonostia Health Research Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)Universidad del País Vasco (UPV/EHU)San SebastiánSpain
| | - Maialen Barrero
- Department of OncologyHospital Universitario DonostiaSan SebastiánSpain
| | - Antoni Castells
- Gastroenterology Department, Hospital Clínic, IDIBAPS, CIBERehdUniversity of BarcelonaSpain
| | - Francesc Balaguer
- Gastroenterology Department, Hospital Clínic, IDIBAPS, CIBERehdUniversity of BarcelonaSpain
| | - Rodrigo Jover
- Servicio de Medicina Digestiva, Hospital General Universitario Dr. Balmis ISABIALUniversidad Miguel HernándezAlicanteSpain
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC)BadalonaSpain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC)MadridSpain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
- Physiological Sciences Department, School of Medicine and Health SciencesUniversity of Barcelona (UB)Spain
| | - Antoni Tardío Baiges
- Department of PathologyHospital Álvaro Cunqueiro, Instituto de Investigación Biomédica Galicia SurVigoSpain
| | | | - Joaquín Cubiella
- Department of GastroenterologyComplexo Hospitalario Universitario de Ourense, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)OurenseSpain
| | - Loretta De Chiara
- CINBIO, Universidade de VigoSpain
- Department of Biochemistry, Genetics and ImmunologyUniversidade de VigoSpain
- Galicia Sur Health Research Institute (IIS Galicia Sur)SERGAS‐UVIGOSpain
| |
Collapse
|
48
|
Huang T, Zhang Y, Wu Y, Han X, Li L, Guo Z, Li K, Xin Y, Wang W. CEBPB dampens the cuproptosis sensitivity of colorectal cancer cells by facilitating the PI3K/AKT/mTOR signaling pathway. Saudi J Gastroenterol 2024; 30:381-388. [PMID: 39246119 PMCID: PMC11630481 DOI: 10.4103/sjg.sjg_169_24] [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: 05/13/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND Cuproptosis is a novel pathway that differs from other forms of cell death and has been confirmed to be applicable for predicting tumor prognosis and clinical treatment response. However, the mechanism underlying the resistance of colorectal cancer (CRC) to cuproptosis at the molecular level has not been elucidated. METHODS Using bioinformatics analysis, the expression of CCAAT/enhancer-binding protein beta (CEBPB) in CRC tissues and its enrichment in biological processes were detected. Quantitative reverse transcription polymerase chain reaction and western blotting (WB) were employed to test the expression of CEBPB in CRC cells. WB was utilized to assess the levels of proteins related to cuproptosis and the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway. The MTT assay was used to test cell viability. Cell proliferation was assessed by a colony formation assay. Transwell assays were used to measure cell migration and invasion ability. DLAT-aggregate formation was determined by immunofluorescence. RESULTS CEBPB was highly upregulated in CRC cells to enhance cell viability, proliferation, migration, and invasion. CEBPB was strongly implicated in copper ion homeostasis and the mTOR signaling pathway in CRC. In a CRC cuproptosis cell model, rescue experiments revealed that a PI3K/AKT/mTOR pathway inhibitor attenuated the promoting effect of CEBPB overexpression on the PI3K/AKT/mTOR pathway and rescued the sensitivity of CRC to cuproptosis. CONCLUSION This work demonstrated that CEBPB can activate the PI3K/AKT/mTOR signaling pathway, thereby decreasing the sensitivity of CRC to cuproptosis. These data suggested that targeting CEBPB or the PI3K/AKT/mTOR pathway may enhance the sensitivity of CRC patients to cuproptosis, providing a combined therapeutic strategy for cuproptosis-induced therapy.
Collapse
Affiliation(s)
- Tianchen Huang
- The Fourth Department of General Surgery, Anyang Tumor Hospital, Anyang, China
| | - Yong Zhang
- The Fourth Department of General Surgery, Anyang Tumor Hospital, Anyang, China
| | - Yachao Wu
- The Fourth Department of General Surgery, Anyang Tumor Hospital, Anyang, China
| | - Xiaodong Han
- The Fourth Department of General Surgery, Anyang Tumor Hospital, Anyang, China
| | - Lei Li
- The Fourth Department of General Surgery, Anyang Tumor Hospital, Anyang, China
| | - Zhipeng Guo
- The Fourth Department of General Surgery, Anyang Tumor Hospital, Anyang, China
| | - Kan Li
- The Fourth Department of General Surgery, Anyang Tumor Hospital, Anyang, China
| | - Yanshan Xin
- The Fourth Department of General Surgery, Anyang Tumor Hospital, Anyang, China
| | - Weijie Wang
- Thoracic Surgery Department, Anyang Tumor Hospital, Anyang, China
| |
Collapse
|
49
|
Collatuzzo G, Seyyedsalehi MS, Rashidian H, Hadji M, Safari-Faramani R, Rezaianzadeh A, Malekzadeh R, Zendehdel K, Boffetta P. Determinants of early-onset colorectal cancer: a multicenter case-control study in Iran. Eur J Cancer Prev 2024; 33:533-540. [PMID: 38687254 DOI: 10.1097/cej.0000000000000888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
BACKGROUND We aimed to study the risk factors of early-onset colorectal cancer (CRC) incidence in the Iranian population. Early onset CRC in Iran is a relevant health issue that deserves further epidemiological efforts to be defined and controlled as far as possible. Early age screening of low-tract of the intestine would be particularly useful in families of colorectal cancer patients. METHODS We analyzed data from a multicenter hospital-based case-control study in Iran (The Iranian Study of Opium and Cancer). Sociodemographic and lifestyle information was collected using validated questionnaires. Multivariate logistic regressions estimated the odds ratios (OR) and 95% confidence intervals (CIs) for the association of early-onset CRC in individuals under the age of 50 and potential risk factors, including physical activity, socioeconomic status, body shape at age 15, dietary factors, vitamin D, cigarettes and waterpipe smoking, opium use and family history of CRC. Additionally, a subgroup analysis was conducted for individuals with a very young age of CRC onset (i.e. <35 years). RESULTS We analyzed data of 189 developed CRC below age 50 (99 colon and 90 rectum), and 66 patients under the age 35 (13 colon and 21 rectum). Early CRC was inversely associated with vegetables (OR, 0.59; 95% CI, 0.38-0.92 for 422-576 g/day) and vitamin D (OR, 0.49; 95% CI, 0.26-0.94), and positively associated with red meat intake (OR, 1.80; 1.15-2.83 per 25.65 g/day). Vegetables (OR, 0.51; 95% CI, 0.27-0.98 for 576 g/day), red meat (OR, 2.05; 95% CI, 1.11-3.79 for 25.65 g/day), vitamin D (OR, 0.29; 95% CI, 0.10-0.86) and opium use (OR, 2.61; 95% CI, 1.01-6.74) were associated with early rectum cancer. Results were heterogeneous by cancer site for high fruit and vegetables intakes and cigarette smoking. Family history was associated with CRC (OR, 3.16; 95% CI, 1.29-10.9) and rectum cancer (OR, 3.22; 95% CI, 1.24-14.4) in subjects younger than 35, and, to a lesser extent, with CRC and rectum cancer before age 50. CONCLUSION Early-onset CRC was related to the intake of vegetables, vitamin D and red meat in Iran. Early-onset rectum cancer was associated with regular opium use. Family history was associated with early CRC and early rectum cancer, particularly below the age of 35.
Collapse
Affiliation(s)
- Giulia Collatuzzo
- Department of Medical and Surgical Sciences, University of Bologna, Italy
| | - Monireh Sadat Seyyedsalehi
- Department of Medical and Surgical Sciences, University of Bologna, Italy
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamideh Rashidian
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Hadji
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
- Health Sciences Unit, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Roya Safari-Faramani
- Research Center for Environmental Determinants of Health, School of Public Health, Kermanshah Medical Sciences University, Kermanshah, Iran
| | - Abbas Rezaianzadeh
- Colorectal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Malekzadeh
- Digestive Diseases Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Kazem Zendehdel
- Department of Medical and Surgical Sciences, University of Bologna, Italy
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Biology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Italy
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, USA
- Department of Family, Population and Preventive Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
50
|
Li Q, Geng S, Luo H, Wang W, Mo YQ, Luo Q, Wang L, Song GB, Sheng JP, Xu B. Signaling pathways involved in colorectal cancer: pathogenesis and targeted therapy. Signal Transduct Target Ther 2024; 9:266. [PMID: 39370455 PMCID: PMC11456611 DOI: 10.1038/s41392-024-01953-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/25/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Its complexity is influenced by various signal transduction networks that govern cellular proliferation, survival, differentiation, and apoptosis. The pathogenesis of CRC is a testament to the dysregulation of these signaling cascades, which culminates in the malignant transformation of colonic epithelium. This review aims to dissect the foundational signaling mechanisms implicated in CRC, to elucidate the generalized principles underpinning neoplastic evolution and progression. We discuss the molecular hallmarks of CRC, including the genomic, epigenomic and microbial features of CRC to highlight the role of signal transduction in the orchestration of the tumorigenic process. Concurrently, we review the advent of targeted and immune therapies in CRC, assessing their impact on the current clinical landscape. The development of these therapies has been informed by a deepening understanding of oncogenic signaling, leading to the identification of key nodes within these networks that can be exploited pharmacologically. Furthermore, we explore the potential of integrating AI to enhance the precision of therapeutic targeting and patient stratification, emphasizing their role in personalized medicine. In summary, our review captures the dynamic interplay between aberrant signaling in CRC pathogenesis and the concerted efforts to counteract these changes through targeted therapeutic strategies, ultimately aiming to pave the way for improved prognosis and personalized treatment modalities in colorectal cancer.
Collapse
Affiliation(s)
- Qing Li
- The Shapingba Hospital, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Shan Geng
- Central Laboratory, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Wang
- Chongqing Municipal Health and Health Committee, Chongqing, China
| | - Ya-Qi Mo
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Lu Wang
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Guan-Bin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Jian-Peng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|